51
|
Alquadan KF, Santos AH, Leghrouz M, Ozrazgat-Baslanti T, Bozorgmehri S, Gupta G, Womer KL. A pilot study of immunosuppression resumption following BK viremia resolution. Transpl Infect Dis 2020; 23:e13508. [PMID: 33176016 DOI: 10.1111/tid.13508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/23/2020] [Accepted: 10/25/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Immunosuppression reduction for BK viremia is associated with de novo humoral responses, which are a risk factor for rejection and graft loss. In this pilot project, we tested a protocol of immunosuppression resumption to standard dose after viral clearance for optimal protection against humoral immunity in patients undergoing treatment for BK viremia. METHODS Thirty-six consecutive kidney transplant recipients who developed BK viremia from 7/1/2014 to 11/18/2016 underwent immunosuppression reduction. After 4 weeks of absent viremia, mycophenolate mofetil (MMF) was increased by 500mg/day every 2 weeks up to standard dosage, followed by increase of tacrolimus trough levels to 5-7 ng/mL. If viremia recurred during the increase, immunosuppression was reduced in this same stepwise fashion, with stepwise increase again after 2 months of negative viremia. RESULTS Mean tacrolimus trough level (ng/mL) was 8.3 ± 2.7 at viremia onset, 5.3 ± 3.6 at resolution, and 5.6 ± 2.0 at study end date. Mean daily dose (mg) of MMF was 1574 ± 355 at onset, 910 ± 230 at resolution, and 1377 ± 451 at study end date. Only one patient developed low level viremia recurrence (peak 2875 copies/mL) during the period of immunosuppression resumption that ultimately resolved. CONCLUSIONS The results of our pilot project indicate that following BK viremia resolution, resumption of standard immunosuppression can be achieved safely without BK viremia recurrence. Larger trials with long-term follow up are required to determine whether such an approach improves long-term graft survival.
Collapse
Affiliation(s)
| | - Alfonso H Santos
- Medicine- Nephrology, University of Florida, Gainesville, FL, USA
| | | | | | | | - Gaurav Gupta
- Medicine-Nephrology, Virginia Commonwealth University, Richmond, VA, USA
| | - Karl L Womer
- Medicine- Nephrology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
52
|
Steines L, Poth H, Herrmann M, Schuster A, Banas B, Bergler T. B Cell Activating Factor (BAFF) Is Required for the Development of Intra-Renal Tertiary Lymphoid Organs in Experimental Kidney Transplantation in Rats. Int J Mol Sci 2020; 21:ijms21218045. [PMID: 33126753 PMCID: PMC7662293 DOI: 10.3390/ijms21218045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/22/2022] Open
Abstract
Intra-renal tertiary lymphoid organs (TLOs) are associated with worsened outcome in kidney transplantation (Ktx). We used an anti-BAFF (B cell activating factor) intervention to investigate whether BAFF is required for TLO formation in a full MHC-mismatch Ktx model in rats. Rats received either therapeutic immunosuppression (no rejection, NR) or subtherapeutic immunosuppression (chronic rejection, CR) and were sacrificed on d56. One group additionally received an anti-BAFF antibody (CR + AB). Intra-renal T (CD3+) and B (CD20+) cells, their proliferation (Ki67+), and IgG+ plasma cells were analyzed by immunofluorescence microscopy. Formation of T and B cell zones and TLOs was assessed. Intra-renal expression of TLO-promoting factors, molecules of T:B crosstalk, and B cell differentiation was analyzed by qPCR. Intra-renal B and T cell zones and TLOs were detected in CR and were associated with elevated intra-renal mRNA expression of TLO-promoting factors, including CXCL13, CCL19, lymphotoxin-β, and BAFF. Intra-renal plasma cells were also elevated in CR. Anti-BAFF treatment significantly decreased intra-renal B cell zones and TLO, as well as intra-renal B cell-derived TLO-promoting factors and B cell differentiation markers. We conclude that BAFF-dependent intra-renal B cells promote TLO formation and advance local adaptive alloimmune responses in chronic rejection.
Collapse
Affiliation(s)
- Louisa Steines
- Correspondence: ; Tel.: +49-941-9447301; Fax: +49-941-9447302
| | | | | | | | | | | |
Collapse
|
53
|
Stites E, Kumar D, Olaitan O, John Swanson S, Leca N, Weir M, Bromberg J, Melancon J, Agha I, Fattah H, Alhamad T, Qazi Y, Wiseman A, Gupta G. High levels of dd-cfDNA identify patients with TCMR 1A and borderline allograft rejection at elevated risk of graft injury. Am J Transplant 2020; 20:2491-2498. [PMID: 32056331 PMCID: PMC7496411 DOI: 10.1111/ajt.15822] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 01/30/2020] [Accepted: 02/02/2020] [Indexed: 01/25/2023]
Abstract
The clinical importance of subclinical, early T cell-mediated rejection (Banff TCMR 1A and borderline lesions) remains unclear, due, in part to the fact that histologic lesions used to characterize early TCMR can be nonspecific. Donor-derived cell-free DNA (dd-cfDNA) is an important molecular marker of active graft injury. Over a study period from June 2017 to May 2019, we assessed clinical outcomes in 79 patients diagnosed with TCMR 1A/borderline rejection across 11 US centers with a simultaneous measurement of dd-cfDNA. Forty-two patients had elevated dd-cfDNA (≥0.5%) and 37 patients had low levels (<0.5%). Elevated levels of dd-cfDNA predicted adverse clinical outcomes: among patients with elevated cfDNA, estimated glomerular filtration rate declined by 8.5% (interquartile rate [IQR] -16.22% to -1.39%) (-3.50 mL/min/1.73 m2 IQR -8.00 to -1.00) vs 0% (-4.92%, 4.76%) in low dd-cfDNA patients (P = .004), de novo donor-specific antibody formation was seen in 40% (17/42) vs 2.7% (P < .0001), and future or persistent rejection occurred in 9 of 42 patients (21.4%) vs 0% (P = .003). The use of dd-cfDNA may complement the Banff classification and to risk stratify patients with borderline/TCMR 1A identified on biopsy.
Collapse
Affiliation(s)
| | - Dhiren Kumar
- Medicine/NephrologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | | | | | - Nicolae Leca
- Division of NephrologyDepartment of MedicineUniversity of Washington Medical CenterSeattleWashingtonUSA
| | - Matthew Weir
- Division of NephrologyUniversity of MarylandBaltimoreMarylandUSA
| | | | - Joseph Melancon
- SurgeryGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Irfan Agha
- Medical City Dallas HospitalDallasTexasUSA
| | - Hasan Fattah
- University of Kentucky Medical CenterLexingtonKentuckyUSA
| | - Tarek Alhamad
- Washington University in Saint LouisSaint LouisMissouriUSA
| | - Yasir Qazi
- Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Internal MedicineDivision of NephrologyLos AngelesCaliforniaUSA
| | | | - Gaurav Gupta
- Medicine/NephrologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| |
Collapse
|
54
|
Matas AJ, Helgeson ES, Gaston R, Cosio F, Mannon R, Kasiske BL, Hunsicker L, Gourishankar S, Rush D, Michael Cecka J, Connett J, Grande JP. Inflammation in areas of fibrosis: The DeKAF prospective cohort. Am J Transplant 2020; 20:2509-2521. [PMID: 32185865 DOI: 10.1111/ajt.15862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/13/2020] [Accepted: 02/27/2020] [Indexed: 01/25/2023]
Abstract
Inflammation in areas of fibrosis (i-IFTA) in posttransplant biopsy specimens has been associated with decreased death-censored graft survival (DC-GS). Additionally, an i-IFTA score ≥ 2 is part of the diagnostic criteria for chronic active TCMR (CA TCMR). We examined the impact of i-IFTA and t-IFTA (tubulitis in areas of atrophy) in the first biopsy for cause after 90 days posttransplant (n = 598); mean (SD) 1.7 ± 1.4 years posttransplant. I-IFTA, present in 196 biopsy specimens, was strongly correlated with t-IFTA, and Banff i. Of the 196, 37 (18.9%) had a previous acute rejection episode; 96 (49%) had concurrent i score = 0. Unlike previous studies, i-IFTA = 1 (vs 0) was associated with worse 3-year DC-GS: (i-IFTA = 0, 81.7%, [95% CI 77.7 to 85.9%]); i-IFTA = 1, 68.1%, [95% CI 59.7 to 77.6%]; i-IFTA = 2, 56.1%, [95% CI 43.2 to 72.8%], i-IFTA = 3, 48.5%, [95% CI 31.8 to 74.0%]). The association of i-IFTA with decreased DC-GS remained significant when adjusted for serum creatinine at the time of the biopsy, Banff i, ci and ct, C4d and DSA. T-IFTA was similarly associated with decreased DC-GS. Of these indication biopsies, those with i-IFTA ≥ 2, without meeting other criteria for CA TCMR had similar postbiopsy DC-GS as those with CA TCMR. Those with i-IFTA = 1 and t ≥ 2, ti ≥ 2 had postbiopsy DC-GS similar to CA TCMR. Biopsies with i-IFTA = 1 had similar survival as CA TCMR when biopsy specimens also met Banff criteria for TCMR and/or AMR. Studies of i-IFTA and t-IFTA in additional cohorts, integrating analyses of Banff scores meeting criteria for other Banff diagnoses, are needed.
Collapse
Affiliation(s)
- Arthur J Matas
- Transplantation Division, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Erika S Helgeson
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Robert Gaston
- Department of Medicine, University of Alabama, Birmingham, Alabama, USA
| | | | - Roslyn Mannon
- Department of Medicine, University of Alabama, Birmingham, Alabama, USA
| | | | - Lawrence Hunsicker
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Sita Gourishankar
- Division of Nephrology, Department of Medicine, Univeristy of Alberta, Edmonton, Alberta, Canada
| | - David Rush
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - J Michael Cecka
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California, USA
| | - John Connett
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
55
|
Rush DN. Subclinical Rejection: a Universally Held Concept? CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00290-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
56
|
Fu Q, Liao M, Feng C, Tang J, Liao R, Wei L, Yang H, Markmann JF, Chen K, Deng S. Profiling of mRNA of interstitial fibrosis and tubular atrophy with subclinical inflammation in recipients after kidney transplantation. Aging (Albany NY) 2020; 11:5215-5231. [PMID: 31343413 PMCID: PMC6682514 DOI: 10.18632/aging.102115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/16/2019] [Indexed: 12/18/2022]
Abstract
Interstitial fibrosis and tubular atrophy (IFTA) with inflammation (IFTA-I) is strongly correlated with kidney allograft failure. Diagnosis of IFTA-I accurately and early is critical to prevent graft failure and improve graft survival. In the current study, through analyzing the renal allograft biopsy in patients with stable function after kidney transplantation (STA), IFTA and IFTA-I group with semi-supervised principal components methods, we found that CD2, IL7R, CCL5 based signature could not only distinguish STA and IFTA-I well, but predict IFTA-I with a high degree of accuracy with an area under the curve (AUC) of 0.91 (P = 0.00023). Additionally, IRF8 demonstrated significant differences among STA, IFTA and IFTA-I groups, suggesting that IRF8 had the capacity to discriminate the different classifications of graft biopsies well. Also, with Kaplan-Meier and log-rank methods, we found that IRF8 could serve as the prognostic marker for renal graft failure in those biopsies without rejection (AUC = 0.75) and the recipients expressing high had a higher risk for renal graft loss (P < 0.0001). This research may provide new targets for therapeutic prevention and intervention for post-transplantation IFTA with or with inflammation.
Collapse
Affiliation(s)
- Qiang Fu
- Organ Transplantation Center, Sichuan Provincial People's Hospital and School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.,Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02148, USA.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China
| | - Minxue Liao
- Organ Transplantation Center, Sichuan Provincial People's Hospital and School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China.,North Sichuan Medical College, Nanchong 637100, Sichuan, China
| | - Cheng Feng
- Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China.,Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Jichao Tang
- Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China.,Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Rui Liao
- Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China.,Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Liang Wei
- Organ Transplantation Center, Sichuan Provincial People's Hospital and School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China
| | - Hongji Yang
- Organ Transplantation Center, Sichuan Provincial People's Hospital and School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China.,North Sichuan Medical College, Nanchong 637100, Sichuan, China.,Southwest Medical University, Luzhou 646000, Sichuan, China
| | - James F Markmann
- Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02148, USA
| | - Kai Chen
- Organ Transplantation Center, Sichuan Provincial People's Hospital and School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China
| | - Shaoping Deng
- Organ Transplantation Center, Sichuan Provincial People's Hospital and School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.,Transplant Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02148, USA.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan, China.,North Sichuan Medical College, Nanchong 637100, Sichuan, China.,Southwest Medical University, Luzhou 646000, Sichuan, China
| |
Collapse
|
57
|
Abstract
The standardization of renal allograft pathology began in 1991 at the first Banff Conference held in Banff, Alberta, Canada. The first task of transplant pathologists, clinicians, and surgeons was to establish diagnostic criteria for T-cell-mediated rejection (TCMR). The histological threshold for this diagnosis was arbitrarily set at "i2t2": a mononuclear interstitial cell infiltrate present in at least 25% of normal parenchyma and >4 mononuclear cells within the tubular basement membrane of nonatrophic tubules. TCMR was usually found in dysfunctional grafts with an elevation in the serum creatinine; however, our group and others found this extent of inflammation in "routine" or "protocol" biopsies of normally functioning grafts: "subclinical" TCMR. The prevalence of TCMR is higher in the early months posttransplant and has decreased with the increased potency of current immunosuppressive agents. However, the pathogenicity of lesser degrees of inflammation under modern immunosuppression and the relation between ongoing inflammation and development of donor-specific antibody has renewed our interest in subclinical alloreactivity. Finally, the advances in our understanding of pretransplant risk assessment, and our increasing ability to monitor patients less invasively posttransplant, promises to usher in the era of precision medicine.
Collapse
|
58
|
The Pathophysiology and Impact of Inflammation in Nonscarred Renal Interstitium: The Banff i Lesion. Transplantation 2020; 104:835-846. [DOI: 10.1097/tp.0000000000002887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
59
|
Moreso F, Sellarès J, Soler MJ, Serón D. Transcriptome Analysis in Renal Transplant Biopsies Not Fulfilling Rejection Criteria. Int J Mol Sci 2020; 21:ijms21062245. [PMID: 32213927 PMCID: PMC7139324 DOI: 10.3390/ijms21062245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/11/2020] [Accepted: 03/20/2020] [Indexed: 01/02/2023] Open
Abstract
The clinical significance of renal transplant biopsies displaying borderline changes suspicious for T-cell mediated rejection (TCMR) or interstitial fibrosis and tubular atrophy (IFTA) with interstitial inflammation has not been well defined. Molecular profiling to evaluate renal transplant biopsies using microarrays has been shown to be an objective measurement that adds precision to conventional histology. We review the contribution of transcriptomic analysis in surveillance and indication biopsies with borderline changes and IFTA associated with variable degrees of inflammation. Transcriptome analysis applied to biopsies with borderline changes allows to distinguish patients with rejection from those in whom mild inflammation mainly represents a response to injury. Biopsies with IFTA and inflammation occurring in unscarred tissue display a molecular pattern similar to TCMR while biopsies with IFTA and inflammation in scarred tissue, apart from T-cell activation, also express B cell, immunoglobulin and mast cell-related genes. Additionally, patients at risk for IFTA progression can be identified by genes mainly reflecting fibroblast dysregulation and immune activation. At present, it is not well established whether the expression of rejection gene transcripts in patients with fibrosis and inflammation is the consequence of an alloimmune response, tissue damage or a combination of both.
Collapse
|
60
|
Kidney allograft fibrosis: what we learned from latest translational research studies. J Nephrol 2020; 33:1201-1211. [PMID: 32193834 DOI: 10.1007/s40620-020-00726-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
To add new molecular and pathogenetic insights into the biological machinery associated to kidney allograft fibrosis is a major research target in nephrology and organ transplant translational medicine. Interstitial fibrosis associated to tubular atrophy (IF/TA) is, in fact, an inevitable and progressive process that occurs in almost every type of chronic allograft injury (particularly in grafts from expanded criteria donors) characterized by profound remodeling and excessive production/deposition of fibrillar extracellular matrix (ECM) with a great clinical impact. IF/TA is detectable in more than 50% of kidney allografts at 2 years. However, although well studied, the complete cellular/biological network associated with IF/TA is only partially evaluated. In the last few years, then, thanks to the introduction of new biomolecular technologies, inflammation in scarred/fibrotic parenchyma areas (recently acknowledged by the BANFF classification) has been recognized as a pivotal element able to accelerate the onset and development of the allograft chronic damage. Therefore, in this review, we focused on some new pathogenetic elements involved in graft fibrosis (including epithelial/endothelial to mesenchymal transition, oxidative stress, activation of Wnt and Hedgehog signaling pathways, fatty acids oxidation and cellular senescence) that, in our opinion, could become in future good candidates as potential biomarkers and therapeutic targets.
Collapse
|
61
|
Rivero J, Rodríguez F, Soto V, Macedo E, Chawla LS, Mehta RL, Vaingankar S, Garimella PS, Garza C, Madero M. Furosemide stress test and interstitial fibrosis in kidney biopsies in chronic kidney disease. BMC Nephrol 2020; 21:87. [PMID: 32143585 PMCID: PMC7060600 DOI: 10.1186/s12882-020-01721-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 02/11/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Interstitial fibrosis (IF) on kidney biopsy is one of the most potent risk factors for kidney disease progression. The furosemide stress test (FST) is a validated tool that predicts the severity of acute kidney injury (especially at 2 h) in critically ill patients. Since furosemide is secreted through the kidney tubules, the response to FST represents the tubular secretory capacity. To our knowledge there is no data on the correlation between functional tubular capacity assessed by the FST with IF on kidney biopsies from patients with chronic kidney disease (CKD). The aim of this study was to determine the association between urine output (UO), Furosemide Excreted Mass (FEM) and IF on kidney biopsies after a FST. METHODS This study included 84 patients who underwent kidney biopsy for clinical indications and a FST. The percentage of fibrosis was determined by morphometry technique and reviewed by a nephropathologist. All patients underwent a FST prior to the biopsy. Urine volume and urinary sodium were measured in addition to urine concentrations of furosemide at different times (2, 4 and 6 h). We used an established equation to determine the FEM. Values were expressed as mean, standard deviation or percentage and Pearson Correlation. RESULTS The mean age of the participants was 38 years and 44% were male. The prevalence of diabetes mellitus, hypertension and diuretic use was significantly higher with more advanced degree of fibrosis. Nephrotic syndrome and acute kidney graft dysfunction were the most frequent indications for biopsy. eGFR was inversely related to the degree of fibrosis. Subjects with the highest degree of fibrosis (grade 3) showed a significant lower UO at first hour of the FST when compared to lower degrees of fibrosis (p = 0.015). Likewise, the total UO and the FEM was progressively lower with higher degrees of fibrosis. An inversely linear correlation between FEM and the degree of fibrosis (r = - 0.245, p = 0.02) was observed. CONCLUSIONS Our findings indicate that interstitial fibrosis correlates with total urine output and FEM. Further studies are needed to determine if UO and FST could be a non-invasive tool to evaluate interstitial fibrosis. TRIAL REGISTRATION ClinicalTrials.gov NCT02417883.
Collapse
Affiliation(s)
- Jesús Rivero
- Nephrology Department, National Institute of Lung Disease Ismael Cosio Villegas , Mexico City, Mexico
| | - Francisco Rodríguez
- Nephrology Department, National Institute Cardiology Ignacio Chávez, Juan Badiano No. 1, 14080-Tlalpan, Mexico City, Mexico
| | - Virgilia Soto
- Nephrology Department, National Institute Cardiology Ignacio Chávez, Juan Badiano No. 1, 14080-Tlalpan, Mexico City, Mexico
| | - Etienne Macedo
- Division of Nephrology, University of California, San Diego, CA, USA
| | - Lakhmir S Chawla
- Division of Nephrology, University of California, San Diego, CA, USA
| | - Ravindra L Mehta
- Division of Nephrology, University of California, San Diego, CA, USA
| | - Sucheta Vaingankar
- Nephrology Department, National Institute Cardiology Ignacio Chávez, Juan Badiano No. 1, 14080-Tlalpan, Mexico City, Mexico
| | | | - Carlos Garza
- Nephrology Department, National Institute Cardiology Ignacio Chávez, Juan Badiano No. 1, 14080-Tlalpan, Mexico City, Mexico
| | - Magdalena Madero
- Nephrology Department, National Institute Cardiology Ignacio Chávez, Juan Badiano No. 1, 14080-Tlalpan, Mexico City, Mexico.
| |
Collapse
|
62
|
The Causes of Kidney Allograft Failure: More Than Alloimmunity. A Viewpoint Article. Transplantation 2020; 104:e46-e56. [DOI: 10.1097/tp.0000000000003012] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
63
|
Khairwa A. The relevance of complement C4d staining in renal allograft biopsies. INDIAN JOURNAL OF TRANSPLANTATION 2020. [DOI: 10.4103/ijot.ijot_60_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
64
|
Hofmann BB, Krapp N, Li Y, De La Torre C, Sol M, Braun JD, Kolibabka M, Pallavi P, Krämer BK, Yard BA, Kälsch AI. N-Octanoyl-Dopamine inhibits cytokine production in activated T-cells and diminishes MHC-class-II expression as well as adhesion molecules in IFNγ-stimulated endothelial cells. Sci Rep 2019; 9:19338. [PMID: 31853095 PMCID: PMC6920350 DOI: 10.1038/s41598-019-55983-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/29/2019] [Indexed: 01/06/2023] Open
Abstract
IFNγ enhances allograft immunogenicity and facilitates T-cell mediated rejection. This may cause interstitial fibrosis and tubular atrophy (IFTA), contributing to chronic allograft loss. We assessed if inhibition of T-cell activation by N-octanoyl dopamine (NOD) impairs adherence of activated T-cells to endothelial cells and the ability of activated T-cells to produce IFNγ. We also assessed if NOD affects IFNγ mediated gene expression in endothelial cells. The presence of NOD during T-cell activation significantly blunted their adhesion to unstimulated and cytokine stimulated HUVEC. Supernatants of these T-cells displayed significantly lower concentrations of TNFα and IFNγ and were less capable to facilitate T-cell adhesion. In the presence of NOD VLA-4 (CD49d/CD29) and LFA-1 (CD11a/CD18) expression on T-cells was reduced. NOD treatment of IFNγ stimulated HUVEC reduced the expression of MHC class II transactivator (CIITA), of MHC class II and its associated invariant chain CD74. Since IFTA is associated with T-cell mediated rejection and IFNγ to a large extent regulates immunogenicity of allografts, our current data suggest a potential clinical use of NOD in the treatment of transplant recipients. Further in vivo studies are warranted to confirm these in vitro findings and to assess the benefit of NOD on IFTA in clinically relevant models.
Collapse
Affiliation(s)
- Björn B Hofmann
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nicolas Krapp
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Yingchun Li
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carolina De La Torre
- Center of Medical Research, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marloes Sol
- Department of Medical Biology and Pathology, University Medical Center Groningen, Groningen, Netherlands
| | - Jana D Braun
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Matthias Kolibabka
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Prama Pallavi
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bernhard K Krämer
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Benito A Yard
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| | - Anna-Isabelle Kälsch
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
65
|
Taneda S, Koike J, Oda H, Honda K. Practical Issues and Future Perspectives for Inflammation in Areas of Interstitial Fibrosis and Tubular Atrophy in Chronic Active T cell-Mediated Rejection: Three Case Reports with Commentary. Transplant Proc 2019; 51:3286-3292. [PMID: 31732216 DOI: 10.1016/j.transproceed.2019.08.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/30/2019] [Indexed: 10/25/2022]
Abstract
The 2017 Banff meeting provided specific criteria for the diagnosis of tubulointerstitial changes in chronic active T cell-mediated rejection (CATCR), with an emphasis on inflammation in areas of interstitial fibrosis and tubular atrophy, which was thought to reflect an ongoing T cell-mediated alloimmunity. CATCR is considered to occur as a consequence of persistent or recurrent acute T cell-mediated rejection. Acute T cell-mediated rejection is an acute cytotoxic T-cell reaction to HLA antigens on the donor kidneys and causes tubulitis, interstitial inflammation, and intimal arteries. However, unlike early T-cell transplant damage, CATCR can sometimes be difficult to diagnose because the subsequent chronic T-cell damage can become more complex from the accumulation of previous immune and nonimmune injuries. Furthermore, scoring inflammation in areas of interstitial fibrosis and tubular atrophy has potential problems because other diseases and not even native kidneys can have scattered inflammatory cells. Therefore, detailed insights on the pathogenesis of CATCR are indispensable for appropriate diagnosis and further treatment. In this study, the pathologic characteristics and possible factors involved in the interstitial lesions in both typical and complex cases of CATCR are discussed.
Collapse
Affiliation(s)
- Sekiko Taneda
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Junki Koike
- Department of Pathology, St. Marianna University, Tokyo, Japan
| | - Hideaki Oda
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuho Honda
- Department of Anatomy, Showa University School of Medicine, Kanagawa, Japan
| |
Collapse
|
66
|
Mariani LH, Martini S, Barisoni L, Canetta PA, Troost JP, Hodgin JB, Palmer M, Rosenberg AZ, Lemley KV, Chien HP, Zee J, Smith A, Appel GB, Trachtman H, Hewitt SM, Kretzler M, Bagnasco SM. Interstitial fibrosis scored on whole-slide digital imaging of kidney biopsies is a predictor of outcome in proteinuric glomerulopathies. Nephrol Dial Transplant 2019; 33:310-318. [PMID: 28339906 DOI: 10.1093/ndt/gfw443] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022] Open
Abstract
Background Interstitial fibrosis (IF), tubular atrophy (TA) and interstitial inflammation (II) are known determinants of progression of renal disease. Standardized quantification of these features could add value to current classification of glomerulopathies. Methods We studied 315 participants in the Nephrotic Syndrome Study Network (NEPTUNE) study, including biopsy-proven minimal change disease (MCD = 98), focal segmental glomerulosclerosis (FSGS = 121), membranous nephropathy (MN = 59) and IgA nephropathy (IgAN = 37). Cortical IF, TA and II were quantified (%) on digitized whole-slide biopsy images, by five pathologists with high inter-reader agreement (intra-class correlation coefficient >0.8). Tubulointerstitial messenger RNA expression was measured in a subset of patients. Multivariable Cox proportional hazards models were fit to assess association of IF with the composite of 40% decline in estimated glomerular filtration rate (eGFR) and end-stage renal disease (ESRD) and separately as well, and with complete remission (CR) of proteinuria. Results IF was highly correlated with TA (P < 0.001) and II (P < 0.001). Median IF varied by diagnosis: FSGS 17, IgAN 21, MN 7, MCD 1 (P < 0.001). IF was strongly correlated with baseline eGFR (P < 0.001) and proteinuria (P = 0.002). After adjusting for clinical pathologic diagnosis, age, race, global glomerulosclerosis, baseline proteinuria, eGFR and medications, each 10% increase in IF was associated with a hazard ratio of 1.29 (P < 0.03) for ESRD/40% eGFR decline, but was not significantly associated with CR. A total of 981 genes were significantly correlated with IF (|r| > 0.4, false discovery rate (FDR) < 0.01), including upstream regulators such as tumor necrosis factor, interferon gamma (IFN-gamma), and transforming growth factor beta 1 (TGF-B1), and signaling pathways for antigen presentation and hepatic fibrosis. Conclusions The degree of IF is associated with risk of eGFR decline across different types of proteinuric glomerulopathy, correlates with inflammatory and fibrotic gene expression, and may have predictive value in assessing risk of progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jarcy Zee
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA
| | - Abigail Smith
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA
| | | | | | | | | | | |
Collapse
|
67
|
Bouatou Y, Viglietti D, Pievani D, Louis K, Duong Van Huyen JP, Rabant M, Aubert O, Taupin JL, Glotz D, Legendre C, Loupy A, Lefaucheur C. Response to treatment and long-term outcomes in kidney transplant recipients with acute T cell-mediated rejection. Am J Transplant 2019; 19:1972-1988. [PMID: 30748089 DOI: 10.1111/ajt.15299] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/31/2018] [Accepted: 01/22/2019] [Indexed: 01/25/2023]
Abstract
The recent recognition of complex and chronic phenotypes of T cell-mediated rejection (TCMR) has fostered the need to better evaluate the response of acute TCMR-a condition previously considered to lack relevant consequences for allograft survival-to the standard of care. In a prospective cohort of kidney recipients (n = 256) with biopsy-proven acute TCMR receiving corticosteroids, we investigated clinical, histological, and immunological phenotypes at the time of acute TCMR diagnosis and 3 months posttreatment. Independent posttreatment determinants of allograft loss included the glomerular filtration rate (GFR) (HR = 0.94; 95% CI = 0.92-0.96; P < .001), proteinuria (HR = 1.40; 95% CI = 1.10-1.79; P = .007), time since transplantation (HR = 1.02; 95% CI = 1.00-1.03; P = .016), peritubular capillaritis (HR = 2.27; 95% CI = 1.13-4.55; P = .022), interstitial inflammation in sclerotic cortical parenchyma (i-IF/TA) (HR = 1.87; 95% CI = 1.08-3.25; P = .025), and donor-specific anti-HLA antibodies (DSAs) (HR = 2.67; 95% CI = 1.46-4.88; P = .001). Prognostic value was improved using a composite evaluation of response to treatment versus clinical parameters only (cNRI = 0.68; 95% CI = 0.41-0.95; P < .001). A classification tree for allograft loss identified five patterns of response to treatment based on the posttreatment GFR, i-IF/TA, and anti-HLA DSAs (cross-validated accuracy = 0.80). Compared with responders (n = 155, 60.5%), nonresponders (n = 101, 39.5%) had a higher incidence of de novo DSAs, antibody-mediated rejection, and allograft loss at 10 years (P < .001 for all comparisons). Thus, clinical, histological, and immunological assessment of response to treatment of acute TCMR revealed different profiles of the response to treatment with distinct outcomes.
Collapse
Affiliation(s)
- Yassine Bouatou
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Denis Viglietti
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Daniele Pievani
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Kevin Louis
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Jean-Paul Duong Van Huyen
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Pathology Department, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Marion Rabant
- Pathology Department, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Olivier Aubert
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-Luc Taupin
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Department of Immunology and Histocompatibility, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Denis Glotz
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Christophe Legendre
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Carmen Lefaucheur
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
68
|
Kettler B, Scheffner I, Bräsen JH, Hallensleben M, Richter N, Heiringhoff KH, Lehner F, Klempnauer J, Gwinner W. Kidney graft survival of >25 years: a single center report including associated graft biopsy results. Transpl Int 2019; 32:1277-1285. [PMID: 31188498 DOI: 10.1111/tri.13469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/29/2018] [Accepted: 06/07/2019] [Indexed: 11/28/2022]
Abstract
Only few centers have reported their observations on patients with very long-term kidney graft survival of more than 25 years. Eighty-six subjects were identified in our center with graft survival of >25 years. Donor age was 31.3 ± 18.5 years. Mean duration of transplantation was 30.3 ± 3.6 years. At last follow-up, the cystatin C clearance was 47 ± 23 ml/min. Transplant biopsies for cause were performed in 30 subjects at a median of 28.4 years (19.1-40.3) after transplantation. Acute or chronic active T cell-mediated rejection was present in five cases and histological characteristics of acute or chronic active humoral rejection in eight cases. More than 80% of biopsies had inflammatory infiltrates in nonatrophic or atrophic cortical areas. The number of HLA mismatches were higher in biopsied subjects (3.0 ± 1.8 vs. 2.2 ± 1.7 without biopsy). Immunosuppressive therapy was adapted in most biopsied subjects; impaired graft function and proteinuria was unchanged at last follow-up. Sixty percent of all subjects had hyperparathyroidism (iPTH of the whole group: 132 ± 157 pg/ml), which was predominantly secondary, as judged by serum calcium and graft function. Young donor age was certainly a prerequisite of longterm graft survival. Nonetheless, inflammation or rejection in most biopsied patients suggests an important role of alloreactivity even in this late course.
Collapse
Affiliation(s)
- Bastian Kettler
- Clinic for General-, Abdominal- and Transplant-Surgery, Medical School Hannover, Hannover, Germany
| | - Irina Scheffner
- Clinic for Nephrology, Medical School Hannover, Hannover, Germany
| | - Jan H Bräsen
- Institute for Transfusion Medicine, Medical School Hannover, Hannover, Germany
| | - Michael Hallensleben
- Nephropathology Unit, Institute for Pathology, Medical School Hannover, Hannover, Germany
| | - Nicolas Richter
- Clinic for General-, Abdominal- and Transplant-Surgery, Medical School Hannover, Hannover, Germany
| | - Karl-Heinz Heiringhoff
- Clinic for General-, Abdominal- and Transplant-Surgery, Medical School Hannover, Hannover, Germany
| | - Frank Lehner
- Clinic for General-, Abdominal- and Transplant-Surgery, Medical School Hannover, Hannover, Germany
| | - Jürgen Klempnauer
- Clinic for General-, Abdominal- and Transplant-Surgery, Medical School Hannover, Hannover, Germany
| | - Wilfried Gwinner
- Clinic for Nephrology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
69
|
Halloran PF, Matas A, Kasiske BL, Madill-Thomsen KS, Mackova M, Famulski KS. Molecular phenotype of kidney transplant indication biopsies with inflammation in scarred areas. Am J Transplant 2019; 19:1356-1370. [PMID: 30417539 DOI: 10.1111/ajt.15178] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/02/2018] [Accepted: 11/02/2018] [Indexed: 01/25/2023]
Abstract
In kidney transplant biopsies, inflammation in areas of atrophy-fibrosis (i-IFTA) is associated with increased risk of failure, presumably because inflammation is evoked by recent parenchymal injury from rejection or other insults, but some cases also have rejection. The present study explored the frequency of rejection in i-IFTA, by using histology Banff 2015 and a microarray-based molecular diagnostic system (MMDx). In unselected indication biopsies (108 i-IFTA, 73 uninflamed IFTA [i0-IFTA], and 53 no IFTA), i-IFTA biopsies occurred later, showed more scarring, and had more antibody-mediated rejection (ABMR) based on histology (28%) and MMDx (45%). T cell-mediated rejection (TCMR) was infrequent in i-IFTA based on histology (8%) and MMDx (16%). Twelve i-IFTA biopsies (11%) had molecular TCMR not diagnosed by histology, although 6 were called borderline and almost all had histologic TCMR lesions. The prominent feature of i-IFTA biopsies was molecular injury (eg, acute kidney injury [AKI] transcripts). In multivariate analysis of biopsies >1 year posttransplant, the strongest associations with graft loss were AKI transcripts and histologic atrophy-scarring; i-IFTA was not significant when molecular AKI was included. We conclude that i-IFTA in indication biopsies reflects recent/ongoing parenchymal injury, often with concomitant ABMR but few with TCMR. Thus, the application of Banff i-IFTA in the population of late biopsies needs to be reconsidered.
Collapse
Affiliation(s)
- Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Arthur Matas
- Department of Surgery, University of Minnesota at Fairview, Minneapolis, Minnesota
| | | | - Katelynn S Madill-Thomsen
- Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Martina Mackova
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
| | - Konrad S Famulski
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
| |
Collapse
|
70
|
Matas AJ, Fieberg A, Mannon RB, Leduc R, Grande J, Kasiske BL, Cecka M, Gaston R, Hunsicker L, Connett J, Cosio F, Gourishankar S, Rush D. Long-term follow-up of the DeKAF cross-sectional cohort study. Am J Transplant 2019; 19:1432-1443. [PMID: 30506642 PMCID: PMC7653899 DOI: 10.1111/ajt.15204] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/24/2018] [Accepted: 11/19/2018] [Indexed: 01/25/2023]
Abstract
The DeKAF study was developed to better understand the causes of late allograft loss. Preliminary findings from the DeKAF cross-sectional cohort (with follow-up < 20 months) have been published. Herein, we present long-term outcomes in those recipients (mean follow-up ± SD, 6.6 ± 0.7 years). Eligibility included being transplanted prior to October 1, 2005; serum creatinine ≤ 2.0 mg/dL on January 1, 2006; and subsequently developing new-onset graft dysfunction leading to a biopsy. Mean time from transplant to biopsy was 7.5 ± 6.1 years. Histologic findings and DSA were studied in relation to postbiopsy outcomes. Long-term follow-up confirms and expands the preliminary results of each of 3 studies: (1) increasing inflammation in area of atrophy (irrespective of inflammation in nonscarred areas [Banff i]) was associated with increasingly worse postbiopsy death-censored graft survival; (2) hierarchical analysis based on Banff scores defined clusters (entities) that differed in long-term death-censored graft survival; and (3) C4d-/DSA- recipients had significantly better (and C4d+/DSA+ worse) death-censored graft survival than other groups. C4d+/DSA- and C4d-/DSA+ had similar intermediate death-censored graft survival. Clinical and histologic findings at the time of new-onset graft dysfunction define high- vs low-risk groups for long-term death-censored graft survival, even years posttransplant. These findings can help differentiate groups for potential intervention studies.
Collapse
Affiliation(s)
- Arthur J. Matas
- Transplantation Division, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Ann Fieberg
- Biostatistics Division, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Roslyn B. Mannon
- Department of Nephrology, University of Alabama, Birmingham, Alabama
| | - Robert Leduc
- Biostatistics Division, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Joe Grande
- Nephrology and Transplant Center, Mayo Clinic, Rochester, Minnesota
| | - Bertram L. Kasiske
- Chronic Disease and Research Group, Hennepin Healthcare, Minneapolis, Minnesota
| | - Michael Cecka
- Ronald Reagan UCLA Medicine Center, University of California, Los Angeles, California
| | - Robert Gaston
- Department of Nephrology, University of Alabama, Birmingham, Alabama
| | | | - John Connett
- Biostatistics Division, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Fernando Cosio
- Nephrology and Transplant Center, Mayo Clinic, Rochester, Minnesota
| | - Sita Gourishankar
- Division of Nephrology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - David Rush
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
71
|
Ho J, Sharma A, Kroeker K, Carroll R, De Serres S, Gibson IW, Hirt-Minkowski P, Jevnikar A, Kim SJ, Knoll G, Rush DN, Wiebe C, Nickerson P. Multicentre randomised controlled trial protocol of urine CXCL10 monitoring strategy in kidney transplant recipients. BMJ Open 2019; 9:e024908. [PMID: 30975673 PMCID: PMC6500325 DOI: 10.1136/bmjopen-2018-024908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION Subclinical inflammation is an important predictor of death-censored graft loss, and its treatment has been shown to improve graft outcomes. Urine CXCL10 outperforms standard post-transplant surveillance in observational studies, by detecting subclinical rejection and early clinical rejection before graft functional decline in kidney transplant recipients. METHODS AND ANALYSIS This is a phase ii/iii multicentre, international randomised controlled parallel group trial to determine if the early treatment of rejection, as detected by urine CXCL10, will improve kidney allograft outcomes. Incident adult kidney transplant patients (n~420) will be enrolled to undergo routine urine CXCL10 monitoring postkidney transplant. Patients at high risk of rejection, defined as confirmed elevated urine CXCL10 level, will be randomised 1:1 stratified by centre (n=250). The intervention arm (n=125) will undergo a study biopsy to check for subclinical rejection and biopsy-proven rejection will be treated per protocol. The control arm (n=125) will undergo routine post-transplant monitoring. The primary outcome at 12 months is a composite of death-censored graft loss, clinical biopsy-proven acute rejection, de novo donor-specific antibody, inflammation in areas of interstitial fibrosis and tubular atrophy (Banff i-IFTA, chronic active T-cell mediated rejection) and subclinical tubulitis on 12-month surveillance biopsy. The secondary outcomes include decline of graft function, microvascular inflammation at 12 months, development of IFTA at 12 months, days from transplantation to clinical biopsy-proven rejection, albuminuria, EuroQol five-dimension five-level instrument, cost-effectiveness analysis of the urine CXCL10 monitoring strategy and the urine CXCL10 kinetics in response to rejection therapy. ETHICS AND DISSEMINATION The study has been approved by the University of Manitoba Health Research Ethics Board (HS20861, B2017:076) and the local research ethics boards of participating centres. Recruitment commenced in March 2018 and results are expected to be published in 2023. De-identified data may be shared with other researchers according to international guidelines (International Committee of Medical Journal Editors [ICJME]). TRIAL REGISTRATION NUMBER NCT03206801; Pre-results.
Collapse
Affiliation(s)
- Julie Ho
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
- Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Atul Sharma
- Data Science, George and Fay Yee Centre for Healthcare Innovation, Winnipeg, Manitoba, Canada
| | - Kristine Kroeker
- Data Science, George and Fay Yee Centre for Healthcare Innovation, Winnipeg, Manitoba, Canada
| | - Robert Carroll
- Transplant Nephrology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Sacha De Serres
- Internal Medicine & Nephrology, Universite Laval, Québec, Québec, Canada
| | - Ian W Gibson
- Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Anthony Jevnikar
- Internal Medicine & Nephrology, Western University, London, Ontario, Canada
| | - S Joseph Kim
- Internal Medicine & Nephrology, University of Toronto, Toronto, Ontario, Canada
| | - Greg Knoll
- Internal Medicine & Nephrology, University of Ottawa, Ottawa, Ontario, Canada
| | - David N Rush
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
| | - Chris Wiebe
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
| | - Peter Nickerson
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
| |
Collapse
|
72
|
Urine Angiotensin II Signature Proteins as Markers of Fibrosis in Kidney Transplant Recipients. Transplantation 2019; 103:e146-e158. [PMID: 30801542 DOI: 10.1097/tp.0000000000002676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Interstitial fibrosis/tubular atrophy (IFTA) is an important cause of kidney allograft loss; however, noninvasive markers to identify IFTA or guide antifibrotic therapy are lacking. Using angiotensin II (AngII) as the prototypical inducer of IFTA, we previously identified 83 AngII-regulated proteins in vitro. We developed mass spectrometry-based assays for quantification of 6 AngII signature proteins (bone marrow stromal cell antigen 1, glutamine synthetase [GLNA], laminin subunit beta-2, lysophospholipase I, ras homolog family member B, and thrombospondin-I [TSP1]) and hypothesized that their urine excretion will correlate with IFTA in kidney transplant patients. METHODS Urine excretion of 6 AngII-regulated proteins was quantified using selected reaction monitoring and normalized by urine creatinine. Immunohistochemistry was used to assess protein expression of TSP1 and GLNA in kidney biopsies. RESULTS The urine excretion rates of AngII-regulated proteins were found to be increased in 15 kidney transplant recipients with IFTA compared with 20 matched controls with no IFTA (mean log2[fmol/µmol of creatinine], bone marrow stromal cell antigen 1: 3.8 versus 3.0, P = 0.03; GLNA: 1.2 versus -0.4, P = 0.03; laminin subunit beta-2: 6.1 versus 5.4, P = 0.06; lysophospholipase I: 2.1 versus 0.6, P = 0.002; ras homolog family member B: 1.2 versus -0.1, P = 0.006; TSP1_GGV: 2.5 versus 1.9; P = 0.15; and TSP1_TIV: 2.0 versus 0.6, P = 0.0006). Receiver operating characteristic curve analysis demonstrated an area under the curve = 0.86 for the ability of urine AngII signature proteins to discriminate IFTA from controls. Urine excretion of AngII signature proteins correlated strongly with chronic IFTA and total inflammation. In a separate cohort of 19 kidney transplant recipients, the urine excretion of these 6 proteins was significantly lower following therapy with AngII inhibitors (P < 0.05). CONCLUSIONS AngII-regulated proteins may represent markers of IFTA and guide antifibrotic therapies.
Collapse
|
73
|
Nankivell BJ, P'Ng CH, Chapman JR. Does tubulitis without interstitial inflammation represent borderline acute T cell mediated rejection? Am J Transplant 2019; 19:132-144. [PMID: 29687946 DOI: 10.1111/ajt.14888] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 03/30/2018] [Accepted: 03/31/2018] [Indexed: 01/25/2023]
Abstract
Tubulitis without interstitial inflammation (Banff i0), termed "isolated tubulitis" (ISO-T), has been controversially included within the Banff "borderline" category of acute T cell mediated rejection (TCMR). This single-center, retrospective, observational study of 2055 consecutive biopsies from 775 recipients, determined the clinical significance of ISO-T. ISO-T prevalence was 19.1%, comprising mild tubulitis (i0t1) in 97.2%. Independent clinical predictors of tubulitis were HLA mismatch, prior TCMR and antibody-mediated rejection, pulse corticosteroids, and BKVAN (P = .006 to P < .001 by multivariable analysis). Histological associations of tubulitis included interstitial inflammation, peritubular capillaritis, tubular atrophy, and SV40T (P = .005 to <.001). The dominant pathological diagnoses in ISO-T (n = 393) were interstitial fibrosis/tubular atrophy (IF/TA, 44.5%) or normal/minimal (31.8%). Subanalysis of ISO-T from indication biopsies (n = 107) found acute tubular injury (37.4%), IF/TA (28.0%), normal/minimal (12.1%), acute rejection (9.3%, vascular or antibody), chronic-active TCMR (2.8%), and BKVAN (5.6%). Allograft function of ISO-T frequently improved, affected by early biopsy timing and underlying disease diagnosis. Subsequent histology of 1197 ISO-T biopsy-pairs was generally benign. The 1- and 5-year death-censored graft survivals of ISO-T were 98.8% and 92.7%. In summary, tubulitis without inflammation does not represent borderline TCMR. We suggest its removal from the borderline category, and reinstatement of i1 as the diagnostic threshold.
Collapse
Affiliation(s)
| | - Chow H P'Ng
- Tissue Pathology and Diagnostic Oncology, ICPMR, Sydney, Australia
| | - Jeremy R Chapman
- Departments of Renal Medicine, Westmead Hospital, Sydney, Australia
| |
Collapse
|
74
|
Impact of the Current Versus the Previous Diagnostic Threshold on the Outcome of Patients With Borderline Changes Suspicious for T Cell–mediated Rejection Diagnosed on Indication Biopsies. Transplantation 2018; 102:2120-2125. [DOI: 10.1097/tp.0000000000002327] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
75
|
Roufosse C, Simmonds N, Clahsen-van Groningen M, Haas M, Henriksen KJ, Horsfield C, Loupy A, Mengel M, Perkowska-Ptasińska A, Rabant M, Racusen LC, Solez K, Becker JU. A 2018 Reference Guide to the Banff Classification of Renal Allograft Pathology. Transplantation 2018; 102:1795-1814. [PMID: 30028786 PMCID: PMC7597974 DOI: 10.1097/tp.0000000000002366] [Citation(s) in RCA: 517] [Impact Index Per Article: 73.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/07/2018] [Accepted: 05/23/2018] [Indexed: 12/24/2022]
Abstract
The Banff Classification of Allograft Pathology is an international consensus classification for the reporting of biopsies from solid organ transplants. Since its initial conception in 1991 for renal transplants, it has undergone review every 2 years, with attendant updated publications. The rapid expansion of knowledge in the field has led to numerous revisions of the classification. The resultant dispersal of relevant content makes it difficult for novices and experienced pathologists to faithfully apply the classification in routine diagnostic work and in clinical trials. This review shall provide a complete and simple illustrated reference guide of the Banff Classification of Kidney Allograft Pathology based on all publications including the 2017 update. It is intended as a concise desktop reference for pathologists and clinicians, providing definitions, Banff Lesion Scores and Banff Diagnostic Categories. An online website reference guide hosted by the Banff Foundation for Allograft Pathology (www.banfffoundation.org) is being developed, which will be updated with future refinement of the Banff Classification from 2019 onward.
Collapse
Affiliation(s)
- Candice Roufosse
- Department of Medicine, Imperial College, London, United Kingdom
- North West London Pathology, London, United Kingdom
| | - Naomi Simmonds
- Department of Histopathology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | | | - Mark Haas
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Catherine Horsfield
- Department of Histopathology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Paris, France
| | - Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | | | - Marion Rabant
- Department of Pathology, Necker Hospital University Paris Descartes, Paris, France
| | | | - Kim Solez
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Jan U. Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
76
|
Rankin AJ, Kipgen D, Geddes CC, Fox JG, Milne G, Mackinnon B, McQuarrie EP. Assessment of active tubulointerstitial nephritis in non-scarred renal cortex improves prediction of renal outcomes in patients with IgA nephropathy. Clin Kidney J 2018; 12:348-354. [PMID: 31198533 PMCID: PMC6543968 DOI: 10.1093/ckj/sfy093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Indexed: 12/13/2022] Open
Abstract
Background The addition of tubulointerstitial inflammation to the existing pathological classification of IgA nephropathy (IgAN) is appealing but was previously precluded due to reportedly wide inter-observer variability. We report a novel method to score percentage of non-atrophic renal cortex containing active tubulointerstitial inflammation (ATIN) in patients with IgAN and assess its utility to predict clinical outcomes. Methods All adult patients with a native renal biopsy diagnosis of IgAN between 2010 and 2015 in a unit serving 1.5 million people were identified. Baseline characteristics, biopsy reports and outcome data were collected. ATIN was calculated by subtracting the percentage of atrophic cortex from the percentage of total cortex with tubulointerstitial inflammation, with ≥10% representing significant ATIN. The primary outcome was a composite of requiring renal replacement therapy or doubling of serum creatinine. Results In total 153 new cases of IgAN were identified, of which 111 were eligible for inclusion. Of these, 76 (68%) were male and 54 (49%) had ATIN on biopsy. During a median follow-up of 2.3 years, 34 (31%) reached the primary outcome. On univariable Cox regression analysis, ATIN was associated with a five-fold increase in the primary outcome [hazard ratio (HR) (95% confidence interval) 4.9 (95% confidence interval (CI) 2.1–11.3)]. On multivariable analysis, mesangial hypercellularity, tubular atrophy and interstitial fibrosis and ATIN independently associated with renal outcome (P = 0.02 for ATIN). Inter-observer reproducibility revealed fair agreement in the diagnosis of ATIN (κ=0.43, P = 0.05). Conclusions Within our centre, ATIN was significantly associated with renal outcome in patients with IgAN, independently of established histological features and baseline clinical characteristics.
Collapse
Affiliation(s)
- Alastair J Rankin
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK.,Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - David Kipgen
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow, UK
| | - Colin C Geddes
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - Jonathan G Fox
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - Gordon Milne
- Department of Pathology, University Hospital Monklands, Airdrie, UK
| | - Bruce Mackinnon
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - Emily P McQuarrie
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| |
Collapse
|
77
|
Haas M. Mesoamerican nephropathy: pathology in search of etiology. Kidney Int 2018; 93:538-540. [PMID: 29475544 DOI: 10.1016/j.kint.2017.09.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 09/21/2017] [Accepted: 09/25/2017] [Indexed: 12/14/2022]
Abstract
Mesoamerican nephropathy is a progressive, often fatal form of tubulointerstitial nephritis affecting young agricultural laborers in Central America. Initially described as a chronic disease, a study by Fischer and coworkers in this issue of Kidney International suggests that Mesoamerican nephropathy goes through an active, inflammatory phase. Although the pathologic findings are nonspecific and the etiology of Mesoamerican nephropathy remains unclear, inflammatory infiltrates in areas of evolving and established interstitial fibrosis appear to cause progressive kidney injury.
Collapse
Affiliation(s)
- Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
78
|
Hara S. Cell mediated rejection revisited: Past, current, and future directions. Nephrology (Carlton) 2018; 23 Suppl 2:45-51. [PMID: 29968416 DOI: 10.1111/nep.13283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2018] [Indexed: 01/10/2023]
Abstract
The Banff histopathology classification system is the gold standard for assessing the causes of kidney allograft dysfunction triggered by antibody-mediated and T-cell-mediated immune reactions, thereby providing mechanistic insight and guiding therapeutic decisions. The original Banff classification (1993) consisted of four histological categories representing cell-mediated rejection: interstitial inflammation (i), tubulitis (t), endoarteritis (v), and transplant glomerulitis (g). The revised Banff 2007 classification added total inflammation score (ti) from both scarred and unscarred areas based on evolving interpretations of interstitial infiltrates. Further reappraisal of cell-mediated interstitial inflammation led to the introduction of a new inflammation score specific for areas of interstitial fibrosis and tubular atrophy, termed i-IF/TA, in the Banff 2015 scheme, establishment of a new Banff working group on T-cell-mediated rejection (TCMR), and revised criteria of chronic active TCMR in Banff 2017 classification. These Banff scheme updates reflect the general recognition that chronic interstitial inflammation is a common denominator of poor kidney allograft outcome. However, revised theories on the pathogenic importance of interstitial infiltrates have created difficulties in interpretation of chronic tubulointerstitial inflammation, as there are currently no histological criteria to discriminate immune-mediated tissue injury from 'non-specific' injury. Evolving theories on vascular lesions, both active and chronic, have also complicated histological assessment by obscuring the distinction between antibody-mediated and T-cell-mediated tissue injury. This review provides an overview of recent ideas on interstitial inflammation and vascular lesions based on emerging concepts of T-cell-mediated rejection.
Collapse
Affiliation(s)
- Shigeo Hara
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
79
|
Krezdorn N, Tasigiorgos S, Wo L, Lopdrup R, Turk M, Kiwanuka H, Ahmed S, Petruzzo P, Bueno E, Pomahac B, Riella LV. Kidney Dysfunction After Vascularized Composite Allotransplantation. Transplant Direct 2018; 4:e362. [PMID: 30046652 PMCID: PMC6056276 DOI: 10.1097/txd.0000000000000795] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 03/26/2018] [Accepted: 04/02/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Kidney dysfunction is a major complication after nonrenal solid organ transplants. Transplantation of vascularized composite allografts (VCA) has yielded successful midterm outcomes despite high rates of acute rejection and greater requirements of immunosuppression. Whether this translates in higher risks of kidney complications is unknown. METHODS Ninety-nine recipients of facial or extremity transplants from the Brigham and Women's Hospital (BWH) and the International Registry on Hand and Composite Tissue Transplantation (IR) were reviewed. We assessed immunosuppression, markers of renal function over time, as well as pretransplant and posttransplant renal risk factors. RESULTS Data were obtained from 10 patients from BWH (age at transplant, 42.5 ± 13.8 years) and 89 patients (37.8 ± 11.5 years) from IR. A significant rise in creatinine levels (BWH, P = 0.0195; IR, P < 0.0001) and drop in estimated glomerular filtration rate (GFR) within the first year posttransplant was observed. The BWH and IR patients lost a mean of 22 mL/min GFR and 60 mL/min estimated GFR in the first year, respectively. This decrease occurred mostly in the first 6 months posttransplant (BWH). Pretransplant creatinine levels were not restored in either cohort. A mixed linear model identified multiple variables correlating with renal dysfunction, particularly tacrolimus trough levels. CONCLUSIONS Kidney dysfunction represents a major complication posttransplantation in VCA recipients early on. Strategies to mitigate this complication, such as reducing calcineurin inhibitor trough levels or using alternative immunosuppressive agents, may improve long-term patient outcomes. Standardizing laboratory and data collection of kidney parameters and risk factors in VCA patients will be critical for better understanding of this complication.
Collapse
Affiliation(s)
- Nicco Krezdorn
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Sotirios Tasigiorgos
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Luccie Wo
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rachel Lopdrup
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marvee Turk
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Harriet Kiwanuka
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Salman Ahmed
- Transplantation Research Center, Renal Division, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| | - Palmina Petruzzo
- Department of Transplantation, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Department of Surgery, University of Cagliari, Cagliari, Italy
| | - Ericka Bueno
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Bohdan Pomahac
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Leonardo V. Riella
- Transplantation Research Center, Renal Division, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
80
|
Srivastava A, Palsson R, Kaze AD, Chen ME, Palacios P, Sabbisetti V, Betensky RA, Steinman TI, Thadhani RI, McMahon GM, Stillman IE, Rennke HG, Waikar SS. The Prognostic Value of Histopathologic Lesions in Native Kidney Biopsy Specimens: Results from the Boston Kidney Biopsy Cohort Study. J Am Soc Nephrol 2018; 29:2213-2224. [PMID: 29866798 DOI: 10.1681/asn.2017121260] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/18/2018] [Indexed: 11/03/2022] Open
Abstract
Background Few studies have evaluated whether histopathologic lesions on kidney biopsy provide prognostic information beyond clinical and laboratory data.Methods We enrolled 676 individuals undergoing native kidney biopsy at three tertiary care hospitals into a prospective, observational cohort study. Biopsy specimens were adjudicated for semiquantitative scores in 13 categories of histopathology by two experienced renal pathologists. Proportional hazards models tested the association between histopathologic lesions and risk of kidney disease progression (≥40% eGFR decline or RRT).Results Mean baseline eGFR was 57.5±36.0 ml/min per 1.73 m2 During follow-up (median, 34.3 months), 199 individuals suffered kidney disease progression. After adjustment for demographics, clinicopathologic diagnosis, and laboratory values, the following lesions (hazard ratio; 95% confidence interval) were independently associated with progression: inflammation in nonfibrosed interstitium (0.52; 0.32 to 0.83), moderate and severe versus minimal interstitial fibrosis/tubular atrophy (2.14; 1.24 to 3.69 and 3.42; 1.99 to 5.87, respectively), moderate and severe versus minimal global glomerulosclerosis (2.17; 1.36 to 3.45 and 3.31; 2.04 to 5.38, respectively), moderate and severe versus minimal arterial sclerosis (1.78; 1.15 to 2.74 and 1.64; 1.04 to 2.60, respectively), and moderate and severe versus minimal arteriolar sclerosis (1.63; 1.08 to 2.46 and 2.33; 1.42 to 3.83, respectively). An 11-point chronicity score derived from semiquantitative assessments of chronic lesions independently associated with higher risk of kidney disease progression (hazard ratio per one-point increase, 1.19; 95% confidence interval, 1.12 to 1.27).Conclusions Across a diverse group of kidney diseases, histopathologic lesions on kidney biopsy provide prognostic information, even after adjustment for proteinuria and eGFR.
Collapse
Affiliation(s)
- Anand Srivastava
- Renal Division and.,Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | | | | | - Rebecca A Betensky
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Ravi I Thadhani
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts; and.,Departments of Biomedical Sciences and.,Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | | | - Isaac E Stillman
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Helmut G Rennke
- Pathology Department, Brigham and Women's Hospital, Boston, Massachusetts
| | | |
Collapse
|
81
|
Gaston RS, Fieberg A, Hunsicker L, Kasiske BL, Leduc R, Cosio FG, Gourishankar S, Grande J, Mannon RB, Rush D, Cecka JM, Connett J, Matas AJ. Late graft failure after kidney transplantation as the consequence of late versus early events. Am J Transplant 2018; 18:1158-1167. [PMID: 29139625 DOI: 10.1111/ajt.14590] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/10/2017] [Accepted: 11/05/2017] [Indexed: 01/25/2023]
Abstract
Beyond the first posttransplant year, 3% of kidney transplants fail annually. In a prospective, multicenter cohort study, we tested the relative impact of early versus late events on risk of long-term death-censored graft failure (DCGF). In grafts surviving at least 90 days, early events (acute rejection [AR] and delayed graft function [DGF] before day 90) were recorded; serum creatinine (Cr) at day 90 was defined as baseline. Thereafter, a 25% rise in serum Cr or new-onset proteinuria triggered graft biopsy (index biopsy, IBx), allowing comparison of risk of DCGF associated with early events (AR, DGF, baseline serum Cr >2.0 mg/dL) to that associated with later events (IBx). Among 3678 patients followed for 4.7 ± 1.9 years, 753 (20%) had IBx at a median of 15.3 months posttransplant. Early AR (HR = 1.77, P < .001) and elevated Cr at Day 90 (HR = 2.56, P < .0001) were associated with increased risk of DCGF; however, later-onset dysfunction requiring IBx had far greater impact (HR = 13.8, P < .0001). At 90 days, neither clinical characteristics nor early events distinguished those who subsequently did or did not undergo IBx or suffer DCGF. To improve long-term kidney allograft survival, management paradigms should promote prompt diagnosis and treatment of both early and later events.
Collapse
Affiliation(s)
| | - Ann Fieberg
- University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | | | | | | - David Rush
- University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | |
Collapse
|
82
|
Abstract
BACKGROUND Calcineurin inhibitors (CNIs) are commonly given to transplant recipients of kidneys and other solid organs and to patients with immune disorders, such as steroid-resistant nephrotic syndrome, steroid-dependent nephrotic syndrome, and frequent relapse nephrotic syndrome. Although CNIs remain the most effective available immunosuppressant agent, there is clinical concern regarding possible long-term nephrotoxicity. This concern is especially significant in children who have a longer life expectancy and greater growth rate. DATA SOURCES In this review, we analyzed the literatures to identify original articles that examined use of CNIs in children who received organ transplantation and nephropathy to assess the available evidence of their nephrotoxicity. PubMed, Elsevier, and Tompson ISI Web of Knowledge were searched for identifying relevant papers. RESULTS Clinical research supports the presence of CNI-related nephrotoxicity. However, some researchers have questioned the prevalence and seriousness of chronic CNIs nephrotoxicity, especially because the pathological lesions typically associated with long-term CNI use are nonspecific. Many researchers have focused on early markers of CNI nephrotoxicity, and the methods that may help prevent and manage nephrotoxicity. CONCLUSIONS Future research should focus on investigating early markers of CNI nephrotoxicity and strategies for improved immunosuppressant therapy, and developing alternative treatments. CNI-mediated nephrotoxicity should always be taken seriously in clinic.
Collapse
Affiliation(s)
- Fei Liu
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian-Hua Mao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
83
|
Affiliation(s)
- Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
84
|
Nankivell BJ, Shingde M, Keung KL, Fung CLS, Borrows RJ, O'Connell PJ, Chapman JR. The causes, significance and consequences of inflammatory fibrosis in kidney transplantation: The Banff i-IFTA lesion. Am J Transplant 2018; 18:364-376. [PMID: 29194971 DOI: 10.1111/ajt.14609] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/25/2017] [Accepted: 10/28/2017] [Indexed: 01/25/2023]
Abstract
Inflammation within areas of interstitial fibrosis and tubular atrophy (i-IFTA) is associated with adverse outcomes in kidney transplantation. We evaluated i-IFTA in 429 indication- and 2052 protocol-driven biopsy samples from a longitudinal cohort of 362 kidney-pancreas recipients to determine its prevalence, time course, and relationships with T cell-mediated rejection (TCMR), immunosuppression, and outcome. Sequential histology demonstrated that i-IFTA was preceded by cellular interstitial inflammation and followed by IF/TA. The prevalence and intensity of i-IFTA increased with developing chronic fibrosis and correlated with inflammation, tubulitis, and immunosuppression era (P < .001). Tacrolimus era-based immunosuppression was associated with reduced histologic inflammation in unscarred and scarred i-IFTA compartments, ameliorated progression of IF, and increased conversion to inactive IF/TA (compared with cyclosporine era, P < .001). Prior acute (including borderline) TCMR and subclinical TCMR were followed by greater 1-year i-IFTA, remaining predictive by multivariate analysis and independent of humoral markers. One-year i-IFTA was associated with accelerated IF/TA, arterial fibrointimal hyperplasia, and chronic glomerulopathy and with reduced renal function (P < .001 versus no i-IFTA). In summary, i-IFTA is the histologic consequence of active T cell-mediated alloimmunity, representing the interface between inflammation and tubular injury with fibrotic healing. Uncontrolled i-IFTA is associated with adverse structural and functional outcomes.
Collapse
Affiliation(s)
| | - Meena Shingde
- Tissue Pathology and Diagnostic Oncology, Westmead Hospital, Sydney, Australia
| | - Karen L Keung
- Department of Renal Medicine, Westmead Hospital, Sydney, Australia
| | - Caroline L-S Fung
- Tissue Pathology and Diagnostic Oncology, Westmead Hospital, Sydney, Australia
| | | | | | - Jeremy R Chapman
- Department of Renal Medicine, Westmead Hospital, Sydney, Australia
| |
Collapse
|
85
|
Haas M, Loupy A, Lefaucheur C, Roufosse C, Glotz D, Seron D, Nankivell BJ, Halloran PF, Colvin RB, Akalin E, Alachkar N, Bagnasco S, Bouatou Y, Becker JU, Cornell LD, van Huyen JPD, Gibson IW, Kraus ES, Mannon RB, Naesens M, Nickeleit V, Nickerson P, Segev DL, Singh HK, Stegall M, Randhawa P, Racusen L, Solez K, Mengel M. The Banff 2017 Kidney Meeting Report: Revised diagnostic criteria for chronic active T cell-mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials. Am J Transplant 2018; 18:293-307. [PMID: 29243394 PMCID: PMC5817248 DOI: 10.1111/ajt.14625] [Citation(s) in RCA: 776] [Impact Index Per Article: 110.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 01/25/2023]
Abstract
The kidney sessions of the 2017 Banff Conference focused on 2 areas: clinical implications of inflammation in areas of interstitial fibrosis and tubular atrophy (i-IFTA) and its relationship to T cell-mediated rejection (TCMR), and the continued evolution of molecular diagnostics, particularly in the diagnosis of antibody-mediated rejection (ABMR). In confirmation of previous studies, it was independently demonstrated by 2 groups that i-IFTA is associated with reduced graft survival. Furthermore, these groups presented that i-IFTA, particularly when involving >25% of sclerotic cortex in association with tubulitis, is often a sequela of acute TCMR in association with underimmunosuppression. The classification was thus revised to include moderate i-IFTA plus moderate or severe tubulitis as diagnostic of chronic active TCMR. Other studies demonstrated that certain molecular classifiers improve diagnosis of ABMR beyond what is possible with histology, C4d, and detection of donor-specific antibodies (DSAs) and that both C4d and validated molecular assays can serve as potential alternatives and/or complements to DSAs in the diagnosis of ABMR. The Banff ABMR criteria are thus updated to include these alternatives. Finally, the present report paves the way for the Banff scheme to be part of an integrative approach for defining surrogate endpoints in next-generation clinical trials.
Collapse
Affiliation(s)
- M. Haas
- Department of Pathology and Laboratory MedicineCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - A. Loupy
- Paris Translational Research Center for Organ TransplantationINSERM U970 and Necker HospitalUniversity Paris DescartesParisFrance
| | - C. Lefaucheur
- Paris Translational Research Center for Organ Transplantation and Department of Nephrology and TransplantationHopital Saint LouisUniversité Paris VII and INSERM U 1160ParisFrance
| | - C. Roufosse
- Department of MedicineImperial College London and North West London PathologyLondonUK
| | - D. Glotz
- Paris Translational Research Center for Organ Transplantation and Department of Nephrology and TransplantationHopital Saint LouisUniversité Paris VII and INSERM U 1160ParisFrance
| | - D. Seron
- Nephrology DepartmentHospital Vall d'HebronAutonomous University of BarcelonaBarcelonaSpain
| | - B. J. Nankivell
- Department of Renal MedicineWestmead HospitalSydneyAustralia
| | - P. F. Halloran
- Alberta Transplant Applied Genomics CentreUniversity of AlbertaEdmontonAlbertaCanada
| | - R. B. Colvin
- Department of PathologyMassachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Enver Akalin
- Montefiore‐Einstein Center for TransplantationMontefiore Medical CenterBronxNYUSA
| | - N. Alachkar
- Department of MedicineSection of NephrologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - S. Bagnasco
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Y. Bouatou
- Paris Translational Research Center for Organ TransplantationINSERM U970 and Necker HospitalUniversity Paris DescartesParisFrance,Division of NephrologyDepartment of Medical SpecialitiesGeneva University HospitalsGenevaSwitzerland
| | - J. U. Becker
- Institute of PathologyUniversity Hospital of CologneCologneGermany
| | - L. D. Cornell
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - J. P. Duong van Huyen
- Paris Translational Research Center for Organ TransplantationINSERM U970 and Necker HospitalUniversity Paris DescartesParisFrance
| | - I. W. Gibson
- Department of PathologyUniversity of ManitobaWinnipegCanada
| | - Edward S. Kraus
- Division of NephrologyDepartment of MedicineJohns Hopkins UniversityBaltimoreMDUSA
| | - R. B. Mannon
- Division of NephrologyDepartment of MedicineUniversity of Alabama School of MedicineBirminghamALUSA
| | - M. Naesens
- Department of Microbiology and ImmunologyUniversity of Leuven & Department of NephrologyUniversity Hospitals LeuvenLeuvenBelgium
| | - V. Nickeleit
- Division of NephropathologyDepartment of Pathology and Laboratory MedicineThe University of North Carolina School of MedicineChapel HillNCUSA
| | - P. Nickerson
- Department of Internal Medicine and ImmunologyUniversity of ManitobaWinnipegCanada
| | - D. L. Segev
- Department of SurgeryJohns Hopkins Medical InstitutionsBaltimoreMDUSA
| | - H. K. Singh
- Division of NephropathologyDepartment of Pathology and Laboratory MedicineThe University of North Carolina School of MedicineChapel HillNCUSA
| | - M. Stegall
- Departments of Surgery and ImmunologyMayo ClinicRochesterMNUSA
| | - P. Randhawa
- Division of Transplantation PathologyThomas E. Starzl Transplantation InstituteUniversity of PittsburghPittsburghPAUSA
| | - L. Racusen
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - K. Solez
- Department of Laboratory Medicine and PathologyUniversity of AlbertaEdmontonCanada
| | - M. Mengel
- Department of Laboratory Medicine and PathologyUniversity of AlbertaEdmontonCanada
| |
Collapse
|
86
|
Lefaucheur C, Gosset C, Rabant M, Viglietti D, Verine J, Aubert O, Louis K, Glotz D, Legendre C, Duong Van Huyen JP, Loupy A. T cell-mediated rejection is a major determinant of inflammation in scarred areas in kidney allografts. Am J Transplant 2018; 18:377-390. [PMID: 29086461 DOI: 10.1111/ajt.14565] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/22/2017] [Accepted: 10/22/2017] [Indexed: 01/25/2023]
Abstract
Inflammation in fibrosis areas (i-IF/TA) of kidney allografts is associated with allograft loss; however, its diagnostic significance remains to be determined. We investigated the clinicohistologic phenotype and determinants of i-IF/TA in a prospective cohort of 1539 kidney recipients undergoing evaluation of i-IF/TA and tubulitis in atrophic tubules (t-IF/TA) on protocol allograft biopsies performed at 1 year posttransplantation. We considered donor, recipient, and transplant characteristics, immunosuppression, and histological diagnoses in 2260 indication biopsies performed within the first year posttransplantation. Nine hundred forty-six (61.5%) patients presented interstitial fibrosis/tubular atrophy (IF/TA Banff grade > 0) at 1 year posttransplant, among whom 394 (41.6%) showed i-IF/TA. i-IF/TA correlated with concurrent t-IF/TA (P < .001), interstitial inflammation (P < .001), tubulitis (P < .001), total inflammation (P < .001), peritubular capillaritis (P < .001), interstitial fibrosis (P < .001), and tubular atrophy (P = .02). The independent determinants of i-IF/TA were previous T cell-mediated rejection (TCMR) (P < .001), BK virus nephropathy (P = .007), steroid therapy (P = .039), calcineurin inhibitor therapy (P = .011), inosine-5'-monophosphate dehydrogenase inhibitor therapy (P = .011), HLA-B mismatches (P = .012), and HLA-DR mismatches (P = .044). TCMR patients with i-IF/TA on posttreatment biopsy (N = 83/136, 61.0%) exhibited accelerated progression of IF/TA over time (P = .01) and decreased 8-year allograft survival (70.8% vs 83.5%, P = .038) compared to those without posttreatment i-IF/TA. Our results support that i-IF/TA may represent a manifestation of chronic active TCMR.
Collapse
Affiliation(s)
- Carmen Lefaucheur
- Department of Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.,Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Clément Gosset
- Department of Pathology, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Marion Rabant
- Department of Pathology, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Denis Viglietti
- Department of Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.,Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Jérôme Verine
- Department of Pathology, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Olivier Aubert
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Kevin Louis
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Denis Glotz
- Department of Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.,Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Christophe Legendre
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Department of Kidney Transplantation, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-Paul Duong Van Huyen
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Department of Pathology, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Department of Kidney Transplantation, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
87
|
Seeger H, Lindenmeyer MT, Cohen CD, Jaeckel C, Nelson PJ, Chen J, Edenhofer I, Kozakowski N, Regele H, Boehmig G, Brandt S, Wuethrich RP, Heikenwalder M, Fehr T, Segerer S. Lymphotoxin expression in human and murine renal allografts. PLoS One 2018; 13:e0189396. [PMID: 29300739 PMCID: PMC5754061 DOI: 10.1371/journal.pone.0189396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/26/2017] [Indexed: 01/23/2023] Open
Abstract
The kidney is the most frequently transplanted solid organ. Recruitment of inflammatory cells, ranging from diffuse to nodular accumulations with defined microarchitecture, is a hallmark of acute and chronic renal allograft injury. Lymphotoxins (LTs) mediate the communication of lymphocytes and stromal cells and play a pivotal role in chronic inflammation and formation of lymphoid tissue. The aim of this study was to assess the expression of members of the LT system in acute rejection (AR) and chronic renal allograft injury such as transplant glomerulopathy (TG) and interstitial fibrosis/tubular atrophy (IFTA). We investigated differentially regulated components in transcriptomes of human renal allograft biopsies. By microarray analysis, we found the upregulation of LTβ, LIGHT, HVEM and TNF receptors 1 and 2 in AR and IFTA in human renal allograft biopsies. In addition, there was clear evidence for the activation of the NFκB pathway, most likely a consequence of LTβ receptor stimulation. In human renal allograft biopsies with transplant glomerulopathy (TG) two distinct transcriptional patterns of LT activation were revealed. By quantitative RT-PCR robust upregulation of LTα, LTβ and LIGHT was shown in biopsies with borderline lesions and AR. Immunohistochemistry revealed expression of LTβ in tubular epithelial cells and inflammatory infiltrates in transplant biopsies with AR and IFTA. Finally, activation of LT signaling was reproduced in a murine model of renal transplantation with AR. In summary, our results indicate a potential role of the LT system in acute renal allograft rejection and chronic transplant injury. Activation of the LT system in allograft rejection in rodents indicates a species independent mechanism. The functional role of the LT system in acute renal allograft rejection and chronic injury remains to be determined.
Collapse
Affiliation(s)
- Harald Seeger
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
- * E-mail:
| | - Maja T. Lindenmeyer
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Clemens D. Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Carsten Jaeckel
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Peter J. Nelson
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Jin Chen
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
| | - Ilka Edenhofer
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
| | | | - Heinz Regele
- Clinical Institute of Pathology, University of Vienna, Vienna, Austria
| | - Georg Boehmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Austria
| | - Simone Brandt
- Institute of Surgical Pathology, University Hospital Zuerich, Zurich, Switzerland
| | - Rudolf P. Wuethrich
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Fehr
- Department of Internal Medicine, Kantonsspital Graubuenden, Chur, Switzerland
| | - Stephan Segerer
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
- Division of Nephrology, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
88
|
Kühne L, Jung B, Poth H, Schuster A, Wurm S, Ruemmele P, Banas B, Bergler T. Renal allograft rejection, lymphocyte infiltration, and de novo donor-specific antibodies in a novel model of non-adherence to immunosuppressive therapy. BMC Immunol 2017; 18:52. [PMID: 29258420 PMCID: PMC5735914 DOI: 10.1186/s12865-017-0236-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 12/07/2017] [Indexed: 12/18/2022] Open
Abstract
Background Non-adherence has been associated with reduced graft survival. The aim of this study was to investigate the immunological mechanisms underlying chronic renal allograft rejection using a model of non-adherence to immunosuppressive therapy. We used a MHC (major histocompatibility complex) -mismatched rat model of renal transplantation (Brown Norway to Lewis), in which rats received daily oral cyclosporine A. In analogy to non-adherence to therapy, one group received cyclosporine A on alternating days only. Rejection was histologically graded according to the Banff classification. We quantified fibrosis by trichrome staining and intra-graft infiltration of T cells, B cells, and monocytes/macrophages by immunohistochemistry. The distribution of B lymphocytes was assessed using immunofluorescence microscopy. Intra-graft chemokine, chemokine receptor, BAFF (B cell activating factor belonging to the TNF family), and immunoglobulin G transcription levels were analysed by RT-PCR. Finally, we evaluated donor-specific antibodies (DSA) and complement-dependent cytotoxicity using flow cytometry. Results After 28 days, cellular rejection occurred during non-adherence in 5/6 animals, mixed with humoral rejection in 3/6 animals. After non-adherence, the number of T lymphocytes were elevated compared to daily immunosuppression. Monocyte numbers declined over time. Accordingly, lymphocyte chemokine transcription was significantly increased in the graft, as was the transcription of BAFF, BAFF receptor, and Immunoglobulin G. Donor specific antibodies were elevated in non-adherence, but did not induce complement-dependent cytotoxicity. Conclusion Cellular and humoral rejection, lymphocyte infiltration, and de novo DSA are induced in this model of non-adherence. Electronic supplementary material The online version of this article (doi: 10.1186/s12865-017-0236-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Louisa Kühne
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, D-93053, Regensburg, Germany.
| | - Bettina Jung
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, D-93053, Regensburg, Germany
| | - Helen Poth
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, D-93053, Regensburg, Germany
| | - Antonia Schuster
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, D-93053, Regensburg, Germany
| | - Simone Wurm
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, D-93053, Regensburg, Germany
| | - Petra Ruemmele
- Department of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, D-93053, Regensburg, Germany
| | - Tobias Bergler
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, D-93053, Regensburg, Germany
| |
Collapse
|
89
|
Development of CD3 cell quantitation algorithms for renal allograft biopsy rejection assessment utilizing open source image analysis software. Virchows Arch 2017; 472:259-269. [DOI: 10.1007/s00428-017-2260-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/29/2017] [Accepted: 10/24/2017] [Indexed: 12/18/2022]
|
90
|
Seija M, Nin M, Astesiano R, Coitiño R, Santiago J, Ferrari S, Noboa O, González-Martinez F. Rechazo agudo del trasplante renal: diagnóstico y alternativas terapéuticas. NEFROLOGÍA LATINOAMERICANA 2017. [DOI: 10.1016/j.nefrol.2017.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
91
|
Van Loon E, Lerut E, Naesens M. The time dependency of renal allograft histology. Transpl Int 2017; 30:1081-1091. [DOI: 10.1111/tri.13042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/05/2017] [Accepted: 08/21/2017] [Indexed: 01/27/2023]
Affiliation(s)
- Elisabet Van Loon
- Laboratory of Nephrology; Department of Microbiology and Immunology; KU Leuven; Leuven Belgium
- Department of Nephrology and Renal Transplantation; University Hospitals Leuven; Leuven Belgium
| | - Evelyne Lerut
- Translational Cell and Tissue Research; Department of Imaging and Pathology; KU Leuven; Leuven Belgium
- Department of Morphology and Molecular Pathology; University Hospitals Leuven; Leuven Belgium
| | - Maarten Naesens
- Laboratory of Nephrology; Department of Microbiology and Immunology; KU Leuven; Leuven Belgium
- Department of Nephrology and Renal Transplantation; University Hospitals Leuven; Leuven Belgium
| |
Collapse
|
92
|
Désy O, Béland S, Vallin P, Riopel J, Latulippe E, Najafian N, Chandraker A, Agharazii M, Batal I, De Serres SA. IL-6 production by monocytes is associated with graft function decline in patients with borderline changes suspicious for acute T-cell-mediated rejection: a pilot study. Transpl Int 2017; 31:92-101. [DOI: 10.1111/tri.13070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/28/2017] [Accepted: 09/06/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Olivier Désy
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Stéphanie Béland
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Patrice Vallin
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Julie Riopel
- Department of Pathology; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Eva Latulippe
- Department of Pathology; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Nader Najafian
- Renal Division; Schuster Family Transplantation Research Center; Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School; Boston MA USA
| | - Anil Chandraker
- Renal Division; Schuster Family Transplantation Research Center; Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School; Boston MA USA
| | - Mohsen Agharazii
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Ibrahim Batal
- Department of Pathology; Brigham and Women's Hospital and Harvard Medical School; Boston MA USA
| | - Sacha A. De Serres
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| |
Collapse
|
93
|
Torres IB, Reisaeter AV, Moreso F, Âsberg A, Vidal M, Garcia-Carro C, Midtvedt K, Reinholt FP, Scott H, Castellà E, Salcedo M, Dörje C, Sellarés J, Azancot MA, Perello M, Holdaas H, Serón D. Tacrolimus and mycophenolate regimen and subclinical tubulo-interstitial inflammation in low immunological risk renal transplants. Transpl Int 2017; 30:1119-1131. [PMID: 28667664 DOI: 10.1111/tri.13002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/08/2017] [Accepted: 06/23/2017] [Indexed: 10/19/2022]
Abstract
The aim was to evaluate the relationship between maintenance immunosuppression, subclinical tubulo-interstitial inflammation and interstitial fibrosis/tubular atrophy (IF/TA) in surveillance biopsies performed in low immunological risk renal transplants at two transplant centers. The Barcelona cohort consisted of 109 early and 66 late biopsies in patients receiving high tacrolimus (TAC-C0 target at 1-year 6-10 ng/ml) and reduced MMF dose (500 mg bid at 1-year). The Oslo cohort consisted of 262 early and 237 late biopsies performed in patients treated with low TAC-C0 (target 3-7 ng/ml) and standard MMF dose (750 mg bid). Subclinical inflammation, adjusted for confounders, was associated with low TAC-C0 in the early (OR: 0.75, 95% CI: 0.61-0.92; P = 0.006) and late biopsies (OR: 0.69, 95% CI: 0.50-0.95; P = 0.023) from Barcelona. In the Oslo cohort, it was associated with low MMF in early biopsies (OR: 0.90, 95% CI: 0.83-0.98; P = 0.0101) and with low TAC-C0 in late biopsies (OR: 0.77, 95% CI: 0.61-0.97; P = 0.0286). MMF dose was significantly reduced in Oslo between early and late biopsies. IF/TA was not associated with TAC-C0 or MMF dose in the multivariate analysis. Our data suggest that in TAC- and MMF-based regimens, TAC-C0 levels are associated with subclinical inflammation in patients receiving reduced MMF dose.
Collapse
Affiliation(s)
- Irina B Torres
- Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna V Reisaeter
- Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Francesc Moreso
- Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anders Âsberg
- Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,School of Pharmacy, University of Oslo, Norway
| | - Marta Vidal
- Department of Pathology, Hospital Universitari Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Clara Garcia-Carro
- Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Karsten Midtvedt
- Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Finn P Reinholt
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Helge Scott
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Eva Castellà
- Department of Radiology, Hospital Universitari Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Maite Salcedo
- Department of Pathology, Hospital Universitari Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Christina Dörje
- Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Joana Sellarés
- Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria A Azancot
- Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manel Perello
- Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Hallvard Holdaas
- Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Daniel Serón
- Department of Nephrology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
94
|
Pihlstrøm HK, Gatti F, Hammarström C, Eide IA, Kasprzycka M, Wang J, Haraldsen G, Svensson MHS, Midtvedt K, Mjøen G, Dahle DO, Hartmann A, Holdaas H. Early introduction of oral paricalcitol in renal transplant recipients. An open-label randomized study. Transpl Int 2017; 30:827-840. [PMID: 28436117 DOI: 10.1111/tri.12973] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/31/2016] [Accepted: 04/18/2017] [Indexed: 02/01/2023]
Abstract
In stable renal transplant recipients with hyperparathyroidism, previous studies have indicated that vitamin D agonist treatment might have anti-proteinuric effects. Animal studies indicate possible anti-fibrotic and anti-inflammatory effects. Early introduction of paricalcitol in de novo renal transplant recipients might reduce proteinuria and prevent progressive allograft fibrosis. We performed a single-center, prospective, randomized, open-label trial investigating effects of paricalcitol 2 μg/day added to standard care. Participants were included 8 weeks after engraftment and followed for 44 weeks. Primary end point was change in spot urine albumin/creatinine ratio. Exploratory microarray analyses of kidney biopsies at study end investigated potential effects on gene expression. Secondary end points included change in glomerular filtration rate (GFR), pulse wave velocity (PWV), and endothelial function measured by peripheral arterial tonometry as reactive hyperemia index (RHI). Seventy-seven de novo transplanted kidney allograft recipients were included, 37 receiving paricalcitol. Paricalcitol treatment lowered PTH levels (P = 0.01) but did not significantly reduce albuminuria (P = 0.76), change vascular parameters (PWV; P = 0.98, RHI; P = 0.33), or influence GFR (P = 0.57). Allograft gene expression was not influenced. To summarize, in newly transplanted renal allograft recipients, paricalcitol reduced PTH and was well tolerated without negatively affecting kidney function. Paricalcitol did not significantly reduce/prevent albuminuria, improve parameters of vascular health, or influence allograft gene expression.
Collapse
Affiliation(s)
- Hege Kampen Pihlstrøm
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Franscesca Gatti
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Clara Hammarström
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Ivar Anders Eide
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Monika Kasprzycka
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Junbai Wang
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Guttorm Haraldsen
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | | | - Karsten Midtvedt
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Geir Mjøen
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Dag Olav Dahle
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Anders Hartmann
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hallvard Holdaas
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
95
|
Rejection of the Renal Allograft in the Absence of Demonstrable Antibody and Complement. Transplantation 2017; 101:395-401. [PMID: 26901079 DOI: 10.1097/tp.0000000000001118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Recent literature has stressed the prominent role of antibodies in graft loss. This study was designed to assess a growing perception that T cell-mediated rejection (TCMR) is no longer clinically relevant. METHODS Five hundred forty-five renal allograft recipients over a 3-year period were screened for biopsies with: (a) TCMR including borderline change (BL), (b) negative complement protein C4 degradation fragment, and (c) absence of donor-specific antibody at time of transplant, within 30 days of the biopsy, and up to 4 measurements at later time points. RESULTS These stringent requirements identified 28 "pure" cases of late TCMR/BL. Low-grade glomerulitis, peritubular capillaritis, or chronic transplant glomerulopathy were found in 9/28 (32%) biopsies. Serum creatinine showed complete short-term remission in 7/10 (70%) BL and 9/18 (50%) TCMR patients 1 month postbiopsy. Yet, both treated and untreated patients demonstrated further decline in graft function as assessed by serum creatinine and estimated glomerular filtration rate. CONCLUSIONS Late TCMR seen in 7.9% of biopsies can contribute to significant deterioration of graft function in patients in whom the dominant contribution of antibody-mediated injury has been reasonably excluded. Our data also reinforce existing literature showing that microvascular lesions do not have absolute specificity for a diagnosis of antibody-mediated rejection.
Collapse
|
96
|
Abstract
BACKGROUND Early prognostic markers that identify high-risk patients could lead to increased surveillance, personalized immunosuppression, and improved long-term outcomes. The goal of this study was to validate 6-month urinary chemokine ligand 2 (CCL2) as a noninvasive predictor of long-term outcomes and compare it with 6-month urinary CXCL10. METHODS A prospective, observational renal transplant cohort (n = 185; minimum, 5-year follow-up) was evaluated. The primary composite outcome included 1 or more: allograft loss, renal function decline (>20% decrease estimated glomerular filtration rate between 6 months and last follow-up), and biopsy-proven rejection after 6 months. CCL2/CXCL10 are reported in relation to urine creatinine (ng/mmol). RESULTS Fifty-two patients (52/185, 28%) reached the primary outcome at a median 6.0 years, and their urinary CCL2:Cr was significantly higher compared with patients with stable allograft function (median [interquartile range], 38.6 ng/mmol [19.7-72.5] vs 25.9 ng/mmol [16.1-45.8], P = 0.009). Low urinary CCL2:Cr (≤70.0 ng/mmol) was associated with 88% 5-year event-free survival compared with 50% with high urinary CCL2:Cr (P < 0.0001). In a multivariate Cox-regression model, the only independent predictors of the primary outcome were high CCL2:Cr (hazard ratio [HR], 2.86; 95% confidence interval [95% CI], 1.33-5.73) and CXCL10:Cr (HR, 2.35; 95% CI, 1.23-4.88; both P = 0.009). Urinary CCL2:Cr/CXCL10:Cr area under the curves were 0.62 (P = 0.001)/0.63 (P = 0.03), respectively. Time-to-endpoint analysis according to combined high or low urinary chemokines demonstrates that endpoint-free survival depends on the overall early chemokine burden. CONCLUSIONS This study confirms that urinary CCL2:Cr is an independent predictor of long-term allograft outcomes. Urinary CCL2:Cr/CXCL10:Cr alone have similar prognostic performance, but when both are elevated, this suggests a worse prognosis. Therefore, urinary chemokines may be a useful tool for timely identification of high-risk patients.
Collapse
|
97
|
Zununi Vahed S, Poursadegh Zonouzi A, Ghanbarian H, Ghojazadeh M, Samadi N, Omidi Y, Ardalan M. Differential expression of circulating miR-21, miR-142-3p and miR-155 in renal transplant recipients with impaired graft function. Int Urol Nephrol 2017; 49:1681-1689. [PMID: 28455659 DOI: 10.1007/s11255-017-1602-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/17/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND The discovery of circulating microRNAs (miRNAs), as potential noninvasive diagnostic biomarkers, has opened new avenues of research for identifying patients with chronic failure in renal transplantation. The present study aimed to investigate the expression levels of four immune-related miRNAs (miR-21, miR-31, miR-142-3p and miR-155) in plasma samples of renal recipients. METHODS The plasma expression levels of the miRNAs were evaluated by quantitative real-time PCR (qPCR) in 53 renal recipients with long-term stable allograft function, SGF (N = 27), and with biopsy-proven interstitial fibrosis and tubular atrophy (IFTA) (N = 26) and also healthy controls (N = 15). The possible correlation between clinical parameters and the circulating miRNAs and the receiver-operating characteristic (ROC) analysis were performed. RESULTS Our results showed that expression of miR-21 (p = 0.023), miR-142-3p (p = 0.048) and miR-155 (p = 0.005) was significantly upregulated in plasma samples of recipients with IFTA in comparison with SGF and healthy control groups. Concentration of miR-21 (∆Ct value) in plasma was negatively correlated with creatinine (r = -0.432, p = 0.028) and positively correlated with eGFR (r = 0.423, p = 0.031). The multivariate ROC curve analysis indicated that miR-21, miR-142-3p and miR-155 in plasma samples could discriminate almost most of the IFTA patients (area under curve = 0.802, sensitivity = 81%, specificity = 92%). CONCLUSION Our data suggested that altered expression of miR-21, miR-142-3p and miR-155 in plasma samples may be associated with renal dysfunction and can be used for graft monitoring.
Collapse
Affiliation(s)
- Sepideh Zununi Vahed
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Ahmad Poursadegh Zonouzi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Hossein Ghanbarian
- School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Morteza Ghojazadeh
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Nasser Samadi
- School of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Islamic Republic of Iran
| | - Mohammadreza Ardalan
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.
| |
Collapse
|
98
|
Vanhove T, Goldschmeding R, Kuypers D. Kidney Fibrosis: Origins and Interventions. Transplantation 2017; 101:713-726. [PMID: 27941433 PMCID: PMC7228593 DOI: 10.1097/tp.0000000000001608] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/24/2016] [Accepted: 11/10/2016] [Indexed: 02/06/2023]
Abstract
All causes of renal allograft injury, when severe and/or sustained, can result in chronic histological damage of which interstitial fibrosis and tubular atrophy are dominant features. Unless a specific disease process can be identified, what drives interstitial fibrosis and tubular atrophy progression in individual patients is often unclear. In general, clinicopathological factors known to predict and drive allograft fibrosis include graft quality, inflammation (whether "nonspecific" or related to a specific diagnosis), infections, such as polyomavirus-associated nephropathy, calcineurin inhibitors (CNI), and genetic factors. The incidence and severity of chronic histological damage have decreased substantially over the last 3 decades, but it is difficult to disentangle what effects individual innovations (eg, better matching and preservation techniques, lower CNI dosing, BK viremia screening) may have had. There is little evidence that CNI-sparing/minimization strategies, steroid minimization or renin-angiotensin-aldosterone system blockade result in better preservation of intermediate-term histology. Treatment of subclinical rejections has only proven beneficial to histological and functional outcome in studies in which the rate of subclinical rejection in the first 3 months was greater than 10% to 15%. Potential novel antifibrotic strategies include antagonists of transforming growth factor-β, connective tissue growth factor, several tyrosine kinase ligands (epidermal growth factor, platelet-derived growth factor, vascular endothelial growth factor), endothelin and inhibitors of chemotaxis. Although many of these drugs are mainly being developed and marketed for oncological indications and diseases, such as idiopathic pulmonary fibrosis, a number may hold promise in the treatment of diabetic nephropathy, which could eventually lead to applications in renal transplantation.
Collapse
Affiliation(s)
- Thomas Vanhove
- 1 Department of Microbiology and Immunology, KU Leuven-University of Leuven, Leuven, Belgium. 2 Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium. 3 Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | |
Collapse
|
99
|
Yılmaz R, Yıldırım T, Baydar D, Altun B, Erdem Y. Urinary Type III Procollagen Is Associated With Chronic Allograft Dysfunction and Predicts Graft Survival. Transplant Proc 2017; 49:281-287. [DOI: 10.1016/j.transproceed.2016.11.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/31/2016] [Accepted: 11/16/2016] [Indexed: 12/13/2022]
|
100
|
Modena BD, Milam R, Harrison F, Cheeseman JA, Abecassis MM, Friedewald J, Kirk AD, Salomon DR. Changes in Urinary Microbiome Populations Correlate in Kidney Transplants With Interstitial Fibrosis and Tubular Atrophy Documented in Early Surveillance Biopsies. Am J Transplant 2017; 17:712-723. [PMID: 27597148 PMCID: PMC5328852 DOI: 10.1111/ajt.14038] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/25/2023]
Abstract
An unbalanced microbiome may lead to disease by creating aberrant immune responses. A recent association of cellular rejection with the development of interstitial fibrosis and tubular atrophy (IFTA) suggests the role of immune-mediated tissue injury. We hypothesized that developing IFTA correlates with altered urinary tract microbiomes (UMBs). UMBs at two serial time points, 1 and 6-8 months posttransplant, were assessed by 16S microbial ribosomal gene sequencing in 25 patients developing biopsy-proven IFTA compared to 23 transplant patients with normal biopsies and excellent function (TX) and 20 healthy nontransplant controls (HC). Streptococcus, the dominant genera in HC males, was lower in IFTA and TX males at 1 month compared to HCs. At 6-8 months, Streptococcus was further decreased in IFTA males, but normalized in TX. IFTA males and females had increases in number of genera per sample at 6-8 months. UMB composition varied substantially between individuals in all groups. Despite the wide variation in UMBs between individuals, IFTA was associated with a loss in dominant resident urinary microbes in males, and a parallel increase in nonresident, pathogenic bacteria in males and females. UMB changes may contribute to IFTA development by alteration of the host immune response.
Collapse
Affiliation(s)
- Brian D Modena
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - Rachel Milam
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - Frank Harrison
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | | | - Michael M Abecassis
- Northwestern Comprehensive Transplant Center, Northwestern University, Chicago, IL
| | - John Friedewald
- Northwestern Comprehensive Transplant Center, Northwestern University, Chicago, IL
| | | | - Daniel R Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, Scripps Center for Organ Transplantation, Scripps Health, La Jolla, CA
| |
Collapse
|