101
|
Ekdahl KN, Persson B, Mohlin C, Sandholm K, Skattum L, Nilsson B. Interpretation of Serological Complement Biomarkers in Disease. Front Immunol 2018; 9:2237. [PMID: 30405598 PMCID: PMC6207586 DOI: 10.3389/fimmu.2018.02237] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/10/2018] [Indexed: 01/07/2023] Open
Abstract
Complement system aberrations have been identified as pathophysiological mechanisms in a number of diseases and pathological conditions either directly or indirectly. Examples of such conditions include infections, inflammation, autoimmune disease, as well as allogeneic and xenogenic transplantation. Both prospective and retrospective studies have demonstrated significant complement-related differences between patient groups and controls. However, due to the low degree of specificity and sensitivity of some of the assays used, it is not always possible to make predictions regarding the complement status of individual patients. Today, there are three main indications for determination of a patient's complement status: (1) complement deficiencies (acquired or inherited); (2) disorders with aberrant complement activation; and (3) C1 inhibitor deficiencies (acquired or inherited). An additional indication is to monitor patients on complement-regulating drugs, an indication which may be expected to increase in the near future since there is now a number of such drugs either under development, already in clinical trials or in clinical use. Available techniques to study complement include quantification of: (1) individual components; (2) activation products, (3) function, and (4) autoantibodies to complement proteins. In this review, we summarize the appropriate indications, techniques, and interpretations of basic serological complement analyses, exemplified by a number of clinical disorders.
Collapse
Affiliation(s)
- Kristina N Ekdahl
- Rudbeck Laboratory C5:3, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Centre of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Barbro Persson
- Rudbeck Laboratory C5:3, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Camilla Mohlin
- Centre of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Kerstin Sandholm
- Centre of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Lillemor Skattum
- Section of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Clinical Immunology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Bo Nilsson
- Rudbeck Laboratory C5:3, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
102
|
Halloran PF, Reeve J, Aliabadi AZ, Cadeiras M, Crespo-Leiro MG, Deng M, Depasquale EC, Goekler J, Jouven X, Kim DH, Kobashigawa J, Loupy A, Macdonald P, Potena L, Zuckermann A, Parkes MD. Exploring the cardiac response to injury in heart transplant biopsies. JCI Insight 2018; 3:123674. [PMID: 30333303 DOI: 10.1172/jci.insight.123674] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/11/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Because injury is universal in organ transplantation, heart transplant endomyocardial biopsies present an opportunity to explore response to injury in heart parenchyma. Histology has limited ability to assess injury, potentially confusing it with rejection, whereas molecular changes have potential to distinguish injury from rejection. Building on previous studies of transcripts associated with T cell-mediated rejection (TCMR) and antibody-mediated rejection (ABMR), we explored transcripts reflecting injury. METHODS Microarray data from 889 prospectively collected endomyocardial biopsies from 454 transplant recipients at 14 centers were subjected to unsupervised principal component analysis and archetypal analysis to detect variation not explained by rejection. The resulting principal component and archetype scores were then examined for their transcript, transcript set, and pathway associations and compared to the histology diagnoses and left ventricular function. RESULTS Rejection was reflected by principal components PC1 and PC2, and by archetype scores S2TCMR, and S3ABMR, with S1normal indicating normalness. PC3 and a new archetype score, S4injury, identified unexplained variation correlating with expression of transcripts inducible in injury models, many expressed in macrophages and associated with inflammation in pathway analysis. S4injury scores were high in recent transplants, reflecting donation-implantation injury, and both S4injury and S2TCMR were associated with reduced left ventricular ejection fraction. CONCLUSION Assessment of injury is necessary for accurate estimates of rejection and for understanding heart transplant phenotypes. Biopsies with molecular injury but no molecular rejection were often misdiagnosed rejection by histology.TRAIL REGISTRATION. ClinicalTrials.gov NCT02670408FUNDING. Roche Organ Transplant Research Foundation, the University of Alberta Hospital Foundation, and Alberta Health Services.
Collapse
Affiliation(s)
- Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Jeff Reeve
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Martin Cadeiras
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
| | | | - Mario Deng
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
| | | | | | | | - Daniel H Kim
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Peter Macdonald
- The Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Luciano Potena
- Cardiovascular Department, University of Bologna, Bologna, Italy
| | | | - Michael D Parkes
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
| |
Collapse
|
103
|
Advancing Transplantation: New Questions, New Possibilities in Kidney and Liver Transplantation. Transplantation 2018; 101 Suppl 2S:S1-S41. [PMID: 28125449 DOI: 10.1097/tp.0000000000001563] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
104
|
Lohéac C, Aubert O, Loupy A, Legendre C. [Identifying the specific causes of kidney allograft loss: A population-based study]. Nephrol Ther 2018; 14 Suppl 1:S39-S50. [PMID: 29606262 DOI: 10.1016/j.nephro.2018.02.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Results of kidney transplantation have been improving but long-term allograft survival remains disappointing. The objective of the present study was to identify the specific causes of renal allograft loss, to assess their incidence and long-term outcomes. METHODS A total of 4783 patients from four French centres, transplanted between January 2004 and January 2014 were prospectively included. A total of 9959 kidney biopsies (protocol and for cause) performed between January 2004 and March 2015 were included. Donor and recipient clinical and biological parameters as well as anti-HLA antibody directed against the donor were included. The main outcome was the long-term kidney allograft survival, including the study of the associated causes of graft loss, the delay of graft loss according to their causes and the determinants of graft loss. RESULTS There were 732 graft losses during the follow-up period (median time: 4.51 years) with an identified cause in 95.08 %. Kidney allograft survival at 9 years post-transplant was 78 %. The causes of allograft loss were: antibody-mediated rejection (31.69 %), thrombosis (25.55 %), medical intercurrent disease (14.62 %), recurrence of primary renal disease (7.1 %), BK- or CMV-associated nephropathy (n=35, 4.78 %), T cell-mediated rejection (4.78 %), urological disease (2.46 %) and calcineurin inhibitor nephrotoxicity (1.09 %). CONCLUSION The main causes of allograft loss were antibody-mediated rejection and thrombosis. These results encourage efforts to prevent and detect these complications earlier in order to improve allograft survival.
Collapse
Affiliation(s)
- Charlotte Lohéac
- Service de transplantation rénale, hôpital Necker-Enfants-malades, 149, rue de Sèvres, 75015 Paris, France; Université Paris Descartes, 75006 Paris, France
| | - Olivier Aubert
- Service de transplantation rénale, hôpital Necker-Enfants-malades, 149, rue de Sèvres, 75015 Paris, France; Université Paris Descartes, 75006 Paris, France
| | - Alexandre Loupy
- Service de transplantation rénale, hôpital Necker-Enfants-malades, 149, rue de Sèvres, 75015 Paris, France; Université Paris Descartes, 75006 Paris, France
| | - Christophe Legendre
- Service de transplantation rénale, hôpital Necker-Enfants-malades, 149, rue de Sèvres, 75015 Paris, France; Université Paris Descartes, 75006 Paris, France.
| |
Collapse
|
105
|
Barner M, DeKoning J, Kashi Z, Halloran P. Recent Advancements in the Assessment of Renal Transplant Dysfunction with an Emphasis on Microarray Molecular Diagnostics. Clin Lab Med 2018; 38:623-635. [PMID: 30420057 DOI: 10.1016/j.cll.2018.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Conventional assessment of renal transplant rejection and injury through use of histology, C4d staining, and HLA antibody testing, has been the standard approach to transplant management. By many measures, these methods of conventional assessment may be considered flawed, particularly with the subjective nature of histologic diagnoses. The Alberta Transplant Applied Genomics Center has developed the Molecular Microscope diagnostic system, which uses microarrays to measure gene expression. These data are analyzed using classifiers (weighted equations) that compare the tested biopsy to a proprietary reference set of biopsies to provide objective measures of the status of the renal transplant.
Collapse
Affiliation(s)
- Meagan Barner
- Kashi Clinical Laboratories, 10101 Southwest Barbur Boulevard Suite 200, Portland, OR 97219, USA
| | - Jenefer DeKoning
- Kashi Clinical Laboratories, 10101 Southwest Barbur Boulevard Suite 200, Portland, OR 97219, USA
| | - Zahra Kashi
- Kashi Clinical Laboratories, 10101 Southwest Barbur Boulevard Suite 200, Portland, OR 97219, USA.
| | - Phillip Halloran
- Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Alberta Transplant Applied Genomics Center, 250 Heritage Medical Research Centre, Edmonton, Alberta T6G 2S2, Canada; Transcriptome Sciences Inc, Edmonton, Alberta, Canada
| |
Collapse
|
106
|
van der Zwan M, Clahsen-Van Groningen MC, Roodnat JI, Bouvy AP, Slachmuylders CL, Weimar W, Baan CC, Hesselink DA, Kho MML. The Efficacy of Rabbit Anti-Thymocyte Globulin for Acute Kidney Transplant Rejection in Patients Using Calcineurin Inhibitor and Mycophenolate Mofetil-Based Immunosuppressive Therapy. Ann Transplant 2018; 23:577-590. [PMID: 30115901 PMCID: PMC6248318 DOI: 10.12659/aot.909646] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background T cell depleting antibody therapy with rabbit anti-thymocyte globulin (rATG) is the treatment of choice for glucocorticoid-resistant acute kidney allograft rejection (AR) and is used as first-line therapy in severe AR. Almost all studies investigating the effectiveness of rATG for this indication were conducted at the time when cyclosporine A and azathioprine were the standard of care. Here, the long-term outcome of rATG for AR in patients using the current standard immunosuppressive therapy (i.e., tacrolimus and mycophenolate mofetil) is described. Material/Methods Between 2002 to 2012, 108 patients were treated with rATG for AR. Data on kidney function in the year following rATG and long-term outcomes were collected. Results Overall survival after rATG was comparable to overall survival of all kidney transplantation patients (P=0.10). Serum creatinine 1 year after rATG was 179 μmol/L (interquartile range (IQR) 136–234 μmol/L) and was comparable to baseline serum creatinine (P=0.22). Early AR showed better allograft survival than late AR (P=0.0007). In addition, 1 year after AR, serum creatinine was lower in early AR (157 mol/L; IQR 131–203) compared to late AR (216 mol/L; IQR 165–269; P<0.05). The Banff grade of rejection, kidney function at the moment of rejection, and reason for rATG (severe or glucocorticoid resistant AR) did not influence the allograft survival. Conclusions Treatment of AR with rATG is effective in patients using current standard immunosuppressive therapy, even in patients with poor allograft function. Early identification of AR followed by T cell depleting treatment leads to better allograft outcomes.
Collapse
Affiliation(s)
- Marieke van der Zwan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Marian C Clahsen-Van Groningen
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Joke I Roodnat
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Anne P Bouvy
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Casper L Slachmuylders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Willem Weimar
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Marcia M L Kho
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| |
Collapse
|
107
|
Murakami N, Ding Y, Cohen DJ, Chandraker AK, Rennke HG. Recurrent membranous nephropathy and acute cellular rejection in a patient treated with direct anti-HCV therapy (ledipasvir/sofosbuvir). Transpl Infect Dis 2018; 20:e12959. [PMID: 29968947 DOI: 10.1111/tid.12959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/13/2018] [Accepted: 06/19/2018] [Indexed: 01/04/2023]
Abstract
Direct-acting antiviral agents (DAAs) are very effective therapy for chronic hepatitis C infection, and have revolutionized the treatment of hepatitis C in kidney allograft recipients. Although well tolerated in general, rare renal complications have been reported. We describe a case of recurrent membranous nephropathy and acute cellular rejection in a kidney allograft recipient after DAA (ledipasvir/sofosbuvir) therapy, whose allograft function had been stable for more than 30 years. The patient was presented with nephrotic range proteinuria with stable creatinine. The kidney allograft biopsy revealed recurrent membranous nephropathy with fine granular deposits of IgG1/IgG4 codominance and positive phospholipase A2 receptor (PLA2R) staining. The patient was treated with pulse steroid and rituximab, leading to a decrease in proteinuria. As DAAs are more frequently used, physicians should be aware of immune-related renal complications.
Collapse
Affiliation(s)
- Naoka Murakami
- Schuster Transplant Research Center, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yanli Ding
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - David J Cohen
- West Palm Beach VA Medical Canter, West Palm Beach, Florida
| | - Anil K Chandraker
- Schuster Transplant Research Center, Brigham and Women's Hospital, Boston, Massachusetts
| | - Helmut G Rennke
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
108
|
Alkadi MM, Kim J, Aull MJ, Schwartz JE, Lee JR, Watkins A, Lee JB, Dadhania DM, Seshan SV, Serur D, Kapur S, Suthanthiran M, Hartono C, Muthukumar T. Kidney allograft failure in the steroid-free immunosuppression era: A matched case-control study. Clin Transplant 2018; 31. [PMID: 28921709 DOI: 10.1111/ctr.13117] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
We studied the causes and predictors of death-censored kidney allograft failure among 1670 kidney recipients transplanted at our center in the corticosteroid-free maintenance immunosuppression era. As of January 1, 2012, we identified 137 recipients with allograft failure; 130 of them (cases) were matched 1-1 for recipient age, calendar year of transplant, and donor type with 130 recipients with functioning grafts (controls). Median time to allograft failure was 29 months (interquartile range: 18-51). Physician-validated and biopsy-confirmed categories of allograft failure were as follows: acute rejection (21%), glomerular disease (19%), transplant glomerulopathy (13%), interstitial fibrosis tubular atrophy (10%), and polyomavirus-associated nephropathy (7%). Graft failures were attributed to medical conditions in 21% and remained unresolved in 9%. Donor race, donor age, human leukocyte antigen mismatches, serum creatinine, urinary protein, acute cellular rejection, acute antibody-mediated rejection, BK viremia, and CMV viremia were associated with allograft failure. Independent predictors of allograft failure were acute cellular rejection (odds ratio: 18.31, 95% confidence interval: 5.28-63.45) and urine protein ≥1 g/d within the first year post-transplantation (5.85, 2.37-14.45). Serum creatinine ≤1.5 mg/dL within the first year post-transplantation reduced the odds (0.29, 0.13-0.64) of allograft failure. Our study has identified modifiable risk factors to reduce the burden of allograft failure.
Collapse
Affiliation(s)
- Mohamad M Alkadi
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Division of Nephrology, Department of Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Jim Kim
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Meredith J Aull
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Joseph E Schwartz
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - John R Lee
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Anthony Watkins
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Jun B Lee
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,The Rogosin Institute, New York, NY, USA
| | - Darshana M Dadhania
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Surya V Seshan
- Department of Pathology, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - David Serur
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,The Rogosin Institute, New York, NY, USA
| | - Sandip Kapur
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Manikkam Suthanthiran
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Choli Hartono
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,The Rogosin Institute, New York, NY, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| |
Collapse
|
109
|
Aala A, Brennan DC. Transformation in Immunosuppression: Are We Ready for it? J Am Soc Nephrol 2018; 29:1791-1792. [PMID: 29884733 DOI: 10.1681/asn.2018050491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Amtul Aala
- Division of Nephrology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel C Brennan
- Division of Nephrology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
110
|
Mazdak H, Ghavami M, Dolatkhah S, Daneshpajouhnejad P, Fesharakizadeh M, Fesharakizadeh S, Atapour A, Mahzouni P, Hashemi M, Salajegheh R, Taheri D. Pathological assessment of allograft nephrectomy: An Iranian experience. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2018; 23:55. [PMID: 30057639 PMCID: PMC6040153 DOI: 10.4103/jrms.jrms_440_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/20/2017] [Accepted: 04/16/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND The aim of this study was to determine the pathologic causes of renal allograft failure in transplant nephrectomy specimens. MATERIALS AND METHODS In this cross-sectional study performed in the referral transplant center of Isfahan, Iran, medical files of all patients who underwent nephrectomy in 2008-2013 were studied. Age at transplantation, sex, donor's characteristics, causes of primary renal failure, duration of allograft function, and pathologic reasons of nephrectomy were extracted. Slides of nephrectomy biopsies were evaluated. Data were analyzed using SPSS. RESULTS Medical files of 39 individuals (male: 56.4%; mean age: 35.1 ± 16.0 years) were evaluated. The main disease of patients was hypertension (17.9%), and most cases (64.1%) were nephrectomized < 6 months posttransplantation. Renal vein thrombosis (RVT) (51.3%) and T-cell-mediated rejection (TCMR) (41.0%) were the most prevalent causes of transplanted nephrectomy. Cause of primary renal failure was correlated to nephrectomy result (P = 0.04). TCMR was the only pathologic finding in all of patients nephrectomized >2 years posttransplantation. There were 14 cases in which biopsy results showed a relationship between primary disease of patients and pathologic assessment of allograft (P = 0.04). A significant relationship between transplantation-nephrectomy interval and both the nephrectomy result and histopathologic result existed (P < 0.0001). A relationship between primary allograft biopsy appearance and further assessment of nephrectomized specimen (P < 0.001) existed as well. CONCLUSION The most pathologic diagnoses of nephrectomy in a period of less than and more than 6 months posttransplantation were RVT and TCMR, respectively. Early obtained allograft protocol biopsy is suggested, which leads to better diagnosis of allograft failure.
Collapse
Affiliation(s)
- Hamid Mazdak
- Department of Urology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojgan Ghavami
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahaboddin Dolatkhah
- Department of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parnaz Daneshpajouhnejad
- Isfahan Medical Students’ Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Pathology, Isfahan Kidney Disease Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Fesharakizadeh
- Department of Surgery, Najaf Abad Branch of Islamic Azad University, Isfahan, Iran
| | | | - Abdolamir Atapour
- Department of Nephrology, Isfahan University of Medical Sciences, Isfahan, Iran
- Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Mahzouni
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozaffar Hashemi
- Department of Thoracic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roxana Salajegheh
- Department of Pathology, Faculty of Medicine, Yazd University of Medical Sciences, Yazd, Iran
| | - Diana Taheri
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
- Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
111
|
Sicard A, Levings MK, Scott DW. Engineering therapeutic T cells to suppress alloimmune responses using TCRs, CARs, or BARs. Am J Transplant 2018; 18:1305-1311. [PMID: 29603617 PMCID: PMC5992079 DOI: 10.1111/ajt.14747] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/04/2018] [Accepted: 03/20/2018] [Indexed: 01/25/2023]
Abstract
Adoptive cell therapy with therapeutic T cells has become one of the most promising strategies to stimulate or suppress immune responses. Using virus-mediated genetic manipulation, the antigen specificity of T cells can now be precisely redirected. Tailored specificity has not only overcome technical limitations and safety concerns but also considerably broadened the spectrum of therapeutic applications. Different T cell-engineering strategies have now become available to suppress alloimmune responses. We first provide an overview of the allorecognition pathways and effector mechanisms that are responsible for alloimmune injuries in the setting of vascularized organ transplantation. We then discuss the potential to use different T cell-engineering approaches to suppress alloimmune responses. Specifically, expression of allospecific T cell receptors, single-chain chimeric antigen receptors, or antigen domains recognized by B cell receptors (B cell antibody receptors) in regulatory or cytotoxic T cells are considered. The ability of these strategies to control the direct or indirect pathways of allorecognition and the cellular or humoral alloimmune responses is discussed. An intimate understanding of the complex interplay that occurs between the engineered T cells and the alloimmune players is a necessary prerequisite for the design of safe and successful strategies for precise immunomodulation in transplantation.
Collapse
Affiliation(s)
- Antoine Sicard
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada,Department of Surgery, University of British Columbia, Vancouver, BC, Canada,Department of Nephrology, University Hospital of Nice, Nice and CNRS, Institute of Molecular and Cellular Pharmacology, Valbonne, France
| | - Megan K. Levings
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada,Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - David W. Scott
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
112
|
Pascual J, Berger SP, Witzke O, Tedesco H, Mulgaonkar S, Qazi Y, Chadban S, Oppenheimer F, Sommerer C, Oberbauer R, Watarai Y, Legendre C, Citterio F, Henry M, Srinivas TR, Luo WL, Marti A, Bernhardt P, Vincenti F. Everolimus with Reduced Calcineurin Inhibitor Exposure in Renal Transplantation. J Am Soc Nephrol 2018; 29:1979-1991. [PMID: 29752413 DOI: 10.1681/asn.2018010009] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/08/2018] [Indexed: 12/28/2022] Open
Abstract
Background Everolimus permits reduced calcineurin inhibitor (CNI) exposure, but the efficacy and safety outcomes of this treatment after kidney transplant require confirmation.Methods In a multicenter noninferiority trial, we randomized 2037 de novo kidney transplant recipients to receive, in combination with induction therapy and corticosteroids, everolimus with reduced-exposure CNI (everolimus arm) or mycophenolic acid (MPA) with standard-exposure CNI (MPA arm). The primary end point was treated biopsy-proven acute rejection or eGFR<50 ml/min per 1.73 m2 at post-transplant month 12 using a 10% noninferiority margin.Results In the intent-to-treat population (everolimus n=1022, MPA n=1015), the primary end point incidence was 48.2% (493) with everolimus and 45.1% (457) with MPA (difference 3.2%; 95% confidence interval, -1.3% to 7.6%). Similar between-treatment differences in incidence were observed in the subgroups of patients who received tacrolimus or cyclosporine. Treated biopsy-proven acute rejection, graft loss, or death at post-transplant month 12 occurred in 14.9% and 12.5% of patients treated with everolimus and MPA, respectively (difference 2.3%; 95% confidence interval, -1.7% to 6.4%). De novo donor-specific antibody incidence at 12 months and antibody-mediated rejection rate did not differ between arms. Cytomegalovirus (3.6% versus 13.3%) and BK virus infections (4.3% versus 8.0%) were less frequent in the everolimus arm than in the MPA arm. Overall, 23.0% and 11.9% of patients treated with everolimus and MPA, respectively, discontinued the study drug because of adverse events.Conclusions In kidney transplant recipients at mild-to-moderate immunologic risk, everolimus was noninferior to MPA for a binary composite end point assessing immunosuppressive efficacy and preservation of graft function.
Collapse
Affiliation(s)
- Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain;
| | - Stefan P Berger
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Oliver Witzke
- Department of Infectious Diseases and Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Germany
| | - Helio Tedesco
- Nephrology Division, Hospital do Rim, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Shamkant Mulgaonkar
- Renal and Pancreas Division, St. Barnabas Medical Center, Livingston, New Jersey
| | - Yasir Qazi
- Division of Nephrology, Keck School of Medicine Renal Transplant Program, University of Southern California, Los Angeles, California
| | - Steven Chadban
- Department of Renal Medicine and Transplantation, Renal Medicine and Transplantation, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Federico Oppenheimer
- Department of Nephrology and Renal Transplantation, Renal Transplant Unit, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Claudia Sommerer
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, University Clinic for Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Yoshihiko Watarai
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya-City, Aich, Japan
| | - Christophe Legendre
- Department of Kidney Transplantation, Adult Transplantation Service, Paris Descartes University and Necker Hospital, Paris, France
| | - Franco Citterio
- Department of Surgery, Renal Transplantation, Catholic University, Rome, Italy
| | - Mitchell Henry
- Department of Surgery, The Comprehensive Transplant Center, The Ohio State University, Wexner Medical Center, Columbus, Ohio
| | - Titte R Srinivas
- Division of Nephrology, Medical University of South Carolina, Mount Pleasant, South Carolina
| | - Wen-Lin Luo
- Department of Biometrics and Statistical Science, Novartis Pharmaceuticals, East Hanover, New Jersey
| | - AnaMaria Marti
- Department of Research and Development, Novartis Pharma AG, Basel, Switzerland; and
| | - Peter Bernhardt
- Department of Research and Development, Novartis Pharma AG, Basel, Switzerland; and
| | - Flavio Vincenti
- Department of Surgery, Kidney Transplant Service, University of California, San Francisco, California
| | | |
Collapse
|
113
|
Severova-Andreevska G, Grcevska L, Petrushevska G, Cakalaroski K, Sikole A, Stojceva–Taneva O, Danilovska I, Ivanovski N. The Spectrum of Histopathological Changes in the Renal Allograft - a 12 Months Protocol Biopsy Study. Open Access Maced J Med Sci 2018; 6:606-612. [PMID: 29731924 PMCID: PMC5927487 DOI: 10.3889/oamjms.2018.162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/16/2017] [Accepted: 03/17/2018] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Renal transplantation became a routine and successful medical treatment for Chronic Kidney Disease in the last 30 years all over the world. Introduction of Luminex based Single Antigen Beads (SAB) and recent BANFF consensus of histopathological phenotypes of different forms of rejection enables more precise diagnosis and changes the therapeutic approach. The graft biopsies, protocol or cause, indicated, remain a golden diagnostic tool for clinical follow up of kidney transplant recipients (KTR). AIM The study aimed to analyse the histopathological changes in renal grafts 12 months after the surgery in KTR with satisfactory kidney function. MATERIAL AND METHODS A 12-month protocol biopsy study was performed in a cohort of 50 Kidney transplant recipients (42 from living and 8 from deceased donors). Usual work-up for suitable donors and recipients, standard surgical procedure, basic principles of peri and postoperative care and follow up were done in all KTR. Sequential quadruple immunosuppression including induction with Anti-thymocyte globulin (ATG) or Interleukin-2R antagonist (IL-2R), and triple drug maintenance therapy with Calcineurin Inhibitors (CNI), Mycophenolate Mofetil (MMF) and Steroids were prescribed to all pts. Different forms of Glomerulonephritis (16), Hypertension (10), End Stage Renal Disease (13), Hereditary Nephropathies (6), Diabetes (3) and Vesicoureteral Reflux (2) were the underlying diseases. All biopsies were performed under ultrasound guidance. The 16 gauge needles with automated "gun" were used to take 2 cores of tissue. The samples were stained with HE, PAS, Trichrome Masson and Silver and reviewed by the same pathologist. A revised and uploaded BANFF 2013 classification in 6 categories (Cat) was used. RESULTS Out of 48 biopsies, 15 (31%) were considered as normal, 4 (8%), Borderline (BL-Cat 3), 5 (10%) as Interstitial Fibrosis/Tubular Atrophy (IF/TA-Cat 5), 5 (10%) were classified as non-immunological (Cat 6), 2 as a pure antibody-mediated rejection (ABMR-Cat 2) and T-cell Mediated Rejection (TCMR-Cat 4). The remaining 17 samples were classified as a "mixed" rejection: 7 (41%) ABMR + IF/TA, 5 (29%) ABMR + BL + IF/TA, 2 (11%) BL + IF/TA, 1 (5%) ABMR + BL, 1 (5%) ABMR + TCMR and 1 (5%) TCMR + IF/TA. The mean serum creatinine at the time of the biopsy was 126.7 ± 23.4 µmol/L, while GFR-MDRD 63.4 ± 20.7 ml/min, which means that the majority of the findings were subclinical. Among the non-immunological histological findings (Cat 6), 3 cases belonged to CNI toxicity, 1 to BK nephropathy and 1 to recurrence of the primary disease. CONCLUSION Our 12-month protocol biopsy study revealed the presence of different forms of mixed subclinical rejection. Use of recent BANFF classification and scoring system enables more precise diagnosis and subsequently different approach to the further treatment of the KTR. More correlative long-term studies including Anti HLA antibodies and Endothelial Cell Activation- Associated Transcripts (ENDAT) are needed.
Collapse
Affiliation(s)
- Galina Severova-Andreevska
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Ladislava Grcevska
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Gordana Petrushevska
- Institute for Pathology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Koco Cakalaroski
- Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Aleksandar Sikole
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Olivera Stojceva–Taneva
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Ilina Danilovska
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Ninoslav Ivanovski
- Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
114
|
Halloran PF, Venner JM, Madill-Thomsen KS, Einecke G, Parkes MD, Hidalgo LG, Famulski KS. Review: The transcripts associated with organ allograft rejection. Am J Transplant 2018; 18:785-795. [PMID: 29178397 DOI: 10.1111/ajt.14600] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/31/2017] [Accepted: 11/17/2017] [Indexed: 01/25/2023]
Abstract
The molecular mechanisms operating in human organ transplant rejection are best inferred from the mRNAs expressed in biopsies because the corresponding proteins often have low expression and short half-lives, while small non-coding RNAs lack specificity. Associations should be characterized in a population that rigorously identifies T cell-mediated (TCMR) and antibody-mediated rejection (ABMR). This is best achieved in kidney transplant biopsies, but the results are generalizable to heart, lung, or liver transplants. Associations can be universal (all rejection), TCMR-selective, or ABMR-selective, with universal being strongest and ABMR-selective weakest. Top universal transcripts are IFNG-inducible (eg, CXCL11 IDO1, WARS) or shared by effector T cells (ETCs) and NK cells (eg, KLRD1, CCL4). TCMR-selective transcripts are expressed in activated ETCs (eg, CTLA4, IFNG), activated (eg, ADAMDEC1), or IFNG-induced macrophages (eg, ANKRD22). ABMR-selective transcripts are expressed in NK cells (eg, FGFBP2, GNLY) and endothelial cells (eg, ROBO4, DARC). Transcript associations are highly reproducible between biopsy sets when the same rejection definitions, case mix, algorithm, and technology are applied, but exact ranks will vary. Previously published rejection-associated transcripts resemble universal and TCMR-selective transcripts due to incomplete representation of ABMR. Rejection-associated transcripts are never completely rejection-specific because they are shared with the stereotyped response-to-injury and innate immunity.
Collapse
Affiliation(s)
- Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Medicine, Division of Nephrology and Transplant Immunology, University of Alberta, Edmonton, AB, Canada
| | - Jeffery M Venner
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada
| | - Katelynn S Madill-Thomsen
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Medicine, Division of Nephrology and Transplant Immunology, University of Alberta, Edmonton, AB, Canada
| | | | - Michael D Parkes
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada
| | - Luis G Hidalgo
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Konrad S Famulski
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
115
|
Einecke G, Reeve J, Halloran PF. A molecular biopsy test based on arteriolar under-hyalinosis reflects increased probability of rejection related to under-immunosuppression. Am J Transplant 2018; 18:821-831. [PMID: 28985016 DOI: 10.1111/ajt.14532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/15/2017] [Accepted: 09/26/2017] [Indexed: 01/25/2023]
Abstract
Calcineurin inhibitor immunosuppressive drugs induce changes such as arteriolar hyalinosis (ah) in kidney transplants, raising the possibility that molecular changes in biopsies related to histologic ah can provide information about drug exposure. We hypothesized that molecular changes associated with less-than-expected hyalinosis might highlight a subpopulation of patients with under-immunosuppression/nonadherence at intermediate times of biopsy posttransplant (TxBx). Using gene expression data from 562 indication biopsies, we developed a molecular classifier for predicting the expected ah lesions (Mah ) at a particular TxBx. Mah -scores increased linearly with log(TxBx), but some biopsies had lower scores than expected for TxBx. The deviation of individual Mah -scores below the predicted regression line of Mah -scores vs TxBx is defined as "low hyalinosis index." Low hyalinosis indices were frequent in biopsies between 3 months and 3 years posttransplant, particularly among biopsies lacking histologic hyalinosis (ah0), and were associated with T cell-mediated rejection and a subset of recent-onset antibody-mediated rejection without glomerular double contours. In patients with medical records available for review, low hyalinosis indices were frequently associated with physician-recorded concerns about nonadherence (suspected or proven). We conclude that the Mah classifier and hyalinosis index identify indication biopsies with rejection for which the possibility of patient nonadherence should be considered.
Collapse
Affiliation(s)
- Gunilla Einecke
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Jeff Reeve
- Alberta Transplant Applied Genomics Centre Edmonton, Edmonton, AB, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Philip F Halloran
- Alberta Transplant Applied Genomics Centre Edmonton, Edmonton, AB, Canada
- Department of Medicine, Division of Nephrology and Transplant Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
116
|
Affiliation(s)
- Konrad S Famulski
- University of Alberta, Edmonton, Alberta, Canada.,Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
| | - Philip F Halloran
- University of Alberta, Edmonton, Alberta, Canada.,Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
| |
Collapse
|
117
|
Chen CC, Koenig A, Saison C, Dahdal S, Rigault G, Barba T, Taillardet M, Chartoire D, Ovize M, Morelon E, Defrance T, Thaunat O. CD4+ T Cell Help Is Mandatory for Naive and Memory Donor-Specific Antibody Responses: Impact of Therapeutic Immunosuppression. Front Immunol 2018. [PMID: 29515582 PMCID: PMC5825980 DOI: 10.3389/fimmu.2018.00275] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Antibody-mediated rejection is currently the leading cause of transplant failure. Prevailing dogma predicts that B cells differentiate into anti-donor-specific antibody (DSA)-producing plasma cells only with the help of CD4+ T cells. Yet, previous studies have shown that dependence on helper T cells decreases when high amounts of protein antigen are recruited to the spleen, two conditions potentially met by organ transplantation. This could explain why a significant proportion of transplant recipients develop DSA despite therapeutic immunosuppression. Using murine models, we confirmed that heart transplantation, but not skin grafting, is associated with accumulation of a high quantity of alloantigens in recipients’ spleen. Nevertheless, neither naive nor memory DSA responses could be observed after transplantation of an allogeneic heart into recipients genetically deficient for CD4+ T cells. These findings suggest that DSA generation rather result from insufficient blockade of the helper function of CD4+ T cells by therapeutic immunosuppression. To test this second theory, different subsets of circulating T cells: CD8+, CD4+, and T follicular helper [CD4+CXCDR5+, T follicular helper cells (Tfh)], were analyzed in 9 healthy controls and 22 renal recipients. In line with our hypothesis, we observed that triple maintenance immunosuppression (CNI + MMF + steroids) efficiently blocked activation-induced upregulation of CD25 on CD8+, but not on CD4+ T cells. Although the level of expression of CD40L and ICOS was lower on activated Tfh of immunosuppressed patients, the percentage of CD40L-expressing Tfh was the same than control patients, as was Tfh production of IL21. Induction therapy with antithymocyte globulin (ATG) resulted in prolonged depletion of Tfh and reduction of CD4+ T cells number with depleting monoclonal antibody in murine model resulted in exponential decrease in DSA titers. Furthermore, induction with ATG also had long-term beneficial influence on Tfh function after immune reconstitution. We conclude that CD4+ T cell help is mandatory for naive and memory DSA responses, making Tfh cells attractive targets for improving the prevention of DSA generation and to prolong allograft survival. Waiting for innovative treatments to be translated into the clinical field ATG induction seems to currently offer the best clinical prospect to achieve this goal.
Collapse
Affiliation(s)
- Chien-Chia Chen
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,IHU OPERA, Cardioprotection Laboratory, Hospices Civils de Lyon, CIC, Bron, France
| | - Alice Koenig
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Carole Saison
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
| | - Suzan Dahdal
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
| | - Guillaume Rigault
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Thomas Barba
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Morgan Taillardet
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Dimitri Chartoire
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Michel Ovize
- IHU OPERA, Cardioprotection Laboratory, Hospices Civils de Lyon, CIC, Bron, France.,Lyon-Est Medical Faculty, Claude Bernard University Lyon 1, Lyon, France
| | - Emmanuel Morelon
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,IHU OPERA, Cardioprotection Laboratory, Hospices Civils de Lyon, CIC, Bron, France.,Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University Lyon 1, Lyon, France
| | - Thierry Defrance
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Olivier Thaunat
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,IHU OPERA, Cardioprotection Laboratory, Hospices Civils de Lyon, CIC, Bron, France.,Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University Lyon 1, Lyon, France
| |
Collapse
|
118
|
de Leur K, Clahsen-van Groningen MC, van den Bosch TPP, de Graav GN, Hesselink DA, Samsom JN, Baan CC, Boer K. Characterization of ectopic lymphoid structures in different types of acute renal allograft rejection. Clin Exp Immunol 2018; 192:224-232. [PMID: 29319177 DOI: 10.1111/cei.13099] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2018] [Indexed: 12/15/2022] Open
Abstract
We hypothesize that T cells such as interleukin (IL)-21+ B cell lymphoma 6 (BCL6)+ T follicular helper cells can regulate B cell-mediated immunity within the allograft during acute T cell-mediated rejection; this process may feed chronic allograft rejection in the long term. To investigate this mechanism, we determined the presence and activation status of organized T and B cells in so-called ectopic lymphoid structures (ELSs) in different types of acute renal allograft rejection. Biopsies showing the following primary diagnosis were included: acute/active antibody-mediated rejection, C4d+ (a/aABMR), acute T cell-mediated rejection grade I (aTCMRI) and acute T cell-mediated rejection grade II (aTCMRII). Paraffin sections were stained for T cells (CD3 and CD4), B cells (CD20), follicular dendritic cells (FDCs, CD23), activated B cells (CD79A), immunoglobulin (Ig)D, cell proliferation (Ki67) and double immunofluorescent stainings for IL-21 and BCL6 were performed. Infiltrates of T cells were detected in all biopsies. In aTCMRI, B cells formed aggregates surrounded by T cells. In these aggregates, FDCs, IgD and Ki67 were detected, suggesting the presence of ELSs. In contrast, a/aABMR and aTCMRII showed diffuse infiltrates of T and B cells but no FDCs and IgD. IL-21 was present in all biopsies. However, co-localization with BCL6 was observed mainly in aTCMRI biopsies. In conclusion, ELSs with an activated phenotype are found predominantly in aTCMRI where T cells co-localize with B cells. These findings suggest a direct pathway of B cell alloactivation at the graft site during T cell mediated rejection.
Collapse
Affiliation(s)
- K de Leur
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.,Division of HPB and Transplant Surgery, Department of Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - T P P van den Bosch
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - G N de Graav
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - D A Hesselink
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C C Baan
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - K Boer
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
119
|
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: 765] [Impact Index Per Article: 109.3] [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
|
120
|
Human Cytomegalovirus Infection Increases Both Antibody- and Non-Antibody-Dependent Cellular Reactivity by Natural Killer Cells. Transplant Direct 2017. [PMID: 29536036 PMCID: PMC5828690 DOI: 10.1097/txd.0000000000000750] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Supplemental digital content is available in the text. Background Antibody-mediated rejection in solid organ transplantation is an important immunological barrier to successful long-term graft survival. Next to complement activation, natural killer (NK) cells have been implicated in the process. Human cytomegalovirus (CMV), independently associated with decreased graft survival, has a strong imprint on the immune response. Here, we assessed the effect of CMV status on alloreactive NK cell reactivity. Methods We compared antibody-mediated NK cell cytolytic activity (CD107a expression) and IFNγ production between healthy CMV-seropositive (n = 8) and CMV-seronegative (n = 11) individuals, in cocultures of NK cells with anti-HLA class I or rituximab (control) antibody-coated Raji cells. Results First, we showed that within the NKG2C+ NK cells, it is specifically the NKG2C+/A− subset that is enriched in CMV+ individuals. We then observed that in particular the NK cell antibody-dependent cell mediated cytotoxicity (ADCC), but also non-ADCC alloreactivity toward HLA-positive target cells was increased in CMV+ individuals as compared to CMV− ones. This enhanced ADCC as well as non-ADCC NK cell reactivity in CMV+ individuals was particularly characterized by a significantly higher number of ILT2+ and NKG2C+ NK cells that possessed cytolytic activity and/or produced IFNγ in response to HLA-positive target cells. Conclusions With regard to organ transplantation, these data suggest that CMV infection enhances NK cell alloreactivity, which may pose an additional adverse effect on graft survival, especially in the presence of donor specific antibodies.
Collapse
|
121
|
Chen CC, Pouliquen E, Broisat A, Andreata F, Racapé M, Bruneval P, Kessler L, Ahmadi M, Bacot S, Saison-Delaplace C, Marcaud M, Van Huyen JPD, Loupy A, Villard J, Demuylder-Mischler S, Berney T, Morelon E, Tsai MK, Kolopp-Sarda MN, Koenig A, Mathias V, Ducreux S, Ghezzi C, Dubois V, Nicoletti A, Defrance T, Thaunat O. Endothelial chimerism and vascular sequestration protect pancreatic islet grafts from antibody-mediated rejection. J Clin Invest 2017; 128:219-232. [PMID: 29202467 DOI: 10.1172/jci93542] [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: 02/23/2017] [Accepted: 10/12/2017] [Indexed: 12/31/2022] Open
Abstract
Humoral rejection is the most common cause of solid organ transplant failure. Here, we evaluated a cohort of 49 patients who were successfully grafted with allogenic islets and determined that the appearance of donor-specific anti-HLA antibodies (DSAs) did not accelerate the rate of islet graft attrition, suggesting resistance to humoral rejection. Murine DSAs bound to allogeneic targets expressed by islet cells and induced their destruction in vitro; however, passive transfer of the same DSAs did not affect islet graft survival in murine models. Live imaging revealed that DSAs were sequestrated in the circulation of the recipients and failed to reach the endocrine cells of grafted islets. We used murine heart transplantation models to confirm that endothelial cells were the only accessible targets for DSAs, which induced the development of typical microvascular lesions in allogeneic transplants. In contrast, the vasculature of DSA-exposed allogeneic islet grafts was devoid of lesions because sprouting of recipient capillaries reestablished blood flow in grafted islets. Thus, we conclude that endothelial chimerism combined with vascular sequestration of DSAs protects islet grafts from humoral rejection. The reduced immunoglobulin concentrations in the interstitial tissue, confirmed in patients, may have important implications for biotherapies such as vaccines and monoclonal antibodies.
Collapse
Affiliation(s)
- Chien-Chia Chen
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Eric Pouliquen
- Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
| | - Alexis Broisat
- French National Institute of Health and Medical Research (INSERM) Unit 1039, Grenoble, France; Bioclinical Radiopharmaceutical Laboratory, Joseph Fourier University (Grenoble 1), Grenoble, France
| | - Francesco Andreata
- French National Institute of Health and Medical Research (INSERM) Unit 1148, Laboratory of Vascular Translational Science, F-75018, Paris, France; Paris Diderot University, Paris, France
| | - Maud Racapé
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, Paris, France
| | - Patrick Bruneval
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, Paris, France
| | - Laurence Kessler
- Department of Diabetology, University Hospital, Strasbourg, France; Federation of Translational Medicine of Strasbourg, University of Strasbourg, Strasbourg, France.,Groupe Rhin-Rhône-Alpes-Genève pour la Greffe d'Ilots de Langerhans (GRAGIL) Consortium
| | - Mitra Ahmadi
- French National Institute of Health and Medical Research (INSERM) Unit 1039, Grenoble, France; Bioclinical Radiopharmaceutical Laboratory, Joseph Fourier University (Grenoble 1), Grenoble, France
| | - Sandrine Bacot
- French National Institute of Health and Medical Research (INSERM) Unit 1039, Grenoble, France; Bioclinical Radiopharmaceutical Laboratory, Joseph Fourier University (Grenoble 1), Grenoble, France
| | - Carole Saison-Delaplace
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
| | - Marina Marcaud
- Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
| | - Jean-Paul Duong Van Huyen
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, Paris, France
| | - Alexandre Loupy
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, Paris, France.,Department of Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean Villard
- Department of Immunology and Allergy and Department of Laboratory Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Sandrine Demuylder-Mischler
- Department of Surgery, Islet Isolation, and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Thierry Berney
- Groupe Rhin-Rhône-Alpes-Genève pour la Greffe d'Ilots de Langerhans (GRAGIL) Consortium.,Department of Surgery, Islet Isolation, and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Emmanuel Morelon
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France.,Groupe Rhin-Rhône-Alpes-Genève pour la Greffe d'Ilots de Langerhans (GRAGIL) Consortium.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Meng-Kun Tsai
- Department of Surgery, National Taiwan University Hospital and National Taiwan University, College of Medicine, Taipei, Taiwan
| | | | - Alice Koenig
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Virginie Mathias
- French National Blood Service (EFS), HLA Laboratory, Lyon, France
| | | | - Catherine Ghezzi
- French National Institute of Health and Medical Research (INSERM) Unit 1039, Grenoble, France; Bioclinical Radiopharmaceutical Laboratory, Joseph Fourier University (Grenoble 1), Grenoble, France
| | - Valerie Dubois
- French National Blood Service (EFS), HLA Laboratory, Lyon, France
| | - Antonino Nicoletti
- French National Institute of Health and Medical Research (INSERM) Unit 1148, Laboratory of Vascular Translational Science, F-75018, Paris, France; Paris Diderot University, Paris, France
| | - Thierry Defrance
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Olivier Thaunat
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Edouard Herriot University Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France.,Groupe Rhin-Rhône-Alpes-Genève pour la Greffe d'Ilots de Langerhans (GRAGIL) Consortium.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
122
|
Evolving Approaches in the Identification of Allograft-Reactive T and B Cells in Mice and Humans. Transplantation 2017; 101:2671-2681. [PMID: 28604446 DOI: 10.1097/tp.0000000000001847] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Whether a transplanted allograft is stably accepted, rejected, or achieves immunological tolerance is dependent on the frequency and function of alloreactive lymphocytes, making the identification and analysis of alloreactive T and B cells in transplant recipients critical for understanding mechanisms, and the prediction of allograft outcome. In animal models, tracking the fate of graft-reactive T and B cells allows investigators to uncover their biology and develop new therapeutic strategies to protect the graft. In the clinic, identification and quantification of graft-reactive T and B cells allows for the early diagnosis of immune reactivity and therapeutic intervention to prevent graft loss. In addition to rejection, probing of T and B cell fate in vivo provides insights into the underlying mechanisms of alloimmunity or tolerance that may lead to biomarkers predicting graft fate. In this review, we discuss existing and developing approaches to track and analyze alloreactive T and B cells in mice and humans and provide examples of discoveries made utilizing these techniques. These approaches include mixed lymphocyte reactions, trans-vivo delayed-type hypersensitivity, enzyme-linked immunospot assays, the use of antigen receptor transgenic lymphocytes, and utilization of peptide-major histocompatibility multimers, along with imaging techniques for static multiparameter analysis or dynamic in vivo tracking. Such approaches have already refined our understanding of the alloimmune response and are pointing to new ways to improve allograft outcomes in the clinic.
Collapse
|
123
|
Halloran PF, Potena L, Van Huyen JPD, Bruneval P, Leone O, Kim DH, Jouven X, Reeve J, Loupy A. Building a tissue-based molecular diagnostic system in heart transplant rejection: The heart Molecular Microscope Diagnostic (MMDx) System. J Heart Lung Transplant 2017; 36:1192-1200. [DOI: 10.1016/j.healun.2017.05.029] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
|
124
|
Halloran PF, Reeve J, Akalin E, Aubert O, Bohmig GA, Brennan D, Bromberg J, Einecke G, Eskandary F, Gosset C, Duong Van Huyen JP, Gupta G, Lefaucheur C, Malone A, Mannon RB, Seron D, Sellares J, Weir M, Loupy A. Real Time Central Assessment of Kidney Transplant Indication Biopsies by Microarrays: The INTERCOMEX Study. Am J Transplant 2017; 17:2851-2862. [PMID: 28449409 DOI: 10.1111/ajt.14329] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/13/2017] [Accepted: 04/14/2017] [Indexed: 01/25/2023]
Abstract
The authors conducted a prospective trial to assess the feasibility of real time central molecular assessment of kidney transplant biopsy samples from 10 North American or European centers. Biopsy samples taken 1 day to 34 years posttransplantation were stabilized in RNAlater, sent via courier overnight at ambient temperature to the central laboratory, and processed (29 h workflow) using microarrays to assess T cell- and antibody-mediated rejection (TCMR and ABMR, respectively). Of 538 biopsy samples submitted, 519 (96%) were sufficient for microarray analysis (average length, 3 mm). Automated reports were generated without knowledge of histology and HLA antibody, with diagnoses assigned based on Molecular Microscope Diagnostic System (MMDx) classifier algorithms and signed out by one observer. Agreement between MMDx and histology (balanced accuracy) was 77% for TCMR, 77% for ABMR, and 76% for no rejection. A classification tree derived to provide automated sign-outs predicted the observer sign-outs with >90% accuracy. In 451 biopsy samples where feedback was obtained, clinicians indicated that MMDx more frequently agreed with clinical judgment (87%) than did histology (80%) (p = 0.0042). In 81% of feedback forms, clinicians reported that MMDx increased confidence in management compared with conventional assessment alone. The authors conclude that real time central molecular assessment is feasible and offers a useful new dimension in biopsy interpretation. ClinicalTrials.gov NCT#01299168.
Collapse
Affiliation(s)
- P F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - J Reeve
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada
| | - E Akalin
- Montefiore Medical Center, Bronx, NY
| | - O Aubert
- Paris Translational Research Center for Organ Transplantation, INSERM, Uss-S970, Paris, France
| | - G A Bohmig
- Medizinische Universität Wien, Vienna, Austria
| | - D Brennan
- Washington University at St. Louis, St. Louis, MO
| | - J Bromberg
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - G Einecke
- Medizinische Hochschule Hannover, Hannover, Germany
| | - F Eskandary
- Medizinische Universität Wien, Vienna, Austria
| | - C Gosset
- Paris Translational Research Center for Organ Transplantation, INSERM, Uss-S970, Paris, France.,Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - J-P Duong Van Huyen
- Paris Translational Research Center for Organ Transplantation, INSERM, Uss-S970, Paris, France
| | - G Gupta
- Virginia Commonwealth University, Richmond, VA
| | - C Lefaucheur
- Paris Translational Research Center for Organ Transplantation, INSERM, Uss-S970, Paris, France.,Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - A Malone
- Washington University at St. Louis, St. Louis, MO
| | - R B Mannon
- University of Alabama at Birmingham, Birmingham, AL
| | - D Seron
- Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - J Sellares
- Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - M Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - A Loupy
- Paris Translational Research Center for Organ Transplantation, INSERM, Uss-S970, Paris, France.,Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
125
|
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
|
126
|
Young JS, Khiew SHW, Yang J, Vannier A, Yin D, Sciammas R, Alegre ML, Chong AS. Successful Treatment of T Cell-Mediated Acute Rejection with Delayed CTLA4-Ig in Mice. Front Immunol 2017; 8:1169. [PMID: 28970838 PMCID: PMC5609110 DOI: 10.3389/fimmu.2017.01169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/04/2017] [Indexed: 12/25/2022] Open
Abstract
Clinical observations that kidney transplant recipients receiving belatacept who experienced T cell-mediated acute rejection can be successfully treated and subsequently maintained on belatacept-based immunosuppression suggest that belatacept is able to control memory T cells. We recently reported that treatment with CTLA4-Ig from day 6 posttransplantation successfully rescues allografts from acute rejection in a BALB/c to C57BL/6 heart transplant model, in part, by abolishing B cell germinal centers and reducing alloantibody titers. Here, we show that CTLA4-Ig is additionally able to inhibit established T cell responses independently of B cells. CTLA4-Ig inhibited the in vivo cytolytic activity of donor-specific CD8+ T cells, and the production of IFNγ by graft-infiltrating T cells. Delayed CTLA4-Ig treatment did not reduce the numbers of graft-infiltrating T cells nor prevented the accumulation of antigen-experienced donor-specific memory T cells in the spleen. Nevertheless, delayed CTLA4-Ig treatment successfully maintained long-term graft acceptance in the majority of recipients that had experienced a rejection crisis, and enabled the acceptance of secondary BALB/c heart grafts transplanted 30 days after the first transplantation. In summary, we conclude that delayed CTLA4-Ig treatment is able to partially halt ongoing T cell-mediated acute rejection. These findings extend the functional efficacy of CTLA4-Ig therapy to effector T cells and provide an explanation for why CTLA4-Ig-based immunosuppression in the clinic successfully maintains long-term graft survival after T cell-mediated rejection.
Collapse
Affiliation(s)
- James S Young
- Department of Surgery, Section of Transplantation, The University of Chicago, Chicago, IL, United States
| | - Stella H-W Khiew
- Department of Surgery, Section of Transplantation, The University of Chicago, Chicago, IL, United States
| | - Jinghui Yang
- Department of Surgery, Section of Transplantation, The University of Chicago, Chicago, IL, United States.,Department of Organ Transplantation, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Augustin Vannier
- Department of Surgery, Section of Transplantation, The University of Chicago, Chicago, IL, United States
| | - Dengping Yin
- Department of Surgery, Section of Transplantation, The University of Chicago, Chicago, IL, United States
| | - Roger Sciammas
- Center for Comparative Medicine, University of California, Davis, Davis, CA, United States
| | - Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL, United States
| | - Anita S Chong
- Department of Surgery, Section of Transplantation, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
127
|
Impact of monocyte-macrophage inhibition by ibandronate on graft function and survival after kidney transplantation: a single-centre follow-up study over 15 years. Clin Exp Nephrol 2017; 22:474-480. [PMID: 28856476 DOI: 10.1007/s10157-017-1470-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/18/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the effect of ibandronate administration on long-term graft function and graft survival after successful renal transplantation. METHODS Seventy-two renal transplant recipients (36 patients each in the treatment and control group) were included and followed over a 15-year period. Data on graft function and death-censored transplant outcome were recorded at 1, 5, 10, and 15 years. RESULTS Death-censored Kaplan-Meier analysis showed significantly improved graft survival of the treatment group (p = 0.026), whereas Cox regression analysis showed that ibandronate was positively associated with improved transplant survival (p = 0.028, hazard ratio 0.24, 95% confidence interval 0.07-0.86). Although general linear modelling did not indicate that ibandronate had a significant effect on transplant function (calculated using the estimated glomerular filtration rate according to Chronic Kidney Disease Epidemiology Collaboration equation) over the entire 15-year period (p = 0.650), there was a tendency towards improved graft function 1-year post-transplantion (p = 0.056). CONCLUSIONS Ibandronate treatment within the first year of transplantation resulted in a trend towards better graft function within the first few year post-transplant, and was associated with increased transplant survival at long-term follow-up.
Collapse
|
128
|
Kozakowski N, Eskandary F, Herkner H, Bond G, Oberbauer R, Regele H, Böhmig GA, Kikić Ž. Diffuse Extent of Peritubular Capillaritis in Late Antibody-Mediated Rejection: Associations With Levels of Donor-Specific Antibodies and Chronic Allograft Injury. Transplantation 2017; 101:e178-e187. [PMID: 28252564 DOI: 10.1097/tp.0000000000001707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Recently, diffuse peritubular capillaritis (ptc) has been suggested to independently predict chronic transplant injury and loss, and although the ptc score is a diagnostic criterion for antibody-mediated rejection, the utility of diffuse ptc is under debate. METHODS We evaluated the diagnostic value of ptc characteristics in this cross-sectional study including 85 biopsies of patients with donor-specific antibodies (DSA). Biopsies were reevaluated for the extent (diffuse vs focal), score and leukocytic composition in relation to DSA binding strength (mean fluorescence intensity [MFI]_max). Chronic allograft injury (transplant chronic glomerulopathy [cg] or chronic lesion score CLS]) were associated with ptc features. RESULTS Peritubular capillaritis was detected in 50% (76% mononuclear ptc). Peritubular capillaritis scores 1, 2, and 3 were present in 36%, 55%, and 9%, and focal or diffuse ptc in 36% or 64%. Diffuse ptc was associated with DSA MFI_max (median: 4407 vs 2419 [focal ptc; P = 0.04] or 1946 [no ptc; P = 0.004]), cg (58% vs no ptc 24% [P = 0.02]), and higher CLS (mean: 6.81 vs 4.67 [focal ptc, P = 0.01] or 5.18 [no ptc, P = 0.001]), respectively. The association of ptc score of 2 or greater with cg was slightly better than with diffuse ptc. Diffuse ptc and ptc score of 2 or greater remained independently related to cg after adjusting for DSA_MFI_max, C4d, or previous rejection episodes, however lost their independent relation after adjusting for total microcirculation scores. Diffuse ptc was the only ptc characteristic independently related to CLS. CONCLUSIONS Our results emphasize the clinical relevance of reporting diffuse ptc, which may relate to DSA binding strength and potentially to chronic graft injury.
Collapse
Affiliation(s)
- Nicolas Kozakowski
- 1 Institute of Clinical Pathology, Medical University Vienna, Vienna, Austria. 2 Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria. 3 Department of Emergency Medicine, Medical University Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
129
|
Mulley WR, Huang LL, Ramessur Chandran S, Longano A, Amos LAR, Polkinghorne KR, Nikolic-Paterson DJ, Kanellis J. Long-term graft survival in patients with chronic antibody-mediated rejection with persistent peritubular capillaritis treated with intravenous immunoglobulin and rituximab. Clin Transplant 2017. [DOI: 10.1111/ctr.13037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- William R. Mulley
- Department of Nephrology; Monash Medical Centre; Clayton Vic. Australia
- Department of Medicine; Centre for Inflammatory Diseases; Monash University; Clayton Vic. Australia
| | - Louis L. Huang
- Department of Nephrology; Monash Medical Centre; Clayton Vic. Australia
| | - Sharmila Ramessur Chandran
- Department of Nephrology; Monash Medical Centre; Clayton Vic. Australia
- Department of Medicine; Centre for Inflammatory Diseases; Monash University; Clayton Vic. Australia
| | - Anthony Longano
- Department of Anatomical Pathology; Monash Medical Centre; Clayton Vic. Australia
| | - Liv A. R. Amos
- Department of Nephrology; Monash Medical Centre; Clayton Vic. Australia
- Department of Medicine; Centre for Inflammatory Diseases; Monash University; Clayton Vic. Australia
| | - Kevan R. Polkinghorne
- Department of Nephrology; Monash Medical Centre; Clayton Vic. Australia
- Department of Medicine; Centre for Inflammatory Diseases; Monash University; Clayton Vic. Australia
- Department of Epidemiology and Preventive Medicine; Monash University; Prahran Vic. Australia
| | - David J. Nikolic-Paterson
- Department of Nephrology; Monash Medical Centre; Clayton Vic. Australia
- Department of Medicine; Centre for Inflammatory Diseases; Monash University; Clayton Vic. Australia
| | - John Kanellis
- Department of Nephrology; Monash Medical Centre; Clayton Vic. Australia
- Department of Medicine; Centre for Inflammatory Diseases; Monash University; Clayton Vic. Australia
| |
Collapse
|
130
|
Halloran PF, Venner JM, Famulski KS. Comprehensive Analysis of Transcript Changes Associated With Allograft Rejection: Combining Universal and Selective Features. Am J Transplant 2017; 17:1754-1769. [PMID: 28101959 DOI: 10.1111/ajt.14200] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/06/2017] [Accepted: 01/08/2017] [Indexed: 01/25/2023]
Abstract
We annotated the top transcripts associated with kidney transplant rejection by p-value, either universal for all rejection or selective for T cell-mediated rejection (TCMR) or antibody-mediated rejection (ABMR; ClinicalTrials.gov NCT01299168). We used eight class-comparison algorithms to interrogate microarray results from 703 biopsies, 205 with rejection. The positive comparators were all rejection, TCMR, or ABMR; the negative comparators varied from normal biopsies to all nonrejecting biopsies, including other diseases. The universal algorithm, rejection versus all nonrejection, identified transcripts mainly inducible by interferon γ. Selectivity for ABMR or TCMR required the other rejection class as well as nonrejection biopsies in the comparator to avoid selecting universal transcripts. Direct comparison of ABMR versus TCMR yielded only transcripts related to TCMR, the stronger signal. Transcripts highly associated with rejection were never completely specific for rejection: Many were increased in biopsies without rejection, reflecting sharing between rejection and injury-induced innate immunity. Union of the top 200 transcripts from universal and selective algorithms yielded 454 transcripts that permitted unsupervised analysis of biopsies in principal component analysis: PC1 was rejection, and PC2 was separation of TCMR from ABMR. Appreciating rejection-associated molecular changes requires a diverse case mix, accurate histologic classification (including C4d-negative ABMR), and both selective and universal algorithms.
Collapse
Affiliation(s)
- P F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - J M Venner
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada
| | - K S Famulski
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
131
|
Tracing Donor-MHC Class II Reactive B cells in Mouse Cardiac Transplantation: Delayed CTLA4-Ig Treatment Prevents Memory Alloreactive B-Cell Generation. Transplantation 2017; 100:1683-91. [PMID: 27362308 DOI: 10.1097/tp.0000000000001253] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND The dual role of B cells as drivers and suppressors of the immune responses have underscored the need to trace the fate of B cells recognizing donor major histocompatibility complex class I and class II after allograft transplantation. METHODS In this study, we used donor class II tetramers to trace the fate of I-E-specific B cells after immunization with BALB/c spleen cells or cardiac transplantation, in naive or sensitized C57BL/6 recipients. We combined this approach with genetic lineage tracing of memory B cells in activation-induced cytidine deaminase regulated Cre transgenic mice crossed to the ROSA26-enhanced yellow fluorescent protein reporter mice to track endogenous I-E-specific memory B cell generation. RESULTS Immunization with BALB/c splenocytes or heart transplantation induced an expansion and differentiation of I-E-specific B cells into germinal center B cells, whereas BALB/c heart transplantation into sensitized recipients induced the preferential differentiation into antibody-secreting cells. A 10.8-fold increase in the frequency of I-E-specific memory B cells was observed by day 42 postimmunization. Treatment with CTLA4-Ig starting on day 0 or day 7 postimmunization abrogated I-E-specific memory B cell generation and sensitized humoral responses, but not if treatment commenced on day 14. CONCLUSIONS The majority of donor-specific memory B cells are generated between days 7 and 14 postimmunization, thus revealing a flexible timeframe whereby delayed CTLA4-Ig administration can inhibit sensitization and the generation of memory graft-reactive B cells.
Collapse
|
132
|
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
|
133
|
Wekerle T, Segev D, Lechler R, Oberbauer R. Strategies for long-term preservation of kidney graft function. Lancet 2017; 389:2152-2162. [PMID: 28561006 DOI: 10.1016/s0140-6736(17)31283-7] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 12/21/2022]
Abstract
Kidney transplantation has become a routine procedure in the treatment of patients with kidney failure, and requires collaboration of experts from different disciplines, such as nephrology, surgery, immunology, pathology, infectious disease medicine, cardiology, and oncology. Grafts can be obtained from deceased or living donors, with different logistical requirements and implications for long-term graft patency. 1-year graft survival rates are greater than 95% in many centres but improvement of long-term function remains a challenge. New developments in molecular immunology and computational biology have increased precision of donor and recipient matching of HLA and non-HLA compatibility. Individual omics-wide molecular diagnostics, extracorporeal therapies, and drug developments allow for precise individual decision making and treatment. Tolerance induction by mixed chimerism without toxic conditioning and with a low risk of graft versus host disease is a visionary but realistic goal. Some of these innovations are already used in modern transplant centres and will allow advancement in long-term allograft preservation.
Collapse
Affiliation(s)
- Thomas Wekerle
- Department of Surgery, Section of Transplantation Immunology, Medical University of Vienna, Vienna, Austria
| | - Dorry Segev
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Robert Lechler
- MRC Centre for Transplantation, King's College London, London, UK
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
134
|
Diagnostic Contribution of Donor-Specific Antibody Characteristics to Uncover Late Silent Antibody-Mediated Rejection-Results of a Cross-Sectional Screening Study. Transplantation 2017; 101:631-641. [PMID: 27120452 DOI: 10.1097/tp.0000000000001195] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Circulating donor-specific antibodies (DSA) detected on bead arrays may not inevitably indicate ongoing antibody-mediated rejection (AMR). Here, we investigated whether detection of complement-fixation, in parallel to IgG mean fluorescence intensity (MFI), allows for improved prediction of AMR. METHODS Our study included 86 DSA+ kidney transplant recipients subjected to protocol biopsy, who were identified upon cross-sectional antibody screening of 741 recipients with stable graft function at 6 months or longer after transplantation. IgG MFI was analyzed after elimination of prozone effect, and complement-fixation was determined using C1q, C4d, or C3d assays. RESULTS Among DSA+ study patients, 44 recipients (51%) had AMR, 24 of them showing C4d-positive rejection. Although DSA number or HLA class specificity were not different, patients with AMR or C4d + AMR showed significantly higher IgG, C1q, and C3d DSA MFI than nonrejecting or C4d-negative patients, respectively. Overall, the predictive value of DSA characteristics was moderate, whereby the highest accuracy was computed for peak IgG MFI (AMR, 0.73; C4d + AMR, 0.71). Combined analysis of antibody characteristics in multivariate models did not improve AMR prediction. CONCLUSIONS We estimate a 50% prevalence of silent AMR in DSA+ long-term recipients and conclude that assessment of IgG MFI may add predictive accuracy, without an independent diagnostic advantage of detecting complement-fixation.
Collapse
|
135
|
Einecke G, Reeve J, Halloran PF. Hyalinosis Lesions in Renal Transplant Biopsies: Time-Dependent Complexity of Interpretation. Am J Transplant 2017; 17:1346-1357. [PMID: 27873464 DOI: 10.1111/ajt.14136] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/28/2016] [Indexed: 01/25/2023]
Abstract
Because calcineurin inhibitor (CNI) immunosuppressive drugs induce arteriolar hyalinosis (ah) in kidney transplants, ah lesions can potentially provide information about drug exposure. We studied the relationship of ah lesions to findings and outcomes in 562 indication biopsies taken 3 days to 35 years after transplant. Prevalence of ah lesions increased with time of biopsy after transplant (TxBx). The ah scores correlated with arterial intimal thickening and atrophy-fibrosis but, unlike atrophy-fibrosis, did not increase until after 500 days because of a background of ah1 lesions in early biopsies reflecting donor aging. Correlation of ah scores with other features varied with TxBx-in early biopsies, donor age and related changes, and in very late biopsies, chronic antibody-mediated rejection and glomerulonephritis and associated lesions. After correction for TxBx, ah0 in intermediate time periods was associated with increased risk of T cell-mediated rejection and graft loss, probably because of underimmunosuppression and nonadherence. Thus, ah lesions in indication biopsies have multiple associations: donor age (early, usually ah1), chronic glomerular diseases (late, often ah2/3), and adequate exposure to CNIs at intermediate times. This threefold TxBx-dependent complexity must be considered when interpreting indication biopsies: ah lesions often indicate adequate CNI exposure, not toxicity, and unexpected ah0 should increase vigilance for nonadherence and underimmunosuppression.
Collapse
Affiliation(s)
- G Einecke
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - J Reeve
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - P F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada.,Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
136
|
The exciting “bench to bedside” journey of cell therapies for acute kidney injury and renal transplantation. J Nephrol 2017; 30:319-336. [DOI: 10.1007/s40620-017-0384-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/20/2017] [Indexed: 12/15/2022]
|
137
|
Taner T, Park WD, Stegall MD. Unique molecular changes in kidney allografts after simultaneous liver-kidney compared with solitary kidney transplantation. Kidney Int 2017; 91:1193-1202. [PMID: 28233612 DOI: 10.1016/j.kint.2016.12.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 12/13/2022]
Abstract
Kidney allografts transplanted simultaneously with liver allografts from the same donor are known to be immunologically privileged. This is especially evident in recipients with high levels of donor-specific anti-HLA antibodies. Here we investigated the mechanisms of liver's protective impact using gene expression in the kidney allograft. Select solitary kidney transplant or simultaneous liver-kidney transplant recipients were retrospectively reviewed and separated into four groups: 16 cross-match negative kidney transplants, 15 cross-match positive kidney transplants, 12 cross-match negative simultaneous liver-kidney transplants, and nine cross-match-positive simultaneous liver-kidney transplants. Surveillance biopsies of cross-match-positive kidney transplants had increased expression of genes associated with donor-specific antigens, inflammation, and endothelial cell activation compared to cross-match-negative kidney transplants. These changes were not found in cross-match-positive simultaneous liver-kidney transplant biopsies when compared to cross-match-negative simultaneous liver-kidney transplants. In addition, simultaneously transplanting a liver markedly increased renal expression of genes associated with tissue integrity/metabolism, regardless of the cross-match status. While the expression of inflammatory gene sets in cross-match-positive simultaneous liver-kidney transplants was not completely reduced to the level of cross-match-negative kidney transplants, the downstream effects of donor-specific anti-HLA antibodies were blocked. Thus, simultaneous liver-kidney transplants can have a profound impact on the kidney allograft, not only by decreasing inflammation and avoiding endothelial cell activation in cross-match-positive recipients, but also by increasing processes associated with tissue integrity/metabolism by unknown mechanisms.
Collapse
Affiliation(s)
- Timucin Taner
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA.
| | - Walter D Park
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark D Stegall
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
138
|
Menon MC, Murphy B, Heeger PS. Moving Biomarkers toward Clinical Implementation in Kidney Transplantation. J Am Soc Nephrol 2017; 28:735-747. [PMID: 28062570 DOI: 10.1681/asn.2016080858] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Long-term kidney transplant outcomes remain suboptimal, delineating an unmet medical need. Although current immunosuppressive therapy in kidney transplant recipients is effective, dosing is conventionally adjusted empirically on the basis of time after transplant or altered in response to detection of kidney dysfunction, histologic evidence of allograft damage, or infection. Such strategies tend to detect allograft rejection after significant injury has already occurred, fail to detect chronic subclinical inflammation that can negatively affect graft survival, and ignore specific risks and immune mechanisms that differentially contribute to allograft damage among transplant recipients. Assays and biomarkers that reliably quantify and/or predict the risk of allograft injury have the potential to overcome these deficits and thereby, aid clinicians in optimizing immunosuppressive regimens. Herein, we review the data on candidate biomarkers that we contend have the highest potential to become clinically useful surrogates in kidney transplant recipients, including functional T cell assays, urinary gene and protein assays, peripheral blood cell gene expression profiles, and allograft gene expression profiles. We identify barriers to clinical biomarker adoption in the transplant field and suggest strategies for moving biomarker-based individualization of transplant care from a research hypothesis to clinical implementation.
Collapse
Affiliation(s)
- Madhav C Menon
- Renal Division, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Barbara Murphy
- Renal Division, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter S Heeger
- Renal Division, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
139
|
Halloran PF, Famulski KS, Chang J. A Probabilistic Approach to Histologic Diagnosis of Antibody-Mediated Rejection in Kidney Transplant Biopsies. Am J Transplant 2017; 17:129-139. [PMID: 27340822 DOI: 10.1111/ajt.13934] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/25/2023]
Abstract
Histologic diagnosis of antibody-mediated rejection (ABMR) in kidney transplant biopsies uses lesion score cutoffs such as 0 versus >0 rather than actual scores and requires donor-specific antibody (DSA); however, cutoffs lose information, and DSA is not always reliable. Using microarray-derived molecular ABMR scores as a histology-independent estimate of ABMR in 703 biopsies, we reassessed criteria for ABMR to determine relative importance of various lesions, the utility of equations using actual scores rather than cutoffs, and the potential for diagnosing ABMR when DSA is unknown or negative. We confirmed that the important features for ABMR diagnosis were peritubular capillaritis (ptc), glomerulitis (g), glomerular double contours, DSA and C4d staining, but we questioned some features: arterial fibrosis, vasculitis, acute tubular injury, and sum of ptc+g scores. Regression equations using lesion scores predicted molecular ABMR more accurately than score cutoffs (area under the curve 0.85-0.86 vs. 0.75). DSA positivity improved accuracy, but regression equations predicted ABMR with moderate accuracy when DSA was unknown. Some biopsies without detectable DSA had high probability of ABMR by regression, although most had HLA antibody. We concluded that regression equations using lesion scores plus DSA maximized diagnostic accuracy and can estimate probable ABMR when DSA is unknown or undetectable.
Collapse
Affiliation(s)
- P F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Medicine, Division of Nephrology and Transplant Immunology, University of Alberta, Edmonton, AB, Canada
| | - K S Famulski
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - J Chang
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada
| |
Collapse
|
140
|
Acute kidney transplant rejection mediated by angiotensin II type 1 receptor antibodies in a pediatric hyperimmune patient. Pediatr Nephrol 2017; 32:185-188. [PMID: 27752765 DOI: 10.1007/s00467-016-3500-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/21/2016] [Accepted: 09/01/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Several cases of severe antibody-mediated rejection (AMR) secondary to antibodies against the angiotensin II type 1 receptor (AT1R-Ab) have been described with variable outcome. CASE-DIAGNOSIS/TREATMENT We report the case of a 13-year-old boy whose first kidney transplant failed due to steroid-resistant acute cellular rejection, with the subsequent development of sensitization. He received a second kidney transplant which was complicated by early humoral rejection, with weakly positive staining for the complement degradation product C4d. Test results were negative for donor-specific antibodies against human leukocyte antigens (HLA-DSA) and MHC class I-related chain A (MICA) but positive for AT1R-Ab. Retrospective testing of the sera collected during the first kidney transplant was also positive for AT1R-Ab. We therefore hypothesized that the failure of the first transplant was secondary to the same cause. Losartan was immediately introduced into the therapeutic regimen, and the patient showed an excellent clinical and histological recovery. CONCLUSIONS Testing for AT1R-Ab in any hypertensive patient with acute rejection who tests negative or weakly positive for C4d and negative for HLA-DSA and who is refractory to therapy is highly advisable. Pre-transplant AT1R-Ab may be indicative of the outcome in patients whose first transplant failed. Prompt initiation of treatment with losartan-immediately after transplantation in patients with pre-existing AT1R-Ab-should be encouraged.
Collapse
|
141
|
Zhou Y, Li X, Liu Y, Sun Q. Maintenance immunosuppressants in the management of antibody-mediated renal allograft rejection: which regimen is best? Immunotherapy 2016; 9:47-55. [PMID: 28000532 DOI: 10.2217/imt-2016-0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Antibody-mediated rejection (AMR) is a pivotal cause of long-term graft failure following renal transplantation. De novo donor-specific antibody reduction is essential to prevent AMR and improve long-term graft survival in renal transplant recipients. Although the number of early AMR episodes can be successfully controlled by attenuating de novo donor-specific antibodies, the long-term outcomes are unsatisfactory. Numerous studies have focused on new strategies to reverse AMR, but the available evidence suggests that maintenance immunosuppressive agents play important roles. This article reviews data on the use of various maintenance immunosuppressive strategies in the management of AMR, with a focus on antibody-mediated kidney transplant rejection. Its aim is to help provide options benefitting long-term graft survival in renal transplant recipients.
Collapse
Affiliation(s)
- Yiqun Zhou
- Medical Department, Shanghai Roche Pharmaceuticals Ltd, Shanghai 201203, China
| | - Xiaolan Li
- Medical Department, Shanghai Roche Pharmaceuticals Ltd, Shanghai 201203, China
| | - Yun Liu
- Medical Department, Shanghai Roche Pharmaceuticals Ltd, Shanghai 201203, China
| | - Qiquan Sun
- Department of Renal Transplantation, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| |
Collapse
|
142
|
Vanhove T, Vermeulen T, Annaert P, Lerut E, Kuypers DRJ. High Intrapatient Variability of Tacrolimus Concentrations Predicts Accelerated Progression of Chronic Histologic Lesions in Renal Recipients. Am J Transplant 2016; 16:2954-2963. [PMID: 27013142 DOI: 10.1111/ajt.13803] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/17/2016] [Accepted: 03/19/2016] [Indexed: 01/25/2023]
Abstract
High intrapatient variability (IPV) of tacrolimus concentrations is increasingly recognized as a predictor of poor outcome in solid organ recipients. How it relates to evolution of histology has not been explored. We analyzed tacrolimus IPV using the coefficient of variability (CV) from months 6-12 after transplantation in a cohort of 220 renal recipients for whom paired protocol biopsies at 3 mo and 2 years were available. Recipients in the highest CV tertile had an increased risk of moderate to severe fibrosis and tubular atrophy by 2 years compared with the low-IPV tertile (odds ratio [OR] 2.47, 95% confidence interval [CI] 1.09-5.60, p = 0.031; and OR 2.40, 95% CI 1.03-5.60, p = 0.043, respectively). Other predictors were donor age, severity of chronic lesions at 3 mo, and presence of borderline or subclinical rejection at 3 mo. Chronicity score increased significantly more in the high CV tertile group than in the middle and low tertiles (mean increase 1.97 ± 2.03 vs. 1.18 ± 2.44 and 1.12 ± 1.80, respectively; p < 0.05). CV did not predict evolution of renal function, which did not deteriorate within the 2-year follow-up period. These results indicate that high IPV is related to accelerated progression of chronic histologic lesions before any evidence of renal dysfunction.
Collapse
Affiliation(s)
- T Vanhove
- Department of Microbiology and Immunology, KU Leuven - University of Leuven, and Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - T Vermeulen
- Department of Microbiology and Immunology, KU Leuven - University of Leuven, and Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - P Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - E Lerut
- Department of Imaging and Pathology, KU Leuven - University of Leuven, and Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - D R J Kuypers
- Department of Microbiology and Immunology, KU Leuven - University of Leuven, and Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
143
|
Moscarelli L, Antognoli G, Buti E, Dervishi E, Fani F, Caroti L, Tsalouchos A, Romoli E, Ghiandai G, Minetti E. 1,25 Dihydroxyvitamin D circulating levels, calcitriol administration, and incidence of acute rejection, CMV infection, and polyoma virus infection in renal transplant recipients. Clin Transplant 2016; 30:1347-1359. [PMID: 27532453 DOI: 10.1111/ctr.12829] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2016] [Indexed: 02/01/2023]
Abstract
Observation that 1,25-Dihydroxyvitamin-D3 has an immunomodulatory effect on innate and adaptive immunity raises the possible effect on clinical graft outcome. Aim of this study was to evaluate the correlation of biopsy-proven acute rejection, CMV infection, BKV infection, with 1,25-Dihydroxyvitamin-D3 deficiency and the benefit of calcitriol supplementation before and during the transplantation. Risk factors and kidney graft function were also evaluated. All RTRs received induction therapy with basiliximab, cyclosporine, mycophenolic acid, and steroids. During the first year, the incidence of BPAR (4% vs 11%, P=.04), CMV infection (3% vs 9%, P=.04), and BKV infection (6% vs 19%, P=.04) was significantly lower in users compared to controls. By multivariate Cox regression analysis, 1,25-Dihydroxyvitamin-D3 deficiency and no calcitriol exposure were independent risk factors for BPAR (HR=4.30, P<.005 and HR=3.25, P<.05), for CMV infection (HR=2.33, P<.05 and HR=2.31, P=.001), and for BKV infection (HR=2.41, P<.05 and HR=2.45, P=.001). After one year, users had a better renal function: eGFR was 62.5±6.7 mL/min vs 51.4±7.6 mL/min (P<.05). Only one user developed polyomavirus-associated nephropathy vs 15 controls. Two users lost their graft vs 11 controls. 1,25(OH)2-D3 deficiency circulating levels increased the risk of BPAR, CMV infection, BKV infection after kidney transplantation. Administration of calcitriol is a way to obtain adequate 1,25(OH)2-D3 circulating levels.
Collapse
Affiliation(s)
| | | | - Elisa Buti
- Renal Unit, Careggi University Hospital, Florence, Italy
| | | | - Filippo Fani
- Renal Unit, Careggi University Hospital, Florence, Italy
| | | | | | - Elena Romoli
- Renal Unit, Careggi University Hospital, Florence, Italy
| | | | - Enrico Minetti
- Renal Unit, Careggi University Hospital, Florence, Italy
| |
Collapse
|
144
|
Chand S, Atkinson D, Collins C, Briggs D, Ball S, Sharif A, Skordilis K, Vydianath B, Neil D, Borrows R. The Spectrum of Renal Allograft Failure. PLoS One 2016; 11:e0162278. [PMID: 27649571 PMCID: PMC5029903 DOI: 10.1371/journal.pone.0162278] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/21/2016] [Indexed: 01/15/2023] Open
Abstract
Background Causes of “true” late kidney allograft failure remain unclear as study selection bias and limited follow-up risk incomplete representation of the spectrum. Methods We evaluated all unselected graft failures from 2008–2014 (n = 171; 0–36 years post-transplantation) by contemporary classification of indication biopsies “proximate” to failure, DSA assessment, clinical and biochemical data. Results The spectrum of graft failure changed markedly depending on the timing of allograft failure. Failures within the first year were most commonly attributed to technical failure, acute rejection (with T-cell mediated rejection [TCMR] dominating antibody-mediated rejection [ABMR]). Failures beyond a year were increasingly dominated by ABMR and ‘interstitial fibrosis with tubular atrophy’ without rejection, infection or recurrent disease (“IFTA”). Cases of IFTA associated with inflammation in non-scarred areas (compared with no inflammation or inflammation solely within scarred regions) were more commonly associated with episodes of prior rejection, late rejection and nonadherence, pointing to an alloimmune aetiology. Nonadherence and late rejection were common in ABMR and TCMR, particularly Acute Active ABMR. Acute Active ABMR and nonadherence were associated with younger age, faster functional decline, and less hyalinosis on biopsy. Chronic and Chronic Active ABMR were more commonly associated with Class II DSA. C1q-binding DSA, detected in 33% of ABMR episodes, were associated with shorter time to graft failure. Most non-biopsied patients were DSA-negative (16/21; 76.1%). Finally, twelve losses to recurrent disease were seen (16%). Conclusion This data from an unselected population identifies IFTA alongside ABMR as a very important cause of true late graft failure, with nonadherence-associated TCMR as a phenomenon in some patients. It highlights clinical and immunological characteristics of ABMR subgroups, and should inform clinical practice and individualised patient care.
Collapse
Affiliation(s)
- Sourabh Chand
- Department of Nephrology and Kidney Transplantation, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, United Kingdom
- Renal Department, Royal Shrewsbury Hospital, Shropshire, United Kingdom
- * E-mail:
| | - David Atkinson
- Histocompatibility and Immunogenetics Laboratory, NHSBT Birmingham Centre, Vincent Drive, Edgbaston, Birmingham, United Kingdom
| | - Clare Collins
- Histocompatibility and Immunogenetics Laboratory, NHSBT Birmingham Centre, Vincent Drive, Edgbaston, Birmingham, United Kingdom
| | - David Briggs
- Histocompatibility and Immunogenetics Laboratory, NHSBT Birmingham Centre, Vincent Drive, Edgbaston, Birmingham, United Kingdom
| | - Simon Ball
- Department of Nephrology and Kidney Transplantation, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, United Kingdom
| | - Adnan Sharif
- Department of Nephrology and Kidney Transplantation, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, United Kingdom
| | - Kassiani Skordilis
- Department of Renal Histopathology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Bindu Vydianath
- Department of Renal Histopathology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Desley Neil
- Department of Renal Histopathology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Richard Borrows
- Department of Nephrology and Kidney Transplantation, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
145
|
Young JS, Chen J, Miller ML, Vu V, Tian C, Moon JJ, Alegre ML, Sciammas R, Chong AS. Delayed Cytotoxic T Lymphocyte-Associated Protein 4-Immunoglobulin Treatment Reverses Ongoing Alloantibody Responses and Rescues Allografts From Acute Rejection. Am J Transplant 2016; 16:2312-23. [PMID: 26928966 PMCID: PMC4956497 DOI: 10.1111/ajt.13761] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 02/18/2016] [Accepted: 02/21/2016] [Indexed: 01/25/2023]
Abstract
Antibody-mediated rejection has emerged as the leading cause of late graft loss in kidney transplant recipients, and inhibition of donor-specific antibody production should lead to improved transplant outcomes. The fusion protein cytotoxic T lymphocyte-associated protein 4-immunoglobulin (CTLA4-Ig) blocks T cell activation and consequently inhibits T-dependent B cell antibody production, and the current paradigm is that CTLA4-Ig is effective with naïve T cells and less so with activated or memory T cells. In this study, we used a mouse model of allosensitization to investigate the efficacy of continuous CTLA4-Ig treatment, initiated 7 or 14 days after sensitization, for inhibiting ongoing allospecific B cell responses. Delayed treatment with CTLA4-Ig collapsed the allospecific germinal center B cell response and inhibited alloantibody production. Using adoptively transferred T cell receptor transgenic T cells and a novel approach to track endogenous graft-specific T cells, we demonstrate that delayed CTLA4-Ig minimally inhibited graft-specific CD4(+) and T follicular helper responses. Remarkably, delaying CTLA4-Ig until day 6 after transplantation in a fully mismatched heart transplant model inhibited alloantibody production and prevented acute rejection, whereas transferred hyperimmune sera reversed the effects of delayed CTLA4-Ig. Collectively, our studies revealed the unexpected efficacy of CTLA4-Ig for inhibiting ongoing B cell responses even when the graft-specific T cell response was robustly established.
Collapse
Affiliation(s)
- James S. Young
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL
| | - Jianjun Chen
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL
| | - Michelle L. Miller
- Section of Rheumatology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Vinh Vu
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL
| | - Changtai Tian
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL
| | - James J. Moon
- Center for Immunology and Inflammatory Diseases, and Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital; and Harvard Medical School, Boston, MA
| | - Maria-Luisa Alegre
- Section of Rheumatology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Roger Sciammas
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL
| | - Anita S. Chong
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL
| |
Collapse
|
146
|
Modena BD, Kurian SM, Gaber LW, Waalen J, Su AI, Gelbart T, Mondala TS, Head SR, Papp S, Heilman R, Friedewald JJ, Flechner S, Marsh CL, Sung RS, Shidban H, Chan L, Abecassis MM, Salomon DR. Gene Expression in Biopsies of Acute Rejection and Interstitial Fibrosis/Tubular Atrophy Reveals Highly Shared Mechanisms That Correlate With Worse Long-Term Outcomes. Am J Transplant 2016; 16:1982-98. [PMID: 26990570 PMCID: PMC5501990 DOI: 10.1111/ajt.13728] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 01/08/2016] [Accepted: 01/13/2016] [Indexed: 01/25/2023]
Abstract
Interstitial fibrosis and tubular atrophy (IFTA) is found in approximately 25% of 1-year biopsies posttransplant. It is known that IFTA correlates with decreased graft survival when histological evidence of inflammation is present. Identifying the mechanistic etiology of IFTA is important to understanding why long-term graft survival has not changed as expected despite improved immunosuppression and dramatically reduced rates of clinical acute rejection (AR) (Services UDoHaH. http://www.ustransplant.org/annual_reports/current/509a_ki.htm). Gene expression profiles of 234 graft biopsy samples were obtained with matching clinical and outcome data. Eighty-one IFTA biopsies were divided into subphenotypes by degree of histological inflammation: IFTA with AR, IFTA with inflammation, and IFTA without inflammation. Samples with AR (n = 54) and normally functioning transplants (TX; n = 99) were used in comparisons. A novel analysis using gene coexpression networks revealed that all IFTA phenotypes were strongly enriched for dysregulated gene pathways and these were shared with the biopsy profiles of AR, including IFTA samples without histological evidence of inflammation. Thus, by molecular profiling we demonstrate that most IFTA samples have ongoing immune-mediated injury or chronic rejection that is more sensitively detected by gene expression profiling. These molecular biopsy profiles correlated with future graft loss in IFTA samples without inflammation.
Collapse
Affiliation(s)
- B. D. Modena
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - S. M. Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA,DNA Microarray and Next Generation Sequencing Core, The Scripps Research Institute, La Jolla, CA
| | - L. W. Gaber
- Department of Pathology, The Methodist Hospital, Houston, TX
| | - J. Waalen
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - A. I. Su
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - T. Gelbart
- DNA Microarray and Next Generation Sequencing Core, The Scripps Research Institute, La Jolla, CA
| | - T. S. Mondala
- DNA Microarray and Next Generation Sequencing Core, The Scripps Research Institute, La Jolla, CA
| | - S. R. Head
- DNA Microarray and Next Generation Sequencing Core, The Scripps Research Institute, La Jolla, CA
| | - S. Papp
- DNA Microarray and Next Generation Sequencing Core, The Scripps Research Institute, La Jolla, CA
| | - R. Heilman
- Transplant Genomics Collaborative Group (TGCG), La Jolla, CA,Department of Transplant Nephrology, Mayo Clinic, Phoenix, AZ
| | - J. J. Friedewald
- Northwestern Comprehensive Transplant Center, Northwestern University, Chicago, IL
| | - S.M. Flechner
- Transplant Genomics Collaborative Group (TGCG), La Jolla, CA,Glickman Urology and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - C. L. Marsh
- Transplant Genomics Collaborative Group (TGCG), La Jolla, CA,Scripps Center for Organ and Cell Transplantation, Scripps Health, La Jolla, CA
| | - R. S. Sung
- Transplant Genomics Collaborative Group (TGCG), La Jolla, CA,Section of Transplant Surgery, University of Michigan, Ann Arbor, MI
| | - H. Shidban
- Transplant Genomics Collaborative Group (TGCG), La Jolla, CA,Department of Surgery, St Vincent Medical Center, Los Angeles, CA
| | - L. Chan
- Transplant Genomics Collaborative Group (TGCG), La Jolla, CA,Department of Transplant/Nephrology, University of Colorado, Aurora, CO
| | - M. M. Abecassis
- Northwestern Comprehensive Transplant Center, Northwestern University, Chicago, IL
| | - D. R. Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA,DNA Microarray and Next Generation Sequencing Core, The Scripps Research Institute, La Jolla, CA,Transplant Genomics Collaborative Group (TGCG), La Jolla, CA,Corresponding author: Daniel R. Salomon,
| |
Collapse
|
147
|
Sanchez-Fueyo A, Markmann JF. Immune Exhaustion and Transplantation. Am J Transplant 2016; 16:1953-7. [PMID: 26729653 DOI: 10.1111/ajt.13702] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 11/09/2015] [Accepted: 12/13/2015] [Indexed: 01/25/2023]
Abstract
Exhaustion of lymphocyte function through chronic exposure to a high load of foreign antigen is well established for chronic viral infection and antitumor immunity and has been found to be associated with a distinct molecular program and characteristic cell surface phenotype. Although exhaustion has most commonly been studied in the context of CD8 viral responses, recent studies indicate that chronic antigen exposure may affect B cells, NK cells and CD4 T cells in a parallel manner. Limited information is available regarding the extent of lymphocyte exhaustion development in the transplant setting and its impact on anti-graft alloreactivity. By analogy to the persistence of a foreign virus, the large mass of alloantigen presented by an allograft in chronic residence could provide an ideal setting for exhausting donor-reactive T cells. The extent of T cell exhaustion occurring with various allografts, the kinetics of its development, whether exhaustion is influenced positively or negatively by different immunosuppressants, and the impact of exhaustion on graft survival and tolerance development remains a fertile area for investigation. Harnessing or encouraging the natural processes of exhaustion may provide a novel means to promote graft survival and transplantation tolerance.
Collapse
Affiliation(s)
- A Sanchez-Fueyo
- Institute of Liver Studies, Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London University, King's College Hospital, London, UK
| | - J F Markmann
- Division of Transplant Surgery, Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
148
|
Halloran PF, Famulski KS, Reeve J. Molecular assessment of disease states in kidney transplant biopsy samples. Nat Rev Nephrol 2016; 12:534-48. [DOI: 10.1038/nrneph.2016.85] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
149
|
Mechanisms of antibody-mediated acute and chronic rejection of kidney allografts. Curr Opin Organ Transplant 2016; 21:7-14. [PMID: 26575854 DOI: 10.1097/mot.0000000000000262] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Antibody-mediated rejection is responsible for up to half of acute rejection episodes in kidney transplant patients and more than half of late graft failures. Antibodies cause acute graft abnormalities that are distinct from T cell-mediated rejection and at later times posttransplant, a distinct pathologic lesion is associated with capillary basement membrane multilayering and glomerulopathy. Despite the importance of donor-reactive antibodies as the leading cause of kidney graft failure, mechanisms underlying antibody-mediated acute and chronic kidney graft injury are poorly understood. Here, we review recent insights provided from clinical studies as well as from animal models that may help to identify new targets for therapy. RECENT FINDINGS Studies of biopsies from kidney grafts in patients with donor-specific antibody versus those without have utilized analysis of pathologic lesions and gene expression to identify the distinct characteristics of antibody-mediated rejection. These analyses have indicated the presence of natural killer cells and their activation during antibody-mediated rejection. The impact of studies of antibody-mediated allograft injury in animal models have lagged behind these clinical studies, but have been useful in testing the activation of innate immune components within allografts in the presence of donor-specific antibodies. SUMMARY Most insights into processes of antibody-mediated rejection of kidney grafts have come from carefully designed clinical studies. However, several new mouse models of antibody-mediated kidney allograft rejection may replicate the abnormalities observed in clinical kidney grafts and may be useful in directly testing mechanisms that underlie acute and chronic antibody-mediated graft injury.
Collapse
|
150
|
Natural killer cells play a critical role in mediating inflammation and graft failure during antibody-mediated rejection of kidney allografts. Kidney Int 2016; 89:1293-306. [PMID: 27165816 DOI: 10.1016/j.kint.2016.02.030] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/11/2016] [Accepted: 02/04/2016] [Indexed: 12/14/2022]
Abstract
While the incidence of antibody-mediated kidney graft rejection has increased, the key cellular and molecular participants underlying this graft injury remain unclear. Rejection of kidney allografts in mice lacking the chemokine receptor CCR5 is dependent on production of donor-specific antibody. Here we determine if cells expressing cytotoxic function contributed to antibody-mediated kidney allograft rejection in these recipients. Wild-type C57BL/6, B6.CCR5(-/-), and B6.CD8(-/-)/CCR5(-/-) mice were transplanted with complete MHC-mismatched A/J kidney grafts, and intragraft inflammatory components were followed to rejection. B6.CCR5(-/-) and B6.CD8(-/-)/CCR5(-/-) recipients rejected kidney allografts by day 35, whereas 65% of allografts in wild-type recipients survived past day 80 post-transplant. Rejected allografts in wild-type C57BL/6, B6.CCR5(-/-), and B6.CD8(-/-)/CCR5(-/-) recipients expressed high levels of VCAM-1 and MMP7 mRNA that was associated with high serum titers of donor-specific antibody. High levels of perforin and granzyme B mRNA expression peaked on day 6 post-transplant in allografts in all recipients, but were absent in isografts. Depletion of natural killer cells in B6.CD8(-/-)/CCR5(-/-) recipients reduced this expression to background levels and promoted the long-term survival of 40% of the kidney allografts. Thus, natural killer cells have a role in increased inflammation during antibody-mediated kidney allograft injury and in rejection of the grafts.
Collapse
|