1
|
Bane O, Lewis SC, Lim RP, Carney BW, Shah A, Fananapazir G. Contemporary and Emerging MRI Strategies for Assessing Kidney Allograft Complications: Arterial Stenosis and Parenchymal Injury, From the AJR Special Series on Imaging of Fibrosis. AJR Am J Roentgenol 2024; 222:e2329418. [PMID: 37315018 PMCID: PMC11006565 DOI: 10.2214/ajr.23.29418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
MRI plays an important role in the evaluation of kidney allografts for vascular complications as well as parenchymal insults. Transplant renal artery stenosis, the most common vascular complication of kidney transplant, can be evaluated by MRA using gadolinium and nongadolinium contrast agents as well as by unenhanced MRA techniques. Parenchymal injury occurs through a variety of pathways, including graft rejection, acute tubular injury, BK polyomavirus infection, drug-induced interstitial nephritis, and pyelonephritis. Investigational MRI techniques have sought to differentiate among these causes of dysfunction as well as to assess the degree of interstitial fibrosis or tubular atrophy (IFTA)-the common end pathway for all of these processes-which is currently evaluated by invasively obtained core biopsies. Some of these MRI sequences have shown promise in not only assessing the cause of parenchymal injury but also assessing IFTA noninvasively. This review describes current clinically used MRI techniques and previews promising investigational MRI techniques for assessing complications of kidney grafts.
Collapse
Affiliation(s)
- Octavia Bane
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sara C Lewis
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ruth P Lim
- Department of Radiology and Department of Surgery, University of Melbourne, Austin Health, Melbourne, Australia
| | - Benjamin W Carney
- Department of Radiology, University of California Davis Medical Center, 4860 Y St, Ste 3100, Sacramento, CA 95816
| | - Amar Shah
- Department of Radiology, Mayo Clinic Arizona, Phoenix, AZ
| | - Ghaneh Fananapazir
- Department of Radiology, University of California Davis Medical Center, 4860 Y St, Ste 3100, Sacramento, CA 95816
| |
Collapse
|
2
|
Khan MAH, Nolan B, Stavniichuk A, Merk D, Imig JD. Dual soluble epoxide hydrolase inhibitor - farnesoid X receptor agonist interventional treatment attenuates renal inflammation and fibrosis. Front Immunol 2024; 14:1269261. [PMID: 38235144 PMCID: PMC10791967 DOI: 10.3389/fimmu.2023.1269261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
Introduction Renal fibrosis associated with inflammation is a critical pathophysiological event in chronic kidney disease (CKD). We have developed DM509 which acts concurrently as a farnesoid X receptor agonist and a soluble epoxide hydrolase inhibitor and investigated DM509 efficacy as an interventional treatment using the unilateral ureteral obstruction (UUO) mouse model. Methods Male mice went through either UUO or sham surgery. Interventional DM509 treatment (10mg/kg/d) was started three days after UUO induction and continued for 7 days. Plasma and kidney tissue were collected at the end of the experimental protocol. Results UUO mice demonstrated marked renal fibrosis with higher kidney hydroxyproline content and collagen positive area. Interventional DM509 treatment reduced hydroxyproline content by 41% and collagen positive area by 65%. Renal inflammation was evident in UUO mice with elevated MCP-1, CD45-positive immune cell positive infiltration, and profibrotic inflammatory gene expression. DM509 treatment reduced renal inflammation in UUO mice. Renal fibrosis in UUO was associated with epithelial-to-mesenchymal transition (EMT) and DM509 treatment reduced EMT. UUO mice also had tubular epithelial barrier injury with increased renal KIM-1, NGAL expression. DM509 reduced tubular injury markers by 25-50% and maintained tubular epithelial integrity in UUO mice. Vascular inflammation was evident in UUO mice with 9 to 20-fold higher ICAM and VCAM gene expression which was reduced by 40-50% with DM509 treatment. Peritubular vascular density was reduced by 35% in UUO mice and DM509 prevented vascular loss. Discussion Interventional treatment with DM509 reduced renal fibrosis and inflammation in UUO mice demonstrating that DM509 is a promising drug that combats renal epithelial and vascular pathological events associated with progression of CKD.
Collapse
Affiliation(s)
- Md. Abdul Hye Khan
- Drug Discovery Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Benjamin Nolan
- Drug Discovery Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anna Stavniichuk
- Drug Discovery Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians Universität München, Munich, Germany
| | - John D. Imig
- Drug Discovery Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
3
|
McDaniels JM, Shetty AC, Kuscu C, Kuscu C, Bardhi E, Rousselle T, Drachenberg C, Talwar M, Eason JD, Muthukumar T, Maluf DG, Mas VR. Single nuclei transcriptomics delineates complex immune and kidney cell interactions contributing to kidney allograft fibrosis. Kidney Int 2023; 103:1077-1092. [PMID: 36863444 PMCID: PMC10200746 DOI: 10.1016/j.kint.2023.02.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 01/16/2023] [Accepted: 02/07/2023] [Indexed: 03/04/2023]
Abstract
Chronic allograft dysfunction (CAD), characterized histologically by interstitial fibrosis and tubular atrophy, is the major cause of kidney allograft loss. Here, using single nuclei RNA sequencing and transcriptome analysis, we identified the origin, functional heterogeneity, and regulation of fibrosis-forming cells in kidney allografts with CAD. A robust technique was used to isolate individual nuclei from kidney allograft biopsies and successfully profiled 23,980 nuclei from five kidney transplant recipients with CAD and 17,913 nuclei from three patients with normal allograft function. Our analysis revealed two distinct states of fibrosis in CAD; low and high extracellular matrix (ECM) with distinct kidney cell subclusters, immune cell types, and transcriptional profiles. Imaging mass cytometry analysis confirmed increased ECM deposition at the protein level. Proximal tubular cells transitioned to an injured mixed tubular (MT1) phenotype comprised of activated fibroblasts and myofibroblast markers, generated provisional ECM which recruited inflammatory cells, and served as the main driver of fibrosis. MT1 cells in the high ECM state achieved replicative repair evidenced by dedifferentiation and nephrogenic transcriptional signatures. MT1 in the low ECM state showed decreased apoptosis, decreased cycling tubular cells, and severe metabolic dysfunction, limiting the potential for repair. Activated B, T and plasma cells were increased in the high ECM state, while macrophage subtypes were increased in the low ECM state. Intercellular communication between kidney parenchymal cells and donor-derived macrophages, detected several years post-transplantation, played a key role in injury propagation. Thus, our study identified novel molecular targets for interventions aimed to ameliorate or prevent allograft fibrogenesis in kidney transplant recipients.
Collapse
Affiliation(s)
- Jennifer M McDaniels
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cem Kuscu
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Canan Kuscu
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Elissa Bardhi
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Thomas Rousselle
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cinthia Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Manish Talwar
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James D Eason
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Daniel G Maluf
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; Program in Transplantation, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Valeria R Mas
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
4
|
McDaniels JM, Shetty AC, Rousselle TV, Bardhi E, Maluf DG, Mas VR. The cellular landscape of the normal kidney allograft: Main players balancing the alloimmune response. FRONTIERS IN TRANSPLANTATION 2022; 1:988238. [PMID: 38994377 PMCID: PMC11235379 DOI: 10.3389/frtra.2022.988238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/22/2022] [Indexed: 07/13/2024]
Abstract
Despite recent advances made in short-term outcomes; minimal improvements have been observed in long-term kidney transplantation outcomes. Due to an imbalance between organ transplant availability and patient waiting list, expanding kidney allograft longevity is a critical need in the field. Prior studies have either focused on early ischemic and immunological conditions affecting kidney allografts (e.g., delayed graft function, acute rejection) or late stage chronic injury when interventions are no longer feasible. However, studies characterizing kidney allografts with normal function by its cellular distribution, cell-cell interactions, and associated molecular pathways are lacking. Herein, we used single nuclei RNA-sequencing to uncover the cellular landscape and transcriptome of the normal kidney allograft. We profiled 40,950 nuclei from seven human kidney biopsies (normal native, N = 3; normal allograft, N = 4); normal allograft protocol biopsies were collected ≥15-months post-transplant. A total of 17 distinct cell clusters were identified with proximal tubules (25.70 and 21.01%), distal tubules (15.22 and 18.20%), and endothelial cells (EC) (4.26 and 9.94%) constituting the major cell populations of normal native and normal allograft kidneys, respectively. A large proportion of cycling cells from normal native kidneys were in G1-phase (43.96%) whereas cells from normal allograft were predominantly in S-phase (32.69%). This result suggests that transcriptional differences between normal native and normal allograft biopsies are dependent on the new host environment, immunosuppression, and injury-affliction. In the normal allograft, EC-specific genes upregulated metabolism, the immune response, and cellular growth, emphasizing their role in maintaining homeostasis during the ongoing alloreactive stress response. Immune cells, including B (2.81%), macrophages (24.96%), monocytes (15.29%), natural killer (NK) (12.83%), neutrophils (8.44%), and T cells (14.41%, were increased in normal allografts despite lack of histological or clinical evidence of acute rejection. Phenotypic characterization of immune cell markers supported lymphocyte activation and proinflammatory cytokines signaling pathways (i.e., IL-15, IL-32). The activation of B, NK, and T cells reveals potential immune cells underlying subclinical inflammation and repair. These single nuclei analyses provide novel insights into kidney and immune cell associated signaling pathways that portray kidney grafts with normal allograft function beyond 2-years post-transplant, revealing a novel perspective in understanding long-term allograft graft survival.
Collapse
Affiliation(s)
- Jennifer M McDaniels
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| | - Amol C Shetty
- Institute for Genome Sciences, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Thomas V Rousselle
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| | - Elissa Bardhi
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| | - Daniel G Maluf
- Program in Transplantation, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Valeria R Mas
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
5
|
Hydroxychloroquine alleviates renal interstitial fibrosis by inhibiting the PI3K/Akt signaling pathway. Biochem Biophys Res Commun 2022; 610:154-161. [DOI: 10.1016/j.bbrc.2022.04.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/18/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023]
|
6
|
Beunon P, Barat M, Dohan A, Cheddani L, Males L, Fernandez P, Etain B, Bellivier F, Marlinge E, Vrtovsnik F, Vidal-Petiot E, Khalil A, Haymann JP, Flamant M, Tabibzadeh N. MRI-based kidney radiomic analysis during chronic lithium treatment. Eur J Clin Invest 2022; 52:e13756. [PMID: 35104368 DOI: 10.1111/eci.13756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/11/2022] [Accepted: 01/23/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Lithium therapy during bipolar disorder is associated with an increased risk of chronic kidney disease (CKD) that is slowly progressive and undetectable at early stages. We aimed at identifying kidney image texture features as possible imaging biomarkers of decreased measured glomerular filtration rate (mGFR) using radiomics of T2-weighted magnetic resonance imaging (MRI). METHODS One hundred and eight patients treated with lithium were evaluated including mGFR and kidney MRI, with T2-weighted sequence single-shot fast spin-echo. Computed radiomic analysis was performed after kidney segmentation. Significant features were selected to build a radiomic signature using multivariable Cox analysis to detect an mGFR <60 ml/min/1.73 m². The texture index was validated using a training and a validation cohort. RESULTS Texture analysis index was able to detect an mGFR decrease, with an AUC of 0.85 in the training cohort and 0.71 in the validation cohort. Patients with a texture index below the median were older (59 [42-66] vs. 46 [34-54] years, p = .001), with longer treatment duration (10 [3-22] vs. 6 [2-10] years, p = .02) and a lower mGFR (66 [46-84] vs. 83 [71-94] ml/min/1.73m², p < .001). Texture analysis index was independently and negatively associated with age (β = -.004 ± 0.001, p < .001), serum vasopressin (-0.005 ± 0.002, p = .02) and lithium treatment duration (-0.01 ± 0.003, p = .001), with a significant interaction between lithium treatment duration and mGFR (p = .02). CONCLUSIONS A renal texture index was developed among patients treated with lithium associated with a decreased mGFR. This index might be relevant in the diagnosis of lithium-induced renal toxicity.
Collapse
Affiliation(s)
- Paul Beunon
- Sorbonne Université, Paris, France.,Radiologie A, APHP.Centre Hôpital Cochin, Paris, France
| | - Maxime Barat
- Radiologie A, APHP.Centre Hôpital Cochin, Paris, France.,Université de Paris, Paris, France
| | - Anthony Dohan
- Radiologie A, APHP.Centre Hôpital Cochin, Paris, France.,Université de Paris, Paris, France
| | - Lynda Cheddani
- Université Paris Saclay, INSERM U1018, Equipe 5, CESP (Centre de Recherche en Épidémiologie et Santé des Populations), Paris, France.,Nephrologie, APHP Hôpital Ambroise Paré, Paris, France
| | - Lisa Males
- Université de Paris, Paris, France.,Radiologie, APHP.Nord Hôpital Bichat, Paris, France
| | | | - Bruno Etain
- Université de Paris, Paris, France.,Département de Psychiatrie et de Médecine Addictologique, APHP.Nord, GH Lariboisière-Fernand-Widal, DMU Neurosciences, Paris, France
| | - Frank Bellivier
- Université de Paris, Paris, France.,Département de Psychiatrie et de Médecine Addictologique, APHP.Nord, GH Lariboisière-Fernand-Widal, DMU Neurosciences, Paris, France
| | - Emeline Marlinge
- Département de Psychiatrie et de Médecine Addictologique, APHP.Nord, GH Lariboisière-Fernand-Widal, DMU Neurosciences, Paris, France
| | - François Vrtovsnik
- Université de Paris, Paris, France.,Département de Psychiatrie et de Médecine Addictologique, APHP.Nord, GH Lariboisière-Fernand-Widal, DMU Neurosciences, Paris, France.,Néphrologie, APHP.Nord Hôpital Bichat, Paris, France
| | - Emmanuelle Vidal-Petiot
- Université de Paris, Paris, France.,Département de Psychiatrie et de Médecine Addictologique, APHP.Nord, GH Lariboisière-Fernand-Widal, DMU Neurosciences, Paris, France.,Explorations Fonctionnelles, Physiologie, APHP.Nord Hôpital Bichat, Paris, France
| | - Antoine Khalil
- Université de Paris, Paris, France.,Radiologie, APHP.Nord Hôpital Bichat, Paris, France
| | - Jean-Philippe Haymann
- Sorbonne Université, Paris, France.,Explorations Fonctionnelles et laboratoire de la lithiase, APHP. Sorbonne Hôpital Tenon, Paris, France
| | - Martin Flamant
- Université de Paris, Paris, France.,Département de Psychiatrie et de Médecine Addictologique, APHP.Nord, GH Lariboisière-Fernand-Widal, DMU Neurosciences, Paris, France.,Explorations Fonctionnelles, Physiologie, APHP.Nord Hôpital Bichat, Paris, France
| | - Nahid Tabibzadeh
- Explorations Fonctionnelles, Physiologie, APHP.Nord Hôpital Bichat, Paris, France.,Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL 8228-Unité Métabolisme et Physiologie Rénale, Paris, France
| |
Collapse
|
7
|
Mitochondrial Pathophysiology on Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms23031776. [PMID: 35163697 PMCID: PMC8836100 DOI: 10.3390/ijms23031776] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
In healthy kidneys, interstitial fibroblasts are responsible for the maintenance of renal architecture. Progressive interstitial fibrosis is thought to be a common pathway for chronic kidney diseases (CKD). Diabetes is one of the boosters of CKD. There is no effective treatment to improve kidney function in CKD patients. The kidney is a highly demanding organ, rich in redox reactions occurring in mitochondria, making it particularly vulnerable to oxidative stress (OS). A dysregulation in OS leads to an impairment of the Electron transport chain (ETC). Gene deficiencies in the ETC are closely related to the development of kidney disease, providing evidence that mitochondria integrity is a key player in the early detection of CKD. The development of novel CKD therapies is needed since current methods of treatment are ineffective. Antioxidant targeted therapies and metabolic approaches revealed promising results to delay the progression of some markers associated with kidney disease. Herein, we discuss the role and possible origin of fibroblasts and the possible potentiators of CKD. We will focus on the important features of mitochondria in renal cell function and discuss their role in kidney disease progression. We also discuss the potential of antioxidants and pharmacologic agents to delay kidney disease progression.
Collapse
|
8
|
Bobot M, Hache G, Moyon A, Fernandez S, Balasse L, Daniel L, Garrigue P, Brige P, Chopinet S, Dignat-George F, Brunet P, Burtey S, Guillet B. Renal SPECT/CT with 99mTc-dimercaptosuccinic acid is a non-invasive predictive marker for the development of interstitial fibrosis in a rat model of renal insufficiency. Nephrol Dial Transplant 2021; 36:804-810. [PMID: 33367913 DOI: 10.1093/ndt/gfaa374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) increases cardiovascular risk and mortality. Renal fibrosis plays a major role in the progression of CKD but, to date, histology remains the gold standard to assess fibrosis. Non-invasive techniques are needed to assess renal parenchymal impairment and to perform the longitudinal evaluation of renal structure. Thus we evaluated renal isotopic imaging by single-photon emission computed tomography/computed tomography (SPECT/CT) with technetium-99m (99mTc)-dimercaptosuccinic acid (DMSA) to monitor renal impairment during renal insufficiency in rats. METHODS Renal insufficiency was induced by an adenine-rich diet (ARD) at 0.25 and 0.5% for 28 days. Renal dysfunction was evaluated by assaying biochemical markers and renal histology. Renal parenchymal impairment was assessed by SPECT/CT isotopic imaging with 99mTc-DMSA on Days 0, 7, 14, 21, 28, 35 and 49. RESULTS Compared with controls, ARD rats developed renal dysfunction characterized by increased serum creatinine and blood urea nitrogen, fibrosis and tubulointerstitial damage in the kidneys, with a dose-dependent effect of the adenine concentration. 99mTc-DMSA SPECT-CT imaging showed a significant decrease in renal uptake over time in 0.25 and 0.5% ARD rats compared with control rats (P = 0.011 and P = 0.0004, respectively). 99mTc-DMSA uptake on Day 28 was significantly inversely correlated with Sirius red staining evaluated on Day 49 (r = 0.89, P < 0.0001, R2 = 0.67). CONCLUSIONS 99mTc-DMSA renal scintigraphy allows a longitudinal follow-up of risk of renal fibrosis in rats. We found that the reduction of renal parenchyma in ARD rats is inversely proportional to newly formed fibrous tissue in the kidney. Our results suggest that 99mTc-DMSA renal scintigraphy may be a useful non-invasive prognostic marker of the development of renal fibrosis in animals and should be tested in humans.
Collapse
Affiliation(s)
- Mickaël Bobot
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France.,Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France
| | - Guillaume Hache
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France.,Pharmacie, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Anaïs Moyon
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France.,Service de Radiopharmacie, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Samantha Fernandez
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| | - Laure Balasse
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| | - Laurent Daniel
- C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France.,Laboratoire d'Anatomopathologie, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Philippe Garrigue
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France.,Service de Radiopharmacie, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Pauline Brige
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,Laboratoire d'Imagerie Interventionnelle Expérimentale, EA, 4264, Aix Marseille Université, Marseille, France
| | - Sophie Chopinet
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,Laboratoire d'Imagerie Interventionnelle Expérimentale, EA, 4264, Aix Marseille Université, Marseille, France.,Service de Chirurgie Digestive, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | | | - Philippe Brunet
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France.,C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France
| | - Stéphane Burtey
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France.,C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France
| | - Benjamin Guillet
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,C2VN, INSERM 1263, INRAE 1260, Aix-Marseille Université, Marseille, France.,Service de Radiopharmacie, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| |
Collapse
|
9
|
Farris AB, Vizcarra J, Amgad M, Cooper LAD, Gutman D, Hogan J. Artificial intelligence and algorithmic computational pathology: an introduction with renal allograft examples. Histopathology 2021; 78:791-804. [PMID: 33211332 DOI: 10.1111/his.14304] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Whole slide imaging, which is an important technique in the field of digital pathology, has recently been the subject of increased interest and avenues for utilisation, and with more widespread whole slide image (WSI) utilisation, there will also be increased interest in and implementation of image analysis (IA) techniques. IA includes artificial intelligence (AI) and targeted or hypothesis-driven algorithms. In the overall pathology field, the number of citations related to these topics has increased in recent years. Renal pathology is one anatomical pathology subspecialty that has utilised WSIs and IA algorithms; it can be argued that renal transplant pathology could be particularly suited for whole slide imaging and IA, as renal transplant pathology is frequently classified by use of the semiquantitative Banff classification of renal allograft pathology. Hypothesis-driven/targeted algorithms have been used in the past for the assessment of a variety of features in the kidney (e.g. interstitial fibrosis, tubular atrophy, inflammation); in recent years, the amount of research has particularly increased in the area of AI/machine learning for the identification of glomeruli, for histological segmentation, and for other applications. Deep learning is the form of machine learning that is most often used for such AI approaches to the 'big data' of pathology WSIs, and deep learning methods such as artificial neural networks (ANNs)/convolutional neural networks (CNNs) are utilised. Unsupervised and supervised AI algorithms can be employed to accomplish image or semantic classification. In this review, AI and other IA algorithms applied to WSIs are discussed, and examples from renal pathology are covered, with an emphasis on renal transplant pathology.
Collapse
Affiliation(s)
- Alton B Farris
- Department of Pathology and Laboratory Medicine, Atlanta, GA, USA
| | - Juan Vizcarra
- Department of Bioinformatics, Emory University, Atlanta, GA, USA
| | - Mohamed Amgad
- Department of Pathology and Center for Computational Imaging and Signal Analytics, Northwestern University, Chicago, IL, USA
| | - Lee A D Cooper
- Department of Pathology and Center for Computational Imaging and Signal Analytics, Northwestern University, Chicago, IL, USA
| | - David Gutman
- Department of Bioinformatics, Emory University, Atlanta, GA, USA
| | - Julien Hogan
- Department of Surgery, Emory University, Atlanta, GA, USA
| |
Collapse
|
10
|
Ellingsen AR, Jørgensen KA, Østerby R, Petersen SE, Juul S, Marcussen N, Nyengaard JR. Human kidney graft survival correlates with structural parameters in baseline biopsies: a quantitative observational cohort study with more than 14 years' follow-up. Virchows Arch 2020; 478:659-668. [PMID: 32986179 DOI: 10.1007/s00428-020-02924-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 01/05/2023]
Abstract
This prospective cohort study evaluates associations between structural and ultrastructural parameters in baseline biopsies from human kidney transplants and long-term graft survival after more than 14 years' follow-up. Baseline kidney graft biopsies were obtained prospectively from 54 consecutive patients receiving a kidney transplant at a single institution. Quantitative measurements were performed on the baseline biopsies by computer-assisted light microscopy and electron microscopy. Stereology-based techniques estimated the fraction of interstitial tissue, the volume of glomeruli, mesangial fraction, and basement membrane thickness of glomerular capillaries. The fraction of occluded glomeruli and scores according to the Banff classification were achieved. Kidney graft survival was analyzed by Kaplan-Meier estimates and Cox regression. Association to long-term kidney function was also analyzed. The long-term surviving kidney transplants were characterized at implantation by less arteriolar hyaline thickening (P < 0.001) and less interstitial fibrosis (P = 0.001), as well as a lower fraction of occluded glomeruli (P = 0.004) and lower glomerular volume (P = 0.03). At the latest follow-up, eGFR was decreased by 12 ml/min/1.73 m2 per unit increase in the score for arteriolar hyalinosis at implantation (P = 0.02), and eGFR was decreased by 19 ml/min/1.73 m2 per 106 μm3 increase in glomerular volume at baseline (P = 0.03). The unbiased Cavalieri estimate of glomerular volume and the ultrastructural parameters are the first to be evaluated in a cohort study with prospective follow-up for more than 14 years. The study shows that baseline biopsies from human kidney grafts contain extraordinary long-term prognostic information, and it highlights the importance of these intrinsic graft factors.
Collapse
Affiliation(s)
- Anne R Ellingsen
- Electron Microscopy Laboratory, Aarhus University Hospital, 8200, Aarhus N, Denmark. .,Department of Pathology, Aarhus University Hospital, 8200, Aarhus N, Denmark. .,Department of Clinical Pathology, Odense University Hospital, 5000, Odense C, Denmark. .,Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark.
| | - Kaj A Jørgensen
- Department of Nephrology, Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Ruth Østerby
- Electron Microscopy Laboratory, Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Steffen E Petersen
- Department of Urology, Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Svend Juul
- Department of Public Health, Section for Epidemiology, Aarhus University, 8000, Aarhus C, Denmark
| | - Niels Marcussen
- Department of Clinical Pathology, Odense University Hospital, 5000, Odense C, Denmark
| | - Jens R Nyengaard
- Electron Microscopy Laboratory, Aarhus University Hospital, 8200, Aarhus N, Denmark.,Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark.,Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, 8000, Aarhus C, Denmark
| |
Collapse
|
11
|
Magnetic Resonance Imaging for Evaluation of Interstitial Fibrosis in Kidney Allografts. Transplant Direct 2020; 6:e577. [PMID: 33134501 PMCID: PMC7581173 DOI: 10.1097/txd.0000000000001009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/28/2020] [Indexed: 12/16/2022] Open
Abstract
Supplemental Digital Content is available in the text. Interstitial fibrosis (IF) is the common pathway of chronic kidney injury in various conditions. Magnetic resonance imaging (MRI) may be a promising tool for the noninvasive assessment of IF in renal allografts.
Collapse
|
12
|
Mortensen LA, Svane AM, Burton M, Bistrup C, Thiesson HC, Marcussen N, Beck HC. Proteomic Analysis of Renal Biomarkers of Kidney Allograft Fibrosis-A Study in Renal Transplant Patients. Int J Mol Sci 2020; 21:ijms21072371. [PMID: 32235494 PMCID: PMC7177439 DOI: 10.3390/ijms21072371] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022] Open
Abstract
Renal transplantation is the preferred treatment of end stage renal disease, but allograft survival is limited by the development of interstitial fibrosis and tubular atrophy in response to various stimuli. Much effort has been put into identifying new protein markers of fibrosis to support the diagnosis. In the present work, we performed an in-depth quantitative proteomics analysis of allograft biopsies from 31 prevalent renal transplant patients and correlated the quantified proteins with the volume fraction of fibrosis as determined by a morphometric method. Linear regression analysis identified four proteins that were highly associated with the degree of interstitial fibrosis, namely Coagulation Factor XIII A chain (estimate 18.7, adjusted p < 0.03), Uridine Phosphorylase 1 (estimate 19.4, adjusted p < 0.001), Actin-related protein 2/3 subunit 2 (estimate 34.2, adjusted p < 0.05) and Cytochrome C Oxidase Assembly Factor 6 homolog (estimate -44.9, adjusted p < 0.002), even after multiple testing. Proteins that were negatively associated with fibrosis (p < 0.005) were primarily related to normal metabolic processes and respiration, whereas proteins that were positively associated with fibrosis (p < 0.005) were involved in catabolic processes, cytoskeleton organization and the immune response. The identified proteins may be candidates for further validation with regards to renal fibrosis. The results support the notion that cytoskeleton organization and immune responses are prevalent processes in renal allograft fibrosis.
Collapse
Affiliation(s)
- Line Aas Mortensen
- Department of Nephrology, Odense University Hospital, DK-5000 Odense, Denmark; (L.A.M.); (C.B.); (H.C.T.)
| | - Anne Marie Svane
- Department of Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, DK-5000 Odense, Denmark;
| | - Mark Burton
- Department of Clinical Genetics, Odense University Hospital, DK-5000 Odense, Denmark;
| | - Claus Bistrup
- Department of Nephrology, Odense University Hospital, DK-5000 Odense, Denmark; (L.A.M.); (C.B.); (H.C.T.)
| | - Helle Charlotte Thiesson
- Department of Nephrology, Odense University Hospital, DK-5000 Odense, Denmark; (L.A.M.); (C.B.); (H.C.T.)
| | - Niels Marcussen
- Department of Pathology, Odense University Hospital, DK-5000 Odense, Denmark;
| | - Hans Christian Beck
- Department of Clinical Biochemistry and Pharmacology, Centre for Clinical Proteomics, Odense University Hospital, DK-5000 Odense, Denmark
- Correspondence:
| |
Collapse
|
13
|
Bane O, Hectors SJ, Gordic S, Kennedy P, Wagner M, Weiss A, Khaim R, Yi Z, Zhang W, Delaney V, Salem F, He C, Menon MC, Lewis S, Taouli B. Multiparametric magnetic resonance imaging shows promising results to assess renal transplant dysfunction with fibrosis. Kidney Int 2019; 97:414-420. [PMID: 31874802 DOI: 10.1016/j.kint.2019.09.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/31/2019] [Accepted: 09/26/2019] [Indexed: 12/28/2022]
Abstract
Here we assessed the diagnostic value of a quantitative multiparametric magnetic resonance imaging (mpMRI) protocol for evaluation of renal allograft dysfunction with fibrosis. Twenty-seven renal transplant patients, including 15 with stable functional allografts (eGFR mean 71.5 ml/min/1.73m2), and 12 with chronic dysfunction/established fibrosis (eGFR mean 30.1 ml/min/1.73m2), were enrolled in this prospective single-center study. Sixteen of the patients had renal biopsy (mean 150 days) before the MRI. All patients underwent mpMRI at 1.5T including intravoxel-incoherent motion diffusion-weighted imaging, diffusion tensor imaging, blood oxygen level dependent (BOLD R2*) and T1 quantification. True diffusion D, pseudodiffusion D*, perfusion fraction PF, apparent diffusion coefficient (ADC), fractional anisotropy (FA), R2* and T1 were calculated for cortex and medulla. ΔT1 was calculated as (100x(T1 Cortex-T1 Medulla)/T1 Cortex). Test-retest repeatability and inter-observer reproducibility were assessed in four and ten patients, respectively. mpMRI parameters had substantial test-retest and interobserver repeatability (coefficient of variation under 15%), except for medullary PF and D* (coefficient of variation over 25%). Cortical ADC, D, medullary ADC and ΔT1 were all significantly decreased, while cortical T1 was significantly elevated in fibrotic allografts. Cortical T1 showed positive correlation to the Banff fibrosis and tubular atrophy scores. The combination of ΔT1 and cortical ADC had excellent cross-validated diagnostic performance for detection of chronic dysfunction with fibrosis. Cortical ADC and T1 had good performance for predicting eGFR decline at 18 months (4 or more ml/min/1.73m2/year). Thus, the combination of cortical ADC and T1 measurements shows promising results for the non-invasive assessment of renal allograft histology and outcomes.
Collapse
Affiliation(s)
- Octavia Bane
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stefanie J Hectors
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Sonja Gordic
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Radiology, University Hospital Zürich, Zürich, Switzerland
| | - Paul Kennedy
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mathilde Wagner
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amanda Weiss
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rafael Khaim
- Division of Nephrology and Recanati Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zhengzi Yi
- Division of Nephrology and Recanati Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Weijia Zhang
- Division of Nephrology and Recanati Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Veronica Delaney
- Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Cijiang He
- Division of Nephrology and Recanati Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Madhav C Menon
- Division of Nephrology and Recanati Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sara Lewis
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bachir Taouli
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
14
|
Cockfield SM, Wilson S, Campbell PM, Cantarovich M, Gangji A, Houde I, Jevnikar AM, Keough‐Ryan TM, Monroy‐Cuadros F, Nickerson PW, Pâquet MR, Ramesh Prasad GV, Senécal L, Shoker A, Wolff J, Howell J, Schwartz JJ, Rush DN. Comparison of the effects of standard vs low-dose prolonged-release tacrolimus with or without ACEi/ARB on the histology and function of renal allografts. Am J Transplant 2019; 19:1730-1744. [PMID: 30582281 PMCID: PMC6590452 DOI: 10.1111/ajt.15225] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 01/25/2023]
Abstract
Targeting the renin-angiotensin system and optimizing tacrolimus exposure are both postulated to improve outcomes in renal transplant recipients (RTRs) by preventing interstitial fibrosis/tubular atrophy (IF/TA). In this multicenter, prospective, open-label controlled trial, adult de novo RTRs were randomized in a 2 × 2 design to low- vs standard-dose (LOW vs STD) prolonged-release tacrolimus and to angiotensin-converting enzyme inhibitors/angiotensin II receptor 1 blockers (ACEi/ARBs) vs other antihypertensive therapy (OAHT). There were 2 coprimary endpoints: the prevalence of IF/TA at month 6 and at month 24. IF/TA prevalence was similar for LOW vs STD tacrolimus at month 6 (36.8% vs 39.5%; P = .80) and ACEi/ARBs vs OAHT at month 24 (54.8% vs 58.2%; P = .33). IF/TA progression decreased significantly with LOW vs STD tacrolimus at month 24 (mean [SD] change, +0.42 [1.477] vs +1.10 [1.577]; P = .0039). Across the 4 treatment groups, LOW + ACEi/ARB patients exhibited the lowest mean IF/TA change and, compared with LOW + OAHT patients, experienced significantly delayed time to first T cell-mediated rejection. Renal function was stable from month 1 to month 24 in all treatment groups. No unexpected safety findings were detected. Coupled with LOW tacrolimus dosing, ACEi/ARBs appear to reduce IF/TA progression and delay rejection relative to reduced tacrolimus exposure without renin-angiotensin system blockade. ClinicalTrials.gov identifier: NCT00933231.
Collapse
Affiliation(s)
| | - Sam Wilson
- Astellas Pharma Global DevelopmentNorthbrookIllinois
| | | | | | - Azim Gangji
- St. Joseph's Healthcare HamiltonHamiltonOntarioCanada
| | | | | | | | | | | | | | | | | | | | | | - John Howell
- Astellas Pharma Global Development, Inc.MarkhamOntarioCanada
| | | | | |
Collapse
|
15
|
Tabibzadeh N, Wagner S, Metzger M, Flamant M, Houillier P, Boffa JJ, Vrtovsnik F, Thervet E, Stengel B, Haymann JP. Fasting Urinary Osmolality, CKD Progression, and Mortality: A Prospective Observational Study. Am J Kidney Dis 2019; 73:596-604. [PMID: 30777634 DOI: 10.1053/j.ajkd.2018.12.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 12/01/2018] [Indexed: 12/21/2022]
Abstract
RATIONALE & OBJECTIVE Chronic kidney disease (CKD) characterized by decreased glomerular filtration rate (GFR) is often accompanied by various degrees of impaired tubular function in the cortex and medulla. Assessment of tubular function may therefore be useful in establishing the severity of kidney disease and identifying those at greater risk for CKD progression. We explored reductions in urinary concentrating ability, a well-known feature of CKD, as a risk factor for GFR decline and end-stage renal disease (ESRD). STUDY DESIGN Prospective longitudinal cohort study. SETTING & PARTICIPANTS 2,084 adult patients with CKD stages 1 to 4 from the French NephroTest Cohort Study. PREDICTOR Fasting urinary osmolality measured using delta cryoscopy. OUTCOMES ESRD, mortality before ESRD, and measured GFR (mGFR) assessed using 51Cr-EDTA renal clearance. ANALYTICAL APPROACH Cause-specific hazards models were fit to estimate crude and adjusted associations of urinary osmolality with ESRD and death before ESRD. Linear mixed models with random intercepts were fit to evaluate the association of urinary osmolality with slope of decline in mGFR. RESULTS At baseline, mean age was 58.7±15.2 (SD) years with a median mGFR of 40.2 (IQR, 29.1-54.5) mL/min/1.73m2 and a median fasting urinary osmolality of 502.7±151.7mOsm/kg H2O. Baseline fasting urinary osmolality was strongly associated with mGFR (R=0.54; P < 0.001). 380 ESRD events and 225 deaths before ESRD occurred during a median follow-up of 5.9 (IQR, 3.8-8.2) years. Patients with lower baseline fasting urinary osmolality had higher adjusted risk for ESRD but not for mortality (HRs of 1.97 [95% CI, 1.26-3.08] and 0.99 [95% CI, 0.68-1.44], respectively, for the lowest vs highest tertile). Based on a mixed linear model adjusted for baseline mGFR and clinical characteristics, patients in the lowest tertile of baseline urinary osmolality had a steeper decline in kidney function (-4.9% ± 0.9% per year; P < 0.001) compared with patients in the highest tertile. LIMITATIONS Fasting was self-reported. CONCLUSIONS Fasting urinary osmolality may be a useful tool, in addition to GFR and albuminuria, for assessing nonglomerular damage in patients with CKD who are at higher risk for CKD progression.
Collapse
Affiliation(s)
- Nahid Tabibzadeh
- Sorbonne Université, Inserm UMR_S 1155, Paris, France; Unit of Renal Physiology, AP-HP Hôpital Tenon, Paris, France
| | - Sandra Wagner
- CESP, Inserm U1018, Univ Paris-Saclay, Univ Paris-Sud, UVSQ, Villejuif, France; FCRIN INI-CRCT, France
| | - Marie Metzger
- CESP, Inserm U1018, Univ Paris-Saclay, Univ Paris-Sud, UVSQ, Villejuif, France
| | - Martin Flamant
- Université Paris Diderot, Paris, France; Unit of Renal Physiology, AP-HP Hôpital Bichat, Paris, France
| | - Pascal Houillier
- Université Paris Descartes, INSERM UMR_S1138, Paris, France; Unit of Renal Physiology, AP-HP Hôpital Européen Georges Pompidou, Paris, France
| | - Jean-Jacques Boffa
- Sorbonne Université, Inserm UMR_S 1155, Paris, France; Unit of Nephrology, AP-HP Hôpital Tenon, Paris, France
| | - Francois Vrtovsnik
- Université Paris Diderot, Paris, France; Unit of Nephrology, AP-HP Hôpital Bichat, Paris, France
| | - Eric Thervet
- Université Paris Descartes, INSERM UMR_S1138, Paris, France; Unit of Nephrology, AP-HP Hôpital Européen Georges Pompidou, Paris, France
| | - Bénédicte Stengel
- CESP, Inserm U1018, Univ Paris-Saclay, Univ Paris-Sud, UVSQ, Villejuif, France; FCRIN INI-CRCT, France.
| | - Jean-Philippe Haymann
- Sorbonne Université, Inserm UMR_S 1155, Paris, France; Unit of Renal Physiology, AP-HP Hôpital Tenon, Paris, France
| | | |
Collapse
|
16
|
Yang C, Qi R, Yang B. Pathogenesis of Chronic Allograft Dysfunction Progress to Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:101-116. [PMID: 31399963 DOI: 10.1007/978-981-13-8871-2_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Kidney transplantation is a life-change measurement for the patients of end-stage renal disease (ESRD). However, the renal allograft cannot avoid initial acute kidney injury (AKI) and subsequent chronic allograft dysfunction (CAD), gradually develops fibrosis and eventually loses function. It is imperative to disclose the pathogenesis of AKI and CAD in order to facilitate interventions. We have studied the involvement of immunity, inflammation, and apoptosis in ischemia-reperfusion injury (IRI) and/or immunosuppressant induced AKI models, with associated chronic damage. Our research mainly focused on tubular epithelial cells (TECs) that are passive victims and also active participators in injury and mediate following repair or fibrosis. Targeting not only fibroblasts/myofibroblasts, but also TECs, might be a fundamental strategy to prevent and treat renal fibrosis. We have also evaluated the potential application of siRNA targeting caspase-3 and tissue protective erythropoietin derivatives, HBSP and CHBP, aiming to treat AKI and prevent CAD. Significant improvements have been obtained, but timely diagnosis and precise therapy of AKI and prevention of CAD progressing to ESRD are still very challenging. Modern technologies such as microarray and sequencing analysis have been used to identify biomarkers and potentially facilitate individual cell target treatment for transplant patients.
Collapse
Affiliation(s)
- Cheng Yang
- Department of Urology, Zhongshan Hospital, Zhangjiang Technology Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruochen Qi
- Department of Urology, Zhongshan Hospital, Zhangjiang Technology Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Bin Yang
- Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, 226001, Jiangsu, China. .,Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, LE1 7RH, UK.
| |
Collapse
|
17
|
Andrikopoulos P, Kieswich J, Pacheco S, Nadarajah L, Harwood SM, O'Riordan CE, Thiemermann C, Yaqoob MM. The MEK Inhibitor Trametinib Ameliorates Kidney Fibrosis by Suppressing ERK1/2 and mTORC1 Signaling. J Am Soc Nephrol 2018; 30:33-49. [PMID: 30530834 DOI: 10.1681/asn.2018020209] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/13/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND During kidney fibrosis, a hallmark and promoter of CKD (regardless of the underlying renal disorder leading to CKD), the extracellular-regulated kinase 1/2 (ERK1/2) pathway, is activated and has been implicated in the detrimental differentiation and expansion of kidney fibroblasts. An ERK1/2 pathway inhibitor, trametinib, is currently used in the treatment of melanoma, but its efficacy in the setting of CKD and renal fibrosis has not been explored. METHODS We investigated whether trametinib has antifibrotic effects in two mouse models of renal fibrosis-mice subjected to unilateral ureteral obstruction (UUO) or fed an adenine-rich diet-as well as in cultured primary human fibroblasts. We also used immunoblot analysis, immunohistochemical staining, and other tools to study underlying molecular mechanisms for antifibrotic effects. RESULTS Trametinib significantly attenuated collagen deposition and myofibroblast differentiation and expansion in UUO and adenine-fed mice. We also discovered that in injured kidneys, inhibition of the ERK1/2 pathway by trametinib ameliorated mammalian target of rapamycin complex 1 (mTORC1) activation, another key profibrotic signaling pathway. Trametinib also inhibited the ERK1/2 pathway in cultured primary human renal fibroblasts stimulated by application of TGF-β1, the major profibrotic cytokine, thereby suppressing downstream mTORC1 pathway activation. Additionally, trametinib reduced the expression of myofibroblast marker α-smooth muscle actin and the proliferation of renal fibroblasts, corroborating our in vivo data. Crucially, trametinib also significantly ameliorated renal fibrosis progression when administered to animals subsequent to myofibroblast activation. CONCLUSIONS Further study of trametinib as a potential candidate for the treatment of chronic renal fibrotic diseases of diverse etiologies is warranted.
Collapse
Affiliation(s)
- Petros Andrikopoulos
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK; and .,Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julius Kieswich
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK; and.,Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sabrina Pacheco
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK; and.,Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Luxme Nadarajah
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Steven Michael Harwood
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK; and.,Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Caroline E O'Riordan
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Christoph Thiemermann
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK; and.,Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muhammad M Yaqoob
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK; and.,Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
18
|
Ellis RJ, Kalma B, Del Vecchio SJ, Aliano DN, Ng KL, Dimeski G, Ma L, Guard D, Bertram JF, Morais C, Oliver K, Wood ST, Gobe GC, Francis RS. Chronic kidney cortical damage is associated with baseline kidney function and albuminuria in patients managed with radical nephrectomy for kidney tumours. Pathology 2018; 51:32-38. [PMID: 30477884 DOI: 10.1016/j.pathol.2018.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/16/2018] [Accepted: 10/18/2018] [Indexed: 01/12/2023]
Abstract
This study evaluated the relationship between histological markers of chronic kidney damage in patients undergoing radical nephrectomy for kidney tumours and preoperative kidney function, degree of albuminuria, and changes in glomerular volume. A schema to grade chronic kidney damage could be used to identify patients at risk of developing CKD following nephrectomy. Non-neoplastic cortical tissue was sourced from 150 patients undergoing radical nephrectomy for suspected kidney cancer. This tissue was evaluated for indicators of chronic damage, specifically: glomerulosclerosis, arteriosclerosis, interstitial fibrosis, and tubular atrophy. Glomerular volume was determined using the Weibel and Gomez method. Associations between these parameters and both estimated glomerular filtration rate (eGFR) and albumin-creatinine ratio (ACR) were determined using either a Mann-Whitney U-test or a Kruskal-Wallis ANOVA. Associations between both eGFR and ACR and glomerular volume were assessed using linear regression. eGFR was inversely associated with the degree of glomerulosclerosis (p < 0.001), vascular narrowing (p = 0.002), tubular atrophy (p < 0.001), and interstitial fibrosis (p < 0.001). ACR was associated only with the degree of interstitial fibrosis (p = 0.02) and tubular atrophy (p = 0.02). Glomerular volume was greater for males, diabetics, hypertensive patients, and patients with a greater degree of interstitial fibrosis. Glomerular volume was positively associated with ACR. A schema to grade chronic damage was developed. The proposed schema is associated with baseline clinical indices of kidney function and damage. Longitudinal validation is necessary to determine the prognostic utility of this schema.
Collapse
Affiliation(s)
- Robert J Ellis
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia.
| | - Benjamin Kalma
- Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| | - Sharon J Del Vecchio
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| | - Danielle N Aliano
- Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| | - Keng Lim Ng
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| | - Goce Dimeski
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| | - Li Ma
- Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - David Guard
- Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - John F Bertram
- Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Vic, Australia
| | - Christudas Morais
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| | | | - Simon T Wood
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| | - Glenda C Gobe
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia; School of Biomedical Sciences, University of Queensland, Brisbane, Qld, Australia; NHMRC Chronic Kidney Disease Centre for Research Excellence, University of Queensland, Brisbane, Qld, Australia
| | - Ross S Francis
- Princess Alexandra Hospital, Brisbane, Qld, Australia; Centre for Kidney Disease Research, University of Queensland, Brisbane, Qld, Australia; Translational Research Institute, Brisbane, Qld, Australia
| |
Collapse
|
19
|
Kholia S, Herrera Sanchez MB, Cedrino M, Papadimitriou E, Tapparo M, Deregibus MC, Brizzi MF, Tetta C, Camussi G. Human Liver Stem Cell-Derived Extracellular Vesicles Prevent Aristolochic Acid-Induced Kidney Fibrosis. Front Immunol 2018; 9:1639. [PMID: 30072992 PMCID: PMC6060249 DOI: 10.3389/fimmu.2018.01639] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/03/2018] [Indexed: 11/29/2022] Open
Abstract
With limited therapeutic intervention in preventing the progression to end-stage renal disease, chronic kidney disease (CKD) remains a global health-care burden. Aristolochic acid (AA) induced nephropathy is a model of CKD characterised by inflammation, tubular injury, and interstitial fibrosis. Human liver stem cell-derived extracellular vesicles (HLSC-EVs) have been reported to exhibit therapeutic properties in various disease models including acute kidney injury. In the present study, we aimed to investigate the effects of HLSC-EVs on tubular regeneration and interstitial fibrosis in an AA-induced mouse model of CKD. NSG mice were injected with HLSC-EVs 3 days after administering AA on a weekly basis for 4 weeks. Mice injected with AA significantly lost weight over the 4-week period. Deterioration in kidney function was also observed. Histology was performed to evaluate tubular necrosis, interstitial fibrosis, as well as infiltration of inflammatory cells/fibroblasts. Kidneys were also subjected to gene array analyses to evaluate regulation of microRNAs (miRNAs) and pro-fibrotic genes. The effect of HLSC-EVs was also tested in vitro to assess pro-fibrotic gene regulation in fibroblasts cocultured with AA pretreated tubular epithelial cells. Histological analyses showed that treatment with HLSC-EVs significantly reduced tubular necrosis, interstitial fibrosis, infiltration of CD45 cells and fibroblasts, which were all elevated during AA induced injury. At a molecular level, HLSC-EVs significantly inhibited the upregulation of the pro-fibrotic genes α-Sma, Tgfb1, and Col1a1 in vivo and in vitro. Fibrosis gene array analyses revealed an upregulation of 35 pro-fibrotic genes in AA injured mice. Treatment with HLSC-EVs downregulated 14 pro-fibrotic genes in total, out of which, 5 were upregulated in mice injured with AA. Analyses of the total mouse miRnome identified several miRNAs involved in the regulation of fibrotic pathways, which were found to be modulated post-treatment with HLSC-EVs. These results indicate that HLSC-EVs play a regenerative role in CKD possibly through the regulation of genes and miRNAs that are activated during the progression of the disease.
Collapse
Affiliation(s)
- Sharad Kholia
- Department of Medical Sciences, University of Torino, Torino, Italy.,Molecular Biotechnology Centre, University of Torino, Torino, Italy
| | - Maria Beatriz Herrera Sanchez
- Molecular Biotechnology Centre, University of Torino, Torino, Italy.,2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Torino, Torino, Italy
| | - Massimo Cedrino
- Molecular Biotechnology Centre, University of Torino, Torino, Italy.,2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Torino, Torino, Italy
| | - Elli Papadimitriou
- Molecular Biotechnology Centre, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Marta Tapparo
- Department of Medical Sciences, University of Torino, Torino, Italy.,Molecular Biotechnology Centre, University of Torino, Torino, Italy
| | - Maria Chiara Deregibus
- Molecular Biotechnology Centre, University of Torino, Torino, Italy.,2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Torino, Torino, Italy
| | | | | | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
20
|
Zhang C, Liang S, Cheng S, Li W, Wang X, Zheng C, Zeng C, Shi S, Xie L, Zen K, Liu Z. Urinary miR-196a predicts disease progression in patients with chronic kidney disease. J Transl Med 2018; 16:91. [PMID: 29636065 PMCID: PMC5894160 DOI: 10.1186/s12967-018-1470-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/02/2018] [Indexed: 11/13/2022] Open
Abstract
Background Urinary miRNAs may potentially serve as noninvasive biomarkers in various kidney diseases to reflect disease activity, severity and progression, especially those correlated with the pathogenesis of kidney diseases. This study demonstrates that urinary miR-196a, a kidney-enriched miRNA, can predict progression of chronic kidney disease (CKD). Methods Focal segmental glomerulosclerosis (FSGS) cohorts were used as the representative example of CKD. First, correlation of miR-196a with disease activity was analyzed using paired urine and plasma samples from FSGS patients with nephrotic-range proteinuria (FSGS-A), complete remission (FSGS-CR) and normal controls (NCs). Then, the value of urinary miR-196a in predicting disease progression was validated using another cohort of 231 FSGS patients who were followed-up until over 36 months or reaching end-stage renal disease (ESRD). MiR-196a levels were analyzed by quantitative reverse transcription-polymerase chain reaction. Results The results showed that urinary miR-196a significantly increased in FSGS-A compared with FSGS-CR and NCs, clearly distinguishing FSGS-A from FSGS-CR and NCs, whereas plasma miR-196a showed no difference among these groups. Moreover, urinary miR-196a, which was associated with proteinuria, estimated glomerular filtration rate (eGFR), interstitial fibrosis and tubular atrophy, significantly increased in patients progressed to ESRD compared to those not. Furthermore, patients with higher urinary miR-196a displayed poorer renal survival than those with lower urinary miR-196a. Multivariate Cox analysis confirmed urinary miR-196a as an independent risk factor for FSGS progression after adjusting for age, sex, proteinuria and eGFR. Prediction accuracy of ESRD was significantly improved by combining urinary miR-196a with other indicators including eGFR and proteinuria. Conclusion Urinary miR-196a may serve as a biomarker for predicting CKD progression Electronic supplementary material The online version of this article (10.1186/s12967-018-1470-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Changming Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Shaoshan Liang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Shuiqin Cheng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Wei Li
- Shanghai Center for Bioinformation Research Technology, Shanghai Academy of Science and Technology, Shanghai, China
| | - Xia Wang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Chunxia Zheng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Shaolin Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Lu Xie
- Shanghai Center for Bioinformation Research Technology, Shanghai Academy of Science and Technology, Shanghai, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, 210093, Jiangsu, China.
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
21
|
Wang H, Zheng C, Lu Y, Jiang Q, Yin R, Zhu P, Zhou M, Liu Z. Urinary Fibrinogen as a Predictor of Progression of CKD. Clin J Am Soc Nephrol 2017; 12:1922-1929. [PMID: 28903970 PMCID: PMC5718264 DOI: 10.2215/cjn.01360217] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 08/17/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND OBJECTIVES Fibrinogen has been reported to be involved in kidney tubulointerstitial fibrosis and podocyte injury in mouse models. However, the relationship between urinary fibrinogen and kidney outcomes has not been clarified in patients with CKD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We evaluated 402 patients with CKD and kidney biopsies, including 101 with diabetic nephropathy, 94 with idiopathic membranous nephropathy, 55 with idiopathic FSGS, and 152 with IgA nephropathy. We quantified urinary fibrinogen by ELISA and tested associations with kidney histology and progression to ESRD. RESULTS Median (interquartile range) urinary fibrinogen-to-creatinine ratio was 536 (191-1461) ng/mg for patients with CKD, significantly higher than 2 (2-3) ng/mg for healthy controls (P<0.001). Urinary fibrinogen was positively correlated with urine protein (r=0.64; P<0.001) and interstitial fibrosis and tubular atrophy (r=0.10; P=0.04), and it was negatively correlated with eGFR (r=-0.20; P<0.001). Over a median follow-up period of 35 months (interquartile range, 24-78 months), 68 of 402 patients (17%) developed ESRD. Higher urinary fibrinogen level was associated with increased risk of ESRD (hazard ratio, 2.12; 95% confidence interval, 1.31 to 3.26) per log10 higher urinary fibrinogen-to-creatinine ratio (P=0.003) adjusting for age, sex, BP, urine protein, disease type, eGFR, and interstitial fibrosis and tubular atrophy. For prediction of ESRD, the addition of urinary fibrinogen to eGFR, urine protein, and BP increased the area under the receiver operating curve from 0.73 to 0.76, and the Akaike information criterion improved from 333.6 to 327.0. CONCLUSIONS Urinary fibrinogen correlated with interstitial fibrosis and tubular atrophy and was an independent risk factor for progression of CKD to ESRD.
Collapse
Affiliation(s)
- Hongtian Wang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, China
| | - Chunxia Zheng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
| | - Yinghui Lu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
| | - Qi Jiang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
| | - Ru Yin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
| | - Ping Zhu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
| | - Minlin Zhou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, China
| |
Collapse
|
22
|
Development of CD3 cell quantitation algorithms for renal allograft biopsy rejection assessment utilizing open source image analysis software. Virchows Arch 2017; 472:259-269. [DOI: 10.1007/s00428-017-2260-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/29/2017] [Accepted: 10/24/2017] [Indexed: 12/18/2022]
|
23
|
Leaf IA, Duffield JS. What can target kidney fibrosis? Nephrol Dial Transplant 2017; 32:i89-i97. [PMID: 28391346 DOI: 10.1093/ndt/gfw388] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/04/2016] [Indexed: 11/14/2022] Open
Abstract
Fibrosis, a characteristic of all chronic kidney diseases, is now recognized to be an independent predictor of disease progression. Deposition of pathological matrix in the walls of glomerular capillaries, the interstitial space and around arterioles both predicts and contributes to functional demise of the nephron and its surrounding vasculature. Recent identification of the major cell populations of fibroblast precursors in the kidney interstitium as pericytes and tissue-resident mesenchymal stem cells, and in the glomerulus as podocytes, parietal epithelial and mesangial cells, has enabled the study of the fibrogenic process in much greater depth directly in the fibroblast precursors. These cells are not only matrix-producing cells, but are also important innate immune surveillance cells that regulate the inflammatory process, exacerbate tissue damage by release of radicals and cytokines, and contribute to parenchymal and microvascular dysfunction by aberrant wound-healing responses. Innate immune signaling in fibroblasts and their precursors is intimately intertwined with the process of fibrogenesis. In addition, genomic and genetic studies also point to defective responses in loci close to genes involved in solute transport, metabolism, autophagy, protein handling and vascular homeostasis, principally in the epithelium and endothelium, as upstream drivers of the fibrotic process, indicating that cellular crosstalk is vital for development of fibrosis. As we move beyond TGFβ inhibition as a central target for fibrosis, targeting innate immune signaling and metabolic dysfunction appear increasingly tenable alternative targets for novel therapies.
Collapse
Affiliation(s)
- Irina A Leaf
- Research & Development, Biogen, Cambridge, MA, USA
| | - Jeremy S Duffield
- Research & Development, Biogen, Cambridge, MA, USA.,University of Washington, Seattle, WA, USA
| |
Collapse
|
24
|
Ju W, Nair V, Smith S, Zhu L, Shedden K, Song PXK, Mariani LH, Eichinger FH, Berthier CC, Randolph A, Lai JYC, Zhou Y, Hawkins JJ, Bitzer M, Sampson MG, Thier M, Solier C, Duran-Pacheco GC, Duchateau-Nguyen G, Essioux L, Schott B, Formentini I, Magnone MC, Bobadilla M, Cohen CD, Bagnasco SM, Barisoni L, Lv J, Zhang H, Wang HY, Brosius FC, Gadegbeku CA, Kretzler M. Tissue transcriptome-driven identification of epidermal growth factor as a chronic kidney disease biomarker. Sci Transl Med 2016; 7:316ra193. [PMID: 26631632 DOI: 10.1126/scitranslmed.aac7071] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic kidney disease (CKD) affects 8 to 16% people worldwide, with an increasing incidence and prevalence of end-stage kidney disease (ESKD). The effective management of CKD is confounded by the inability to identify patients at high risk of progression while in early stages of CKD. To address this challenge, a renal biopsy transcriptome-driven approach was applied to develop noninvasive prognostic biomarkers for CKD progression. Expression of intrarenal transcripts was correlated with the baseline estimated glomerular filtration rate (eGFR) in 261 patients. Proteins encoded by eGFR-associated transcripts were tested in urine for association with renal tissue injury and baseline eGFR. The ability to predict CKD progression, defined as the composite of ESKD or 40% reduction of baseline eGFR, was then determined in three independent CKD cohorts. A panel of intrarenal transcripts, including epidermal growth factor (EGF), a tubule-specific protein critical for cell differentiation and regeneration, predicted eGFR. The amount of EGF protein in urine (uEGF) showed significant correlation (P < 0.001) with intrarenal EGF mRNA, interstitial fibrosis/tubular atrophy, eGFR, and rate of eGFR loss. Prediction of the composite renal end point by age, gender, eGFR, and albuminuria was significantly (P < 0.001) improved by addition of uEGF, with an increase of the C-statistic from 0.75 to 0.87. Outcome predictions were replicated in two independent CKD cohorts. Our approach identified uEGF as an independent risk predictor of CKD progression. Addition of uEGF to standard clinical parameters improved the prediction of disease events in diverse CKD populations with a wide spectrum of causes and stages.
Collapse
Affiliation(s)
- Wenjun Ju
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Viji Nair
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shahaan Smith
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Li Zhu
- Renal Division, Department of Internal Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing 100034, China
| | - Kerby Shedden
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Statistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X K Song
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura H Mariani
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.,Arbor Research Collaborative for Health, Ann Arbor, MI 48104, USA
| | - Felix H Eichinger
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Celine C Berthier
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ann Randolph
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer Yi-Chun Lai
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yan Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer J Hawkins
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Markus Bitzer
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthew G Sampson
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Martina Thier
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | - Corinne Solier
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | - Gonzalo C Duran-Pacheco
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | | | - Laurent Essioux
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | - Brigitte Schott
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | - Ivan Formentini
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | - Maria C Magnone
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | - Maria Bobadilla
- Roche Pharmaceutical Research and Early Development-Roche Innovation Center, 4070 Basel, Switzerland
| | - Clemens D Cohen
- Division of Nephrology, Institute of Physiology, University of Zurich, CH-8006 Zürich, Switzerland
| | - Serena M Bagnasco
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Laura Barisoni
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Jicheng Lv
- Renal Division, Department of Internal Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing 100034, China
| | - Hong Zhang
- Renal Division, Department of Internal Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing 100034, China
| | - Hai-Yan Wang
- Renal Division, Department of Internal Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing 100034, China
| | - Frank C Brosius
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Crystal A Gadegbeku
- Temple Clinical Research Institute, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Matthias Kretzler
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
25
|
Farris AB, Chan S, Climenhaga J, Adam B, Bellamy COC, Serón D, Colvin RB, Reeve J, Mengel M. Banff fibrosis study: multicenter visual assessment and computerized analysis of interstitial fibrosis in kidney biopsies. Am J Transplant 2014; 14:897-907. [PMID: 24712330 DOI: 10.1111/ajt.12641] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 01/25/2023]
Abstract
Increasing interstitial fibrosis (IF) in native and kidney transplant biopsies is associated with functional decline and serves as a clinical trial end point. A Banff 2009 Conference survey revealed a range in IF assessment practices. Observers from multiple centers were asked to assess 30 renal biopsies with a range of IF and quantitate IF using two approaches on trichrome, Periodic acid-Schiff (PAS) and computer-assisted quantification of collagen III immunohistochemistry (C-IHC) slides, as well as assessing percent of cortical tubular atrophy% (TA%) and Banff total cortical inflammation score (ti-score). C-IHC using whole slide scans was performed. C-IHC assessment showed a higher correlation with organ function (r = -0.48) than did visual assessments (r = -0.32--0.42); computerized and visual C-IHC assessment also correlated (r = 0.64-0.66). Visual assessment of trichrome and C-IHC showed better correlations with organ function and C-IHC, than PAS, TA% and ti-score. However, visual assessment of IF, independent of approach, was variable among observers, and differences in correlations with organ function were not statistically significant among C-IHC image analysis and visual assessment methods. C-IHC image analysis correlated among three centers (r > 0.90, p < 0.0001, between all centers). Given the difficulty of visual IF assessment standardization, C-IHC image could potentially accomplish standardized IF assessment in multicenter settings.
Collapse
Affiliation(s)
- A B Farris
- Department of Pathology, Emory University, Atlanta, GA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Distribution and expression of fibroblast-specific protein chemokine CCL21 and chemokine receptor CCR7 in renal allografts. Transplant Proc 2013; 45:538-45. [PMID: 23498789 DOI: 10.1016/j.transproceed.2012.10.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/27/2012] [Accepted: 10/09/2012] [Indexed: 11/23/2022]
Abstract
We aimed to characterize the expression and distribution of the fibroblast surface protein (FSP), the chemokine CC-ligand 21 (CCL21) secondary lymphoid tissue chemokine CC-chemokine receptor 7 (CCR7) in renal allograft biopsy specimens obtained from patients after transplantation. We recruited 165 patients who received renal transplants at our center for this study. Histological examination of the renal allograft biopsy specimens was performed using hematoxylin-eosin, periodic acid-Schiff, and Masson's trichrome staining. Distribution and expression of FSP, CCL21, and CCR7 were determined using immunohistochemistry staining. Serum creatinine levels were evaluated using an enzymatic sarcosine oxidase method. FSP was mainly localized in the cytoplasm and nucleus of renal interstitial fibroblasts and tubular epithelial cells. Compared with the normal group, an elevated number of FSP-positive fibroblasts were observed in patients with acute/active cellular rejection and chronic/sclerosing allograft nephropathy (P < .05). Patients with chronic/sclerosing allograft nephropathy also showed increased total fibroblasts as compared with borderline changes (P < .05). In a multiple regression analysis, CCR7-positive expression was a strong protective factor for acute/active cellular rejection and recurrent nephropathy (odds ratio [OR] = 0.12, P = .034, and OR = 0.08; P = .036, respectively). In contrast, CCL21-positive expression led to a high susceptibility to recurrent nephropathy among renal transplant patients (OR = 10.41, P = .029). Moreover, FSP and CCL21, or CCL21 and CCR7 were localized in the interstitial fibroblasts and renal tubular epithelium cells. In addition, FSP and CCL21 expression positively correlated with serum creatinine levels. Our results suggested that the CCL21/CCR7 signaling pathway is involved in renal fibrosis in kidney transplant patients. An increased number of FSP-positive fibroblasts may be a risk factor for acute/active cellular rejection and chronic/sclerosing allograft nephropathy after renal transplantation. These findings may help understanding of renal allograft fibrosis.
Collapse
|
27
|
Prunotto M, Budd DC, Gabbiani G, Meier M, Formentini I, Hartmann G, Pomposiello S, Moll S. Epithelial-mesenchymal crosstalk alteration in kidney fibrosis. J Pathol 2012; 228:131-47. [PMID: 22570261 DOI: 10.1002/path.4049] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 04/19/2012] [Accepted: 04/26/2012] [Indexed: 02/06/2023]
Abstract
The incidence of chronic kidney diseases (CKD) is constantly rising, reaching epidemic proportions in the western world and leading to an enormous threat, even to modern health-care systems, in industrialized countries. Therapies of CKD have greatly improved following the introduction of drugs targeting the renin-angiotensin system (RAAS) but even this refined pharmacological approach has failed to stop progression to end-stage renal disease (ESRD) in many individuals. In vitro historical data and recent new findings have suggested that progression of renal fibrosis might occur as a result of an altered tubulo-interstitial microenvironment and, more specifically, as a result of an altered epithelial-mesenchymal crosstalk. Here we the review biological findings that support the hypothesis of an altered cellular crosstalk in an injured local tubulo-interstitial microenvironment leading to renal disease progression. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Marco Prunotto
- CV and Metabolic DTA Department, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Tubulointerstitial injury in the kidney is complex, involving a number of independent and overlapping cellular and molecular pathways, with renal interstitial fibrosis and tubular atrophy (IFTA) as the final common pathway. Furthermore, there are multiple ways to assess IFTA. RECENT FINDINGS Cells involved include tubular epithelial cells, fibroblasts, fibrocytes, myofibroblasts, monocyte/macrophages, and mast cells with complex and still incompletely characterized cell-molecular interactions. Molecular mediators involved are numerous and involve pathways such as transforming growth factor (TGF)-β, bone morphogenic protein (BMP), platelet-derived growth factor (PDGF), and hepatocyte growth factor (HGF). Recent genomic approaches have shed insight into some of these cellular and molecular pathways. Pathologic evaluation of IFTA is central in assessing the severity of chronic disease; however, there are a variety of methods used to assess IFTA. Most assessment of IFTA relies on pathologist assessment of special stains such as trichrome, Sirius Red, and collagen III immunohistochemistry. Visual pathologist assessment can be prone to intra and interobserver variability, but some methods employ computerized morphometery, without a clear consensus as to the best method. SUMMARY IFTA results from on orchestration of cell types and molecular pathways. Opinions vary on the optimal qualitative and quantitative assessment of IFTA.
Collapse
Affiliation(s)
- Alton B Farris
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia 30322, USA.
| | | |
Collapse
|
29
|
New computerized color image analysis for the quantification of interstitial fibrosis in renal transplantation. Transplantation 2011; 92:890-9. [PMID: 21926945 DOI: 10.1097/tp.0b013e31822d879a] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Chronic allograft injury, the primary cause of late allograft failure in renal transplantation, can be diagnosed early at a preclinical stage by histopathological changes such as interstitial fibrosis (IF). Currently, assessed by semiquantitative analysis in the Banff classification, IF quantification is limited by pathologist's subjective interpretation. METHODS We have designed algorithms dedicated to quantify IF by computerized color image analysis. This innovative and objective software automatically extracts the green areas characteristic of IF in Masson's trichrome based on color image segmentation followed by removal of nonspecific IF staining (capsula, sclerosis glomeruli and normal glomeruli, normal basement membrane) and computes an index. It also counts automatically the number of glomeruli. Sixty-seven Masson stained renal transplant biopsies at various IF stages were imaged using a digital color camera mounted on a microscope. We tested the robustness of the method against varying acquisition parameters. RESULTS We demonstrated that the parameters do not have an impact on this quantification and that the algorithm is able to handle biopsy color variations. The intra- and interobserver reproducibility was good (P<0.003). The kappa coefficient that was performed on another set of 90 biopsies to evaluate the concordance of our method with an expert Banff quantification was 0.68, indicating a substantial agreement. Finally, the computerized IF correlated with renal function. CONCLUSION This study demonstrates that computerized color image analysis is a reliable and reproducible method to evaluate renal IF in routine practice and in multi-centric studies.
Collapse
|
30
|
Servais A, Meas-Yedid V, Noël LH, Martinez F, Panterne C, Kreis H, Zuber J, Timsit MO, Legendre C, Olivo-Marin JC, Thervet E. Interstitial fibrosis evolution on early sequential screening renal allograft biopsies using quantitative image analysis. Am J Transplant 2011; 11:1456-63. [PMID: 21672152 DOI: 10.1111/j.1600-6143.2011.03594.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Screening renal biopsies (RB) may assess early changes of interstitial fibrosis (IF) after transplantation. The aim of this study was to quantify IF by automatic color image analysis on sequential RB. We analyzed RB performed at day (D) 0, month (M) 3 and M12 from 140 renal transplant recipients with a program of color segmentation imaging. The mean IF score was 19 ± 9% at D0, 27 ± 11% at M3 and 32 ± 11% at M12 with a 8% progression during the first 3 months and 5% between M3 and M12. IF at M3 was correlated with estimated glomerular rate (eGFR) at M3, 12 and 24 (p < 0.02) and IF at M12 with eGFR at M12 and 48 (p < 0.05). Furthermore, IF evolution between D0 and M3 (ΔIFM3-D0) was correlated with eGFR at M24, 36 and 48 (p < 0.03). IF at M12 was significantly associated with male donor gender and tacrolimus dose (p = 0.03). ΔIFM3-D0 was significantly associated with male donor gender, acute rejection episodes (p = 0.04) and diabetes mellitus (p = 0.02). Thus, significant IF is already present before transplantation. IF evolution is more important during the first 3 months and has some predictive ability for change in GFR. Intervention to decrease IF should be applied early, i.e. before 3 months, after transplantation.
Collapse
Affiliation(s)
- A Servais
- Department of Nephrology, Assistance publique-Hôpitaux de Paris, Necker-Enfants Malades Hospital, Paris, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Birk PE, Gill JS, Blydt-Hansen TD, Gibson IW. Enhanced resolution of interstitial fibrosis in pediatric renal allograft biopsies using image analysis of trichrome stain. Pediatr Transplant 2010; 14:925-30. [PMID: 20946518 DOI: 10.1111/j.1399-3046.2010.01376.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The Banff classification is ill suited to detect subtle histologic progression in renal allografts. We present image analysis methodology to precisely quantify IF in pediatric renal allograft biopsies routinely stained with MT. The mean area %IF was determined in 105 pediatric renal allograft biopsies. Associations between %IF or Banff ci scores and estimated GFR were determined using GEE modeling. Logistic regression was used to estimate IF progression. Percent IF (mean ± s.d.) was 6.83% ± 3.94, 10.39 ± 5.23%, and 20.53 ± 8.74 in patients with ci0, ci1, and ci2, respectively. The difference in %IF between biopsies with ci0, ci1, and ci2 was not proportionately incremental: compared to ci2, ci0 had 67% less IF (p < 0.0001), while ci1 had 48% less IF (p < 0.0001). AR had no impact on the precision of %IF measurements. Each 0.5% decrement in %IF was associated with a 1 mL/min per 1.73 m² increase in GFR (p < 0.004). Histologic progression was demonstrated by increasing %IF values (p < 0.0001) and could be estimated by IF = 2.61 × (months) + 6.43. This readily adaptable methodology may be used for the longitudinal assessment of IF in pediatric protocol renal allograft biopsies.
Collapse
Affiliation(s)
- Patricia E Birk
- Department of Pediatrics, University of Manitoba, Winnipeg, MB, Canada.
| | | | | | | |
Collapse
|
32
|
Blydt-Hansen TD, Gibson IW, Birk PE. Histological progression of chronic renal allograft injury comparing sirolimus and mycophenolate mofetil-based protocols. A single-center, prospective, randomized, controlled study. Pediatr Transplant 2010; 14:909-18. [PMID: 20880093 DOI: 10.1111/j.1399-3046.2010.01374.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
In an effort to mitigate progression of IF/TA associated with chronic renal allograft injury, we hypothesize that adjuvant immunosuppression with sirolimus (SRL) will delay progression compared with MMF. Subjects 5-17 yr old, >1-yr post-transplant with mild or moderate IF/TA (Banff criteria) and tacrolimus dose minimization were randomized to continue MMF or convert to SRL and followed for two yr. For the entire cohort (n = 20), there was significant progression of %GGS, ci, ct, cv, and ah scores over the follow-up period (p < 0.05). There was no difference in rates of progression of Banff scores, %GGS, or % IF over two yr between the two groups, though power was low. Both groups exhibited similar rates of eGFR decline (MMF: -12.3 vs. SRL: -11.8 mL/min/1.73 m²/yr), which was correlated with ct score (p < 0.05). The SRL group had more episodes of acute allograft dysfunction and oral ulcers. Proteinuria at 24 months was significantly increased in the SRL group (6/9 subjects) but was not correlated with eGFR or %GGS. We conclude that neither MMF nor SRL, combined with low-dose tacrolimus, was effective at mitigating progressive histological changes or functional decline associated with chronic renal allograft injury.
Collapse
Affiliation(s)
- Tom D Blydt-Hansen
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada.
| | | | | |
Collapse
|
33
|
Early urinary CCL2 is associated with the later development of interstitial fibrosis and tubular atrophy in renal allografts. Transplantation 2010; 90:394-400. [PMID: 20625355 DOI: 10.1097/tp.0b013e3181e6424d] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Chronic renal allograft injury resulting in progressive interstitial fibrosis and tubular atrophy (IFTA) is a leading cause of graft loss. The goal of this study was to identify early urinary predictors for the subsequent development of IFTA in a prospective cohort of patients (n=111) who underwent serial protocol biopsies at 0, 6, and 24 months. METHODS The urinary proteins evaluated were CCL2, CXCL9, CXCL10, and alpha1-microglobulin (alpha1M) using ELISA and immunonephelometry. RESULTS We first evaluated urines obtained at 1 to 3 months and found that alpha1M and CXCL10 were associated with IFTA at 6 months but not at 24 months. Next, we evaluated urines at 6 months and found that CCL2 was associated with both IFTA and graft dysfunction at 24 months. On univariate analysis, 6-month urinary CCL2 was a risk factor for developing 24-month IFTA, defined as ci+ct score more than 0 (odds ratio 1.045, 95% confidence interval: 1.005-1.084, P=0.028). Furthermore, CCL2 remained an independent predictor of IFTA on multivariate analysis (odds ratio 1.049, 95% confidence interval: 1.006-1.094, P=0.024) when adjusted for donor age, delayed graft function, deceased donation, and angiotensin-converting enzyme inhibitor/angiotensin receptor blocker exposure. In comparison, alpha1M, CXCL9, and CXCL10 were not associated with late graft outcomes. CONCLUSION This study demonstrates that early urinary CCL2 is an independent predictor for the subsequent development of IFTA at 24 months.
Collapse
|
34
|
Mas VR, Archer KJ, Scian M, Maluf DG. Molecular pathways involved in loss of graft function in kidney transplant recipients. Expert Rev Mol Diagn 2010; 10:269-84. [PMID: 20370585 PMCID: PMC6846360 DOI: 10.1586/erm.10.6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interstitial fibrosis (IF) and tubular atrophy (TA) are integral parts of chronic allograft dysfunction and represent in the new classification a separate entity with or without the identification of a specific etiology. Loss of kidney graft function with IF/TA is one of the causes of most kidney allograft losses. Despite progress in immunosuppression, chronic allograft dysfunction remains the main clinical challenge for improving long-term graft survival. The sustained damage to the allograft does not represent a single entity but the summated effects of tissue injury from several pathogenic insults, as well as the kidney's healing response, modified by alloimmunity and immunosuppression. A major challenge in the future of kidney transplantation includes the study of chronic allograft dysfunction pathogenesis to identify early markers of disease progression, as well as potential therapeutics pathways.
Collapse
Affiliation(s)
- Valeria R Mas
- Molecular Transplant Research Laboratory, Transplant Division, Department of Surgery, Molecular Medicine Research Building, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | | | |
Collapse
|
35
|
Mazzucco G, Magnani C, Fortunato M, Todesco A, Monga G. The reliability of pre-transplant donor renal biopsies (PTDB) in predicting the kidney state. A comparative single-centre study on 154 untransplanted kidneys. Nephrol Dial Transplant 2010; 25:3401-8. [DOI: 10.1093/ndt/gfq166] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
36
|
Servais A, Meas-Yedid V, Morelon E, Strupler M, Schanne-Klein MC, Legendre C, Olivo-Marin JC, Thervet É. Apports récents des techniques de quantification de la fibrose pour l’examen anatomopathologique en transplantation rénale. Med Sci (Paris) 2009; 25:945-50. [DOI: 10.1051/medsci/20092511945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
37
|
Abstract
Low molecular-mass plasma proteins play a key role in health and disease. Cystatin C is an endogenous cysteine proteinase inhibitor belonging to the type 2 cystatin superfamily. The mature, active form of human cystatin C is a single non-glycosylated polypeptide chain consisting of 120 amino acid residues, with a molecular mass of 13,343-13,359 Da, and containing four characteristic disulfide-paired cysteine residues. Human cystatin C is encoded by the CST3 gene, ubiquitously expressed at moderate levels. Cystatin C monomer is present in all human body fluids; it is preferentially abundant in cerebrospinal fluid, seminal plasma, and milk. Cystatin C L68Q variant is an amyloid fibril-forming protein with a high tendency to dimerize. It forms self-aggregates with massive amyloid deposits in the brain arteries of young adults, leading to lethal cerebral hemorrhage. The main catabolic site of cystatin C is the kidney: more than 99% of the protein is cleared from the circulation by glomerular ultrafiltration and tubular reabsorption. The diagnostic value of cystatin C as a marker of kidney dysfunction has been extensively investigated in multiple clinical studies on adults, children, and in the elderly. In almost all the clinical studies, cystatin C demonstrated a better diagnostic accuracy than serum creatinine in discriminating normal from impaired kidney function, but controversial results have been obtained by comparing this protein with other indices of kidney disease, especially serum creatinine-based equations. In this review, we present and discuss most of the available data from the literature, critically reviewing conclusions and suggestions for the use of cystatin C in clinical practice. Despite the multitude of clinical data in the literature, cystatin C has not been widely used, perhaps because of a combination of factors, such as a general diffidence among clinicians, the absence of definitive cut-off values, conflicting results in clinical studies, no clear evidence on when and how to request the test, the poor commutability of results, and no accurate examination of costs and of its routine use in a stat laboratory.
Collapse
Affiliation(s)
- Michele Mussap
- Department of Laboratory Medicine, University-Hospital of Padua, Padua, Italy
| | | |
Collapse
|
38
|
Abstract
Chronic allograft nephropathy, characterized by interstitial fibrosis and tubular atrophy, is still a major cause of graft loss after kidney transplantation. The complex pathophysiology of chronic allograft nephropathy is still poorly understood, and could be clarified by a more systematic performance of implantation and protocol biopsies of the renal allograft. This review highlights the contribution of implantation and protocol biopsies to our current knowledge of the complex interaction of multiple processes, ultimately leading to the development of interstitial fibrosis and tubular atrophy in the transplanted kidney. In addition, the safety and the limitations of protocol biopsies are discussed, as well as potential future directions for clinical practice and clinical research.
Collapse
|
39
|
Servais A, Meas-Yedid V, Buchler M, Morelon E, Olivo-Marin JC, Lebranchu Y, Legendre C, Thervet E. Quantification of interstitial fibrosis by image analysis on routine renal biopsy in patients receiving cyclosporine. Transplantation 2008; 84:1595-601. [PMID: 18165770 DOI: 10.1097/01.tp.0000295749.50525.bd] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Renal interstitial fibrosis (IF) is the main histopathological feature of chronic allograft injury. IF is currently assessed by semiquantitative analysis, but automatic color image analysis may be more reliable and reproducible. We performed a retrospective analysis to calculate IF on routine renal biopsies performed at 1 year posttransplant. METHODS Data were obtained from MO2ART, a prospective multicenter trial in which cyclosporine A dose was adjusted based on C2 level. One-year routine biopsies were assessed from 26 patients from two centers. For each biopsy, a section was analyzed by a program of color segmentation imaging, which automatically extracts green color areas characteristic of IF. Results were expressed as percentage of IF and grade (grade 1: <25%, grade 2: 25-50%, and grade 3: >50%). RESULTS Mean IF score was 0.35+/-0.04. Quantitative IF grade 1 was observed in 9 biopsies (34.6%), grade 2 in 12 (46.1%), and grade 3 in 5 (19.2%). Diabetes and cytomegalovirus infection were significantly associated with a higher percentage of IF. There was no correlation between the group of randomization and IF. We found a statistical significant correlation between Banff 05 chronic lesions classification and the IF index (P<0.02). Repeated analysis of variance demonstrated an association between high grade of automated IF and a worsening of creatinine clearance (Modification of Diet in Renal Disease) between 1 and 3 years. CONCLUSIONS Automatic quantification of IF on routine renal biopsies at one year posttransplant is predictive of long-term allograft function and may assist early diagnosis of the interstitial lesions of chronic allograft injury.
Collapse
Affiliation(s)
- Aude Servais
- Department of Renal Transplantation, Université Paris 5 -René Descartes, Necker Hospital, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Lerut E, Naesens M, Kuypers DR, Vanrenterghem Y, Van Damme B. Subclinical peritubular capillaritis at 3 months is associated with chronic rejection at 1 year. Transplantation 2007; 83:1416-22. [PMID: 17565313 DOI: 10.1097/01.tp.0000266676.10550.70] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Peritubular capillaritis has been associated with chronic rejection, but the characteristics of subclinical lesions in peritubular capillaries are unknown. METHODS Fifty-three renal allograft recipients underwent a protocol biopsy at both 3 and 12 months after transplantation. Subclinical chronic antibody-mediated rejection (CAMR) at 1 year was diagnosed when three or more of five criteria were present: basement membrane multilayering of peritubular capillaries (MLPTC), transplant glomerulopathy, increase in intimal fibrosis between 3 and 12 months, C4d deposition in peritubular capillaries, and the presence of anti-human leukocyte antigen antibodies. RESULTS Six (11.3%) patients met the criteria of CAMR. MLPTC was the most sensitive (83.3%) and specific (89.1%) histological criterion (P=0.0008). Five patients had peritubular capillaritis at their 3-month biopsy. They all developed MLPTC at 1 year (P<0.0001). Three of the patients with early peritubular capillaritis met the criteria of CAMR at 1 year (P=0.0002). CONCLUSIONS Through early detection of subclinical peritubular capillaritis, renal allograft recipients who are at risk for development of MLPTC might be identified. Larger series are needed to confirm these preliminary findings, but this report suggests peritubular capillaritis as an early detection marker for patients at risk for CAMR.
Collapse
Affiliation(s)
- Evelyne Lerut
- Department of Morphology and Molecular Pathology, University Hospitals Leuven, Leuven, Belgium.
| | | | | | | | | |
Collapse
|
41
|
Serón D, Moreso F. Protocol biopsies in renal transplantation: prognostic value of structural monitoring. Kidney Int 2007; 72:690-7. [PMID: 17597702 DOI: 10.1038/sj.ki.5002396] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The natural history of renal allograft damage has been characterized in serial protocol biopsies. The prevalence of subclinical rejection (SCR) is maximal during the first months and it is associated with the progression of interstitial fibrosis/tubular atrophy (IF/TA) and a decreased graft survival. IF/TA rapidly progress during the first months and constitutes an independent predictor of graft survival. IF/TA associated with transplant vasculopathy, SCR, or transplant glomerulopathy implies a poorer prognosis than IF/TA without additional lesions. These observations suggest that protocol biopsies could be considered a surrogate of graft survival. Preliminary data suggest that the predictive value of protocol biopsies is not inferior to acute rejection or renal function. Additionally, protocol biopsies have been employed as a secondary efficacy variable in clinical trials. This strategy has been useful to demonstrate a decrease in the progression of IF/TA in some calcineurin-free regimens. Quantification of renal damage is associated with graft survival suggesting that quantitative parameters might improve the predictive value of protocol biopsies. Validation of protocol biopsies as a surrogate of graft survival is actively pursued, as the utility of classical surrogates of graft outcome such as acute rejection has become less useful because of its decreased prevalence with actual immunosuppression.
Collapse
Affiliation(s)
- D Serón
- Nephrology Department, Hospital Universitari de Bellvitge, Barcelona, Spain.
| | | |
Collapse
|
42
|
Yilmaz S, Isik I, Afrouzian M, Monroy M, Sar A, Benediktsson H, McLaughlin K. Evaluating the accuracy of functional biomarkers for detecting histological changes in chronic allograft nephropathy. Transpl Int 2007; 20:608-15. [PMID: 17521383 DOI: 10.1111/j.1432-2277.2007.00494.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The most common cause of late kidney transplant failure is chronic allograft nephropathy (CAN). Much research has focused on identifying biomarkers (or correlates) that would predict subsequent CAN and allow timely intervention. Functional biomarkers such as serum creatinine and estimated glomerular filtration rate (eGFR) have been widely adopted, even though they have not been rigorously evaluated as surrogate markers. This study evaluated serum creatinine and eGFR for predicting the early histopathological changes seen in transplant protocol biopsies (TPB). We prospectively followed 289 kidney transplant patients in the Southern Alberta Transplant Program who had TPB at 6-12 months post-transplant. Tissue samples (n = 280) were independently examined by renal pathologists. The ability of serum creatinine or eGFR to predict the threshold level for abnormal histopathology was evaluated by calculating the area under the receiver operator characteristic curve. Serum creatinine and eGFR had poor predictive value (most confidence intervals included 0.5, indicating no predictive ability) for ten individual histological measurements (Banff 97 scores), and the Chronic Allograft Damage Index. We conclude that serum creatinine and eGFR have a limited clinical role in predicting the early histopathological changes that precede CAN and should not be used for this purpose.
Collapse
Affiliation(s)
- Serdar Yilmaz
- Division of Transplantation, Department of Surgery, University of Calgary, Foothills Medical Centre, 1403-29 Street NW, Calgary, Alberta, Canada.
| | | | | | | | | | | | | |
Collapse
|
43
|
Cosio FG, Amer H, Grande JP, Larson TS, Stegall MD, Griffin MD. Comparison of Low Versus High Tacrolimus Levels in Kidney Transplantation: Assessment of Efficacy by Protocol Biopsies. Transplantation 2007; 83:411-6. [PMID: 17318073 DOI: 10.1097/01.tp.0000251807.72246.7d] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The use of calcineurin inhibitors is generally guided by drug blood levels. However, those levels are chosen based on clinical experience, lacking adequate titration studies. METHODS In these analyses, we compared clinical and histologic endpoints in two groups of kidney transplant recipients: in the first (HiTAC, January 2000 to June 2002, n=245) tacrolimus levels were significantly higher than in the second (LoTAC, July 2002 to September 2004, n=330). This change in drug levels (15% reduction) was made in an attempt to reduce the incidence of polyoma virus nephropathy (PVAN). Other immunosuppressive medications were unchanged during these two time periods. RESULTS The recipient and donor demographics were not statistically different between the two groups. Compared to HiTAC, at one year posttransplant LoTAC had: 1) lower incidence of PVAN (10.5% vs. 2.5%, P<0.0001); 2) lower fasting glucose levels; 3) higher iothalamate glomerular filtration rate (52+/-19 vs. 59+/-17 ml/min/m, P<0.0001); and 4) on protocol one-year biopsies, lower incidence and severity of interstitial fibrosis (67% vs. 45%, P=0.003) and tubular atrophy (82% vs., 66%, P=0.01). The incidence and severity of acute rejection episodes was similar between both groups (7.8% versus 7.6%). CONCLUSIONS Modest reductions in tacrolimus exposure early posttransplant are associated with significant beneficial effects for the patient and the allograft without an increased immunologic risk.
Collapse
Affiliation(s)
- Fernando G Cosio
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Kambham N, Nagarajan S, Shah S, Li L, Salvatierra O, Sarwal MM. A Novel, Semiquantitative, Clinically Correlated Calcineurin Inhibitor Toxicity Score for Renal Allograft Biopsies. Clin J Am Soc Nephrol 2006; 2:135-42. [PMID: 17699397 DOI: 10.2215/cjn.01320406] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Calcineurin inhibitor toxicity (CNIT) is an important cause of chronic allograft nephropathy (CAN), but clinically relevant, diagnostic pathologic criteria remain to be defined. A semiquantitative, clinically correlative CNIT scoring system was developed and validated by pathologic analyses of 254 renal transplant biopsies that were obtained from 50 consecutive pediatric renal transplant recipients. Differentially weighted pathologic criteria (glomerulosclerosis, tubular atrophy, arteriolar medial hyaline, and tubular isometric vacuolization) contributed to the composite CNIT model score. Unlike other established pathology chronicity scores, such as the chronic allograft damage index, Banff, and modified Banff, the CNIT score was highly correlated with future graft function. The 3-mo CNIT score correlated significantly with 12 mo (P = 0.021) and 24 mo (P = 0.03) calculated creatinine clearance. Arteriolar medial hyalinosis seems to be the most important factor contributing to the clinical impact of the CNIT score.
Collapse
|
45
|
Lauronen J, Häyry P, Paavonen T. An image analysis-based method for quantification of chronic allograft damage index parameters. APMIS 2006; 114:440-8. [PMID: 16856966 DOI: 10.1111/j.1600-0463.2006.apm_350.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Chronic allograft damage index (CADI) is a semi-quantitative histopathological score that predicts renal graft outcome. We aimed to develop an objective image analysis-based method for quantification of CADI parameters. Thirty-five kidney transplant biopsies were visually analyzed according to the original CADI criteria, and divided into normal, mildly, moderately and severely altered groups. Digital images of the same samples were then analyzed with IPLab software. Areas of inflammation and fibrosis measured using image analysis increased simultaneously with corresponding visual scores, although the difference between non-inflamed and mildly inflamed groups was not statistically significant. Area of normal tubuli decreased in the images of samples with visually mild/moderate tubular atrophy and tended to be even smaller in the group with severe tubular atrophy. Image analysis-based glomerular sclerosis score increased concurrently with increasing visual score. Mesangial matrix increase score in image analysis was greater in the samples with visually mild/moderate mesangial matrix increase score compared to those with normal glomeruli, and it was highest in the group with severe mesangial matrix increase. An image analysis-based CADI scoring of renal allograft biopsies could provide more precise data for scientific studies, and help pathologists in renal allograft biopsy scoring.
Collapse
Affiliation(s)
- Jouni Lauronen
- Haartman Institute, Transplantation Laboratory, University of Helsinki, Helsinki, Finland.
| | | | | |
Collapse
|
46
|
Esquena S, Planas J, Abascal JM, Trilla E, Cecchini L, Raventós CX, Morote J. Correlation Between the Biopsies in Marginal Donor Kidneys for Transplantation: Is It Necessary to Biopsy Both Kidneys? Transplant Proc 2006; 38:1270-3. [PMID: 16797279 DOI: 10.1016/j.transproceed.2006.03.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Indexed: 10/24/2022]
Abstract
OBJECTIVES The objective of this study was to analyze the correlation between histological findings in both transplanted kidneys from marginal donors. METHODS We retrospectively reviewed the histological information on 92 kidneys obtained between January 2001 and January 2004, corresponding to 46 marginal donors. Criteria for biopsy were age greater than 55 years, hypertension, diabetes, and proteinuria. Scores were established by the pathologist including glomerulosclerosis, tubular atrophy, interstitial fibrosis, and arteriosclerosis. The score for each lesion was classified as 0 if absent; 1 if <20%; 2 if >20% and <50%; and 3 if >50%. Finally, we defined an index of renal severity damage (RSD) in order to classify the kidneys for single transplantation (0), double transplantation (1), and unsuitable for transplantation (2). RESULTS Of the kidneys studied, 82.6% of both kidneys showed similar degrees of glomerulosclerosis (<20% in 71.7% and >20% in 10.9%), while 17.4% showed discrepancies (> vs <20%; P=.008). On the other hand, RSD correlated in 82.6% of both kidneys (in 69.6% RSD=0; in 8.7% RSD=1; and in 4.3% RSD=2), while 17.4% showed discrepancies (P=.001). In one case (2.2%), a great discrepancy was observed; one kidney was valid for single transplantation, and the other one not valid for any transplantation, single or double. CONCLUSIONS This study demonstrated a correlation between the biopsy findings in both kidneys in 82.6% of marginal organ donors. However, in 17.4% of cases we observed discrepancies. The degree of glomerulosclerosis seemed to be a powerful parameter to define renal severity damage. According to these results we would recommend biopsy of both kidneys.
Collapse
Affiliation(s)
- S Esquena
- Hospital Vall d'Hebron, Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
47
|
Roos-van Groningen MC, Scholten EM, Lelieveld PM, Rowshani AT, Baelde HJ, Bajema IM, Florquin S, Bemelman FJ, de Heer E, de Fijter JW, Bruijn JA, Eikmans M. Molecular comparison of calcineurin inhibitor-induced fibrogenic responses in protocol renal transplant biopsies. J Am Soc Nephrol 2006; 17:881-8. [PMID: 16467444 DOI: 10.1681/asn.2005080891] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The calcineurin inhibitor cyclosporine (CsA) induces a fibrogenic response that may lead to scarring of the renal allograft. This study investigated whether tacrolimus, a novel calcineurin inhibitor, exerts fibrogenic effects to a similar extent. Sixty patients were enrolled in a randomized study: 29 received CsA, and 31 received tacrolimus. Patients were subjected to tailored exposure-controlled calcineurin inhibitor regimens. Protocol biopsies were obtained at the time of transplantation and 6 and 12 mo after transplantation. Cortical TGF-beta and collagens alpha1(I) and alpha1(III) mRNA steady-state levels were determined with real-time PCR. The extent of protein deposition of TGF-beta, alpha-smooth muscle actin, and interstitial collagens in the renal cortex was quantified with computer-assisted image analysis. The extent of interstitial collagen deposition measured with Sirius red and the accumulation of alpha-smooth muscle actin and TGF-beta protein after 6 and 12 mo were similar for both immunosuppressive regimens. mRNA levels of TGF-beta and collagens alpha1(I) and alpha1(III) were not significantly different in the treatment groups either. It is concluded that the fibrogenic response in renal allografts is similar in patients who receive CsA-based regimens and patients who receive tacrolimus-based regimens.
Collapse
Affiliation(s)
- Marian C Roos-van Groningen
- Leiden University Medical Center, Department of Pathology, Building 1, L1-Q, PO Box 9600, 2300 RC Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rush D. Protocol Transplant Biopsies: An Underutilized Tool in Kidney Transplantation. Clin J Am Soc Nephrol 2005; 1:138-43. [PMID: 17699200 DOI: 10.2215/cjn.00390705] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- David Rush
- University of Manitoba and Manitoba Adult Renal Transplant Program, Winnipeg, Manitoba, Canada.
| |
Collapse
|
49
|
Abstract
Studies suggest that surveillance or protocol biopsies that are performed during the first year after kidney transplantation may be clinically useful in identifying early acute rejection or chronic allograft nephropathy at a point when they may be amenable to treatment. Although the benefit of this approach has yet to be evaluated in large, multicenter, prospective trials, numerous studies suggest that implementation of protocol biopsies may improve long-term graft function. In particular, a number of reports suggest that detection of chronic allograft nephropathy in early protocol biopsies is predictive of subsequent graft function and loss and that early treatment may have a dramatic effect on the outcome of the graft. Protocol biopsies also have the potential to be of great value in high-risk patients, such as those with delayed graft function, by allowing for early intervention for acute rejection. Furthermore, the procedure seems to be relatively straightforward and safe. Nevertheless, paucity of data has meant that clear proof of a benefit of early treatment of subclinical rejection and chronic allograft nephropathy detected by protocol biopsy is lacking. Moreover, the optimal timing of protocol biopsies and reliable methods to quantify the histologic changes observed in biopsy specimens have yet to be determined. This review discusses the pros and cons of protocol biopsies and considers the place of this procedure in the routine treatment of kidney transplant patients.
Collapse
Affiliation(s)
- Alan Wilkinson
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1693, USA.
| |
Collapse
|
50
|
Cosio FG, Grande JP, Wadei H, Larson TS, Griffin MD, Stegall MD. Predicting subsequent decline in kidney allograft function from early surveillance biopsies. Am J Transplant 2005; 5:2464-72. [PMID: 16162196 DOI: 10.1111/j.1600-6143.2005.01050.x] [Citation(s) in RCA: 264] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Identifying factors that are predictive of allograft loss might be an important step toward prolonging kidney allograft survival. In this study we sought to determine the association between histologic changes on 1-year surveillance biopsies, changes in graft function and survival. This analysis included 292 adults, recipients of kidneys from living donors (69%) or deceased donors (31%), transplanted between 1998 and 2001 and followed up for 46 +/- 14 months. The primary end point was death-censored graft loss or a >50% reduction in GFR beyond 1 year. One-year biopsies were classified as: (i) Normal (N = 87, 30%), (ii) inflammation (N = 6, 2%), (iii) fibrosis (N = 131, 45%), (iv) fibrosis and inflammation (N = 53, 18%) and (v) transplant glomerulopathy (N = 15, 5%). By multivariate Cox analysis, survival related to biopsy classification (HR = 4.2, p = 0.001), graft function (HR = 0.97, p = 0.001) and HLA mismatches (HR = 1.003, p = 0.004). Using normal histology as a reference, fibrosis and inflammation (HR = 8.5, p < 0.0001) and glomerulopathy (HR = 10, p < 0.0001) related to poorer survival but mild fibrosis alone did not. Importantly, the degree of inflammation associated with fibrosis generally did not qualify for the diagnosis of borderline rejection. In conclusion, inflammation and glomerulopathy 1 year post-transplant predict loss of graft function and graft failure independently of function and other variables.
Collapse
Affiliation(s)
- Fernando G Cosio
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic and Foundation, Rochester, Minnnesota, USA.
| | | | | | | | | | | |
Collapse
|