1
|
Stabinska J, Piccolo J, Chhabra A, Liatsou I, Gabrielson K, Li Z, Mohanta Z, Sedaghat F, Hobbs RF, Sgouros G, McMahon MT. MRI detects tubulointerstitial changes in mouse models of radiation-induced nephropathy. Magn Reson Med 2025; 94:251-261. [PMID: 39846230 PMCID: PMC12052301 DOI: 10.1002/mrm.30443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025]
Abstract
PURPOSE We hypothesized that radiation-induced tubulointerstitial changes in the kidney can be assessed using MRI-based T2 relaxation time measurements. METHODS We performed MRI, histology, and serum biochemistry in two mouse models of radiation nephropathy: one involving external beam radiotherapy and the other using internal irradiation with an α-particle-emitting actinium-225 radiolabeled antibody. We compared the mean T2 values of different renal compartments between control and external beam radiotherapy or α-particle-emitting actinium-225 radiolabeled antibody-treated groups and between the two radiation-treated groups using a Wilcoxon rank-sum test. RESULTS Significantly higher T2 values were found in the cortex and outer stripe of the outer medulla in all treated animals compared with the control group (p < 0.05). In addition, these changes in T2 were observed before any changes in serum parameters, animal body weight, and kidney volume occurred. CONCLUSION T2 mapping is sensitive to radiation-induced tubulointerstitial changes in the kidney.
Collapse
Affiliation(s)
- Julia Stabinska
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joe Piccolo
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anupriya Chhabra
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ioanna Liatsou
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathy Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhi Li
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zinia Mohanta
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Farzad Sedaghat
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Robert F. Hobbs
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael T. McMahon
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Tarchi SM, Salvatore M, Lichtenstein P, Sekar T, Capaccione K, Luk L, Shaish H, Makkar J, Desperito E, Leb J, Navot B, Goldstein J, Laifer S, Beylergil V, Ma H, Jambawalikar S, Aberle D, D'Souza B, Bentley-Hibbert S, Marin MP. Radiology of fibrosis part III: genitourinary system. J Transl Med 2024; 22:616. [PMID: 38961396 PMCID: PMC11223291 DOI: 10.1186/s12967-024-05333-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
Fibrosis is a pathological process involving the abnormal deposition of connective tissue, resulting from improper tissue repair in response to sustained injury caused by hypoxia, infection, or physical damage. It can impact any organ, leading to their dysfunction and eventual failure. Additionally, tissue fibrosis plays an important role in carcinogenesis and the progression of cancer.Early and accurate diagnosis of organ fibrosis, coupled with regular surveillance, is essential for timely disease-modifying interventions, ultimately reducing mortality and enhancing quality of life. While extensive research has already been carried out on the topics of aberrant wound healing and fibrogenesis, we lack a thorough understanding of how their relationship reveals itself through modern imaging techniques.This paper focuses on fibrosis of the genito-urinary system, detailing relevant imaging technologies used for its detection and exploring future directions.
Collapse
Affiliation(s)
- Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA.
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Thillai Sekar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Kathleen Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hiram Shaish
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jasnit Makkar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Benjamin Navot
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jonathan Goldstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sherelle Laifer
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Volkan Beylergil
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Dwight Aberle
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Stuart Bentley-Hibbert
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| |
Collapse
|
3
|
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
|
4
|
Zhao WT, Herrmann KH, Sibgatulin R, Nahardani A, Krämer M, Heitplatz B, van Marck V, Reuter S, Reichenbach JR, Hoerr V. Perfusion and T 2 Relaxation Time as Predictors of Severity and Outcome in Sepsis-Associated Acute Kidney Injury: A Preclinical MRI Study. J Magn Reson Imaging 2023; 58:1954-1963. [PMID: 37026419 DOI: 10.1002/jmri.28698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Preventing sepsis-associated acute kidney injury (S-AKI) can be challenging because it develops rapidly and is often asymptomatic. Probability assessment of disease progression for therapeutic follow-up and outcome are important to intervene and prevent further damage. PURPOSE To establish a noninvasive multiparametric MRI (mpMRI) tool, including T1 , T2 , and perfusion mapping, for probability assessment of the outcome of S-AKI. STUDY TYPE Preclinical randomized prospective study. ANIMAL MODEL One hundred and forty adult female SD rats (65 control and 75 sepsis). FIELD STRENGTH/SEQUENCE 9.4T; T1 and perfusion map (FAIR-EPI) and T2 map (multiecho RARE). ASSESSMENT Experiment 1: To identify renal injury in relation to sepsis severity, serum creatinine levels were determined (31 control and 35 sepsis). Experiment 2: Animals underwent mpMRI (T1 , T2 , perfusion) 18 hours postsepsis. A subgroup of animals was immediately sacrificed for histology examination (nine control and seven sepsis). Result of mpMRI in follow-up subgroup (25 control and 33 sepsis) was used to predict survival outcomes at 96 hours. STATISTICAL TESTS Mann-Whitney U test, Spearman/Pearson correlation (r), P < 0.05 was considered statistically significant. RESULTS Severely ill septic animals exhibited significantly increased serum creatinine levels compared to controls (70 ± 30 vs. 34 ± 9 μmol/L, P < 0.0001). Cortical perfusion (480 ± 80 vs. 330 ± 140 mL/100 g tissue/min, P < 0.005), and cortical and medullary T2 relaxation time constants were significantly reduced compared to controls (41 ± 4 vs. 37 ± 5 msec in cortex, P < 0.05, 52 ± 7 vs. 45 ± 6 msec in medulla, P < 0.05). The combination of cortical T2 relaxation time constants and perfusion results at 18 hours could predict survival outcomes at 96 hours with high sensitivity (80%) and specificity (73%) (area under curve of ROC = 0.8, Jmax = 0.52). DATA CONCLUSION This preclinical study suggests combined T2 relaxation time and perfusion mapping as first line diagnostic tool for treatment planning. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Wan-Ting Zhao
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Institute of Medical Microbiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Renat Sibgatulin
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Ali Nahardani
- Heart Center Bonn, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Martin Krämer
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Barbara Heitplatz
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Veerle van Marck
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Stefan Reuter
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital Münster, Münster, Germany
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Verena Hoerr
- Institute of Medical Microbiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Heart Center Bonn, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
- Translational Research Imaging Center (TRIC), Clinic of Radiology, University of Münster, Münster, Germany
| |
Collapse
|
5
|
Ton C, Salehi S, Abasi S, Aggas JR, Liu R, Brandacher G, Guiseppi-Elie A, Grayson WL. Methods of ex vivo analysis of tissue status in vascularized composite allografts. J Transl Med 2023; 21:609. [PMID: 37684651 PMCID: PMC10492401 DOI: 10.1186/s12967-023-04379-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/21/2023] [Indexed: 09/10/2023] Open
Abstract
Vascularized composite allotransplantation can improve quality of life and restore functionality. However, the complex tissue composition of vascularized composite allografts (VCAs) presents unique clinical challenges that increase the likelihood of transplant rejection. Under prolonged static cold storage, highly damage-susceptible tissues such as muscle and nerve undergo irreversible degradation that may render allografts non-functional. Skin-containing VCA elicits an immunogenic response that increases the risk of recipient allograft rejection. The development of quantitative metrics to evaluate VCAs prior to and following transplantation are key to mitigating allograft rejection. Correspondingly, a broad range of bioanalytical methods have emerged to assess the progression of VCA rejection and characterize transplantation outcomes. To consolidate the current range of relevant technologies and expand on potential for development, methods to evaluate ex vivo VCA status are herein reviewed and comparatively assessed. The use of implantable physiological status monitoring biochips, non-invasive bioimpedance monitoring to assess edema, and deep learning algorithms to fuse disparate inputs to stratify VCAs are identified.
Collapse
Affiliation(s)
- Carolyn Ton
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA
- Translational Tissue Engineering Center, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA
| | - Sara Salehi
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA
- Translational Tissue Engineering Center, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA
| | - Sara Abasi
- Department of Biomedical Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B®), Texas A&M University, Emerging Technologies Building 3120, 101 Bizzell St, College Station, TX, 77843, USA
- Department of Electrical and Computer Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B®), Texas A&M University, Emerging Technologies Building 3120, 101 Bizzell St, College Station, TX, 77843, USA
- Media and Metabolism, Wildtype, Inc., 2325 3rd St., San Francisco, CA, 94107, USA
| | - John R Aggas
- Department of Biomedical Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B®), Texas A&M University, Emerging Technologies Building 3120, 101 Bizzell St, College Station, TX, 77843, USA
- Department of Electrical and Computer Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B®), Texas A&M University, Emerging Technologies Building 3120, 101 Bizzell St, College Station, TX, 77843, USA
- Test Development, Roche Diagnostics, 9115 Hague Road, Indianapolis, IN, 46256, USA
| | - Renee Liu
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA
- Translational Tissue Engineering Center, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Reconstructive Transplantation Program, Center for Advanced Physiologic Modeling (CAPM), Johns Hopkins University, Ross Research Building/Suite 749D, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
| | - Anthony Guiseppi-Elie
- Department of Biomedical Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B®), Texas A&M University, Emerging Technologies Building 3120, 101 Bizzell St, College Station, TX, 77843, USA.
- Department of Electrical and Computer Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B®), Texas A&M University, Emerging Technologies Building 3120, 101 Bizzell St, College Station, TX, 77843, USA.
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, USA.
- ABTECH Scientific, Inc., Biotechnology Research Park, 800 East Leigh Street, Richmond, VA, USA.
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA.
- Translational Tissue Engineering Center, Johns Hopkins University, 400 North Broadway, Smith Building 5023, Baltimore, MD, 21231, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
6
|
Nakajima K, Sugikawa A, Yasui H, Higashikawa K, Suzuki C, Natsume T, Suzuki M, Takakura H, Tomita M, Takahashi S, Hirata K, Magata Y, Kuge Y, Ogawa M. In vivo imaging of acute physiological responses after treatment of cancer with near-infrared photoimmunotherapy. Mol Imaging Biol 2023:10.1007/s11307-023-01822-9. [PMID: 37193805 DOI: 10.1007/s11307-023-01822-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/18/2023]
Abstract
PURPOSE Near-infrared photoimmunotherapy (NIR-PIT) is a new cancer phototherapy using an antibody-photosensitizer conjugate (Ab-IR700). By NIR light irradiation, Ab-IR700 forms a water-insoluble aggregation on the plasma membrane of cancer cells, leading to lethal membrane damage of cancer cells with high selectivity. However, IR700 produces singlet oxygen, which induces non-selective inflammatory responses such as edema in normal tissues around the tumor. Understanding such treatment-emergent responses is important to minimize side effects and improve clinical outcomes. Thus, in this study, we evaluated physiological responses during NIR-PIT by magnetic resonance imaging (MRI) and positron emission tomography (PET). PROCEDURES Ab-IR700 was intravenously injected into tumor-bearing mice with two tumors on the right and left sides of the dorsum. At 24 h after injection, a tumor was irradiated with NIR light. Edema formation was examined by T1/T2/diffusion-weighted MRI and inflammation was investigated by PET with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG). Because inflammation can increase vascular permeability via inflammatory mediators, we evaluated changes in oxygen levels in tumors using a hypoxia imaging probe, [18F]fluoromisonidazole ([18F]FMISO). RESULTS The uptake of [18F]FDG in the irradiated tumor was significantly decreased compared to the control tumor, indicating the impairment of glucose metabolism induced by NIR-PIT. MRI and [18F]FDG-PET images showed that inflammatory edema with [18F]FDG accumulation was present in the surrounding normal tissues of the irradiated tumor. Furthermore, [18F]FMISO accumulation in the center of the irradiated tumor was relatively low, indicating the enhancement of oxygen supply due to increased vascular permeability. In contrast, high [18F]FMISO accumulation was observed in the peripheral region, indicating enhancement of hypoxia in the region. This could be because inflammatory edema was formed in the surrounding normal tissues, which blocked blood flow to the tumor. CONCLUSIONS We successfully monitored inflammatory edema and changes in oxygen levels during NIR-PIT. Our findings on the acute physiological responses after light irradiation will help to develop effective measures to minimize the side effects in NIR-PIT.
Collapse
Affiliation(s)
- Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan
- Global Station for Biosurfaces and Drug Discovery, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Hokkaido, Japan
| | - Akiyo Sugikawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan
| | - Hironobu Yasui
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kei Higashikawa
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Chie Suzuki
- Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takahiro Natsume
- Promotion Center for Medical Collaboration & Intellectual Property, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Motofumi Suzuki
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan
| | - Hideo Takakura
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan
| | - Mayu Tomita
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan
| | - Sachi Takahashi
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan
| | - Kenji Hirata
- Department of Nuclear Medicine, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yasuhiro Magata
- Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan.
- Global Station for Biosurfaces and Drug Discovery, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Hokkaido, Japan.
| |
Collapse
|
7
|
Mei Y, Yang G, Guo Y, Zhao K, Wu S, Xu Z, Zhou S, Yan C, Seeliger E, Niendorf T, Xu Y, Feng Y. Parametric MRI Detects Aristolochic Acid Induced Acute Kidney Injury. Tomography 2022; 8:2902-2914. [PMID: 36548535 PMCID: PMC9786286 DOI: 10.3390/tomography8060243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Exposure to aristolochic acid (AA) is of increased concern due to carcinogenic and nephrotoxic effects, and incidence of aristolochic acid nephropathy (AAN) is increasing. This study characterizes renal alterations during the acute phase of AAN using parametric magnetic resonance imaging (MRI). An AAN and a control group of male Wistar rats received administration of aristolochic acid I (AAI) and polyethylene glycol (PEG), respectively, for six days. Both groups underwent MRI before and 2, 4 and 6 days after AAI or PEG administration. T2 relaxation times and apparent diffusion coefficients (ADCs) were determined for four renal layers. Serum creatinine levels (sCr) and blood urea nitrogen (BUN) were measured. Tubular injury scores (TIS) were evaluated based on histologic findings. Increased T2 values were detected since day 2 in the AAN group, but decreased ADCs and increased sCr levels and BUN were not detected until day 4. Significant linear correlations were observed between T2 of the cortex and the outer stripe of outer medulla and TIS. Our results demonstrate that parametric MRI facilitates early detection of renal injury induced by AAI in a rat model. T2 mapping may be a valuable tool for assessing kidney injury during the acute phase of AAN.
Collapse
Affiliation(s)
- Yingjie Mei
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Guixiang Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yihao Guo
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou 570311, China
| | - Kaixuan Zhao
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Shuyu Wu
- Radiotherapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Zhongbiao Xu
- Radiotherapy Center, Guangdong General Hospital, Guangzhou 510080, China
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chenggong Yan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Erdmann Seeliger
- Institute of Translational Physiology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Medical Image Processing & Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou 510515, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence & Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde, Foshan), Foshan 528399, China
- Correspondence:
| |
Collapse
|
8
|
Zhi R, Zhang XD, Hou Y, Jiang KW, Li Q, Zhang J, Zhang YD. RtNet: a deep hybrid neural network for the identification of acute rejection and chronic allograft nephropathy after renal transplantation using multiparametric MRI. Nephrol Dial Transplant 2022; 37:2581-2590. [PMID: 35020923 DOI: 10.1093/ndt/gfac005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Reliable diagnosis of the cause of renal allograft dysfunction is of clinical importance. The aim of this study is to develop a hybrid deep-learning approach for determining acute rejection (AR), chronic allograft nephropathy (CAN) and renal function in kidney-allografted patients by multimodality integration. METHODS Clinical and magnetic resonance imaging (MRI) data of 252 kidney-allografted patients who underwent post-transplantation MRI between December 2014 and November 2019 were retrospectively collected. An end-to-end convolutional neural network, namely RtNet, was designed to discriminate between AR, CAN and stable renal allograft recipient (SR), and secondarily, to predict the impaired renal graft function [estimated glomerular filtration rate (eGFR) ≤50 mL/min/1.73 m2]. Specially, clinical variables and MRI radiomics features were integrated into the RtNet, resulting in a hybrid network (RtNet+). The performance of the conventional radiomics model RtRad, RtNet and RtNet+ was compared to test the effect of multimodality interaction. RESULTS Out of 252 patients, AR, CAN and SR was diagnosed in 20/252 (7.9%), 92/252 (36.5%) and 140/252 (55.6%) patients, respectively. Of all MRI sequences, T2-weighted imaging and diffusion-weighted imaging with stretched exponential analysis showed better performance than other sequences. On pairwise comparison of resulting prediction models, RtNet+ produced significantly higher macro-area-under-curve (macro-AUC) (0.733 versus 0.745; P = 0.047) than RtNet in discriminating between AR, CAN and SR. RtNet+ performed similarly to the RtNet (macro-AUC, 0.762 versus 0.756; P > 0.05) in discriminating between eGFR ≤50 mL/min/1.73 m2 and >50 mL/min/1.73 m2. With decision curve analysis, adding RtRad and RtNet to clinical variables resulted in more net benefits in diagnostic performance. CONCLUSIONS Our study revealed that the proposed RtNet+ model owned a stable performance in revealing the cause of renal allograft dysfunction, and thus might offer important references for individualized diagnostics and treatment strategy.
Collapse
Affiliation(s)
- Rui Zhi
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiao-Dong Zhang
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Ying Hou
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ke-Wen Jiang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qiao Li
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jing Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yu-Dong Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
9
|
Liu J, Shelton EL, Crescenzi R, Colvin DC, Kirabo A, Zhong J, Delpire EJ, Yang HC, Kon V. Kidney Injury Causes Accumulation of Renal Sodium That Modulates Renal Lymphatic Dynamics. Int J Mol Sci 2022; 23:ijms23031428. [PMID: 35163352 PMCID: PMC8836121 DOI: 10.3390/ijms23031428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 01/02/2023] Open
Abstract
Lymphatic vessels are highly responsive to changes in the interstitial environment. Previously, we showed renal lymphatics express the Na-K-2Cl cotransporter. Since interstitial sodium retention is a hallmark of proteinuric injury, we examined whether renal sodium affects NKCC1 expression and the dynamic pumping function of renal lymphatic vessels. Puromycin aminonucleoside (PAN)-injected rats served as a model of proteinuric kidney injury. Sodium 23Na/1H-MRI was used to measure renal sodium and water content in live animals. Renal lymph, which reflects the interstitial composition, was collected, and the sodium analyzed. The contractile dynamics of isolated renal lymphatic vessels were studied in a perfusion chamber. Cultured lymphatic endothelial cells (LECs) were used to assess direct sodium effects on NKCC1. MRI showed elevation in renal sodium and water in PAN. In addition, renal lymph contained higher sodium, although the plasma sodium showed no difference between PAN and controls. High sodium decreased contractility of renal collecting lymphatic vessels. In LECs, high sodium reduced phosphorylated NKCC1 and SPAK, an upstream activating kinase of NKCC1, and eNOS, a downstream effector of lymphatic contractility. The NKCC1 inhibitor furosemide showed a weaker effect on ejection fraction in isolated renal lymphatics of PAN vs controls. High sodium within the renal interstitium following proteinuric injury is associated with impaired renal lymphatic pumping that may, in part, involve the SPAK-NKCC1-eNOS pathway, which may contribute to sodium retention and reduce lymphatic responsiveness to furosemide. We propose that this lymphatic vessel dysfunction is a novel mechanism of impaired interstitial clearance and edema in proteinuric kidney disease.
Collapse
Affiliation(s)
- Jing Liu
- Department of Nephrology, Tongji University School of Medicine, Shanghai 200070, China;
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Elaine L. Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Rachelle Crescenzi
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (R.C.); (D.C.C.)
| | - Daniel C. Colvin
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (R.C.); (D.C.C.)
| | - Annet Kirabo
- Department of Medicine, Division of Clinal Pharmacology and Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (A.K.); (J.Z.)
| | - Jianyong Zhong
- Department of Medicine, Division of Clinal Pharmacology and Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (A.K.); (J.Z.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Eric J. Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hai-Chun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence: (H.-C.Y.); (V.K.); Tel.: +1-615-343-0110 (H.-C.Y.); +1-615-322-7416 (V.K.)
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence: (H.-C.Y.); (V.K.); Tel.: +1-615-343-0110 (H.-C.Y.); +1-615-322-7416 (V.K.)
| |
Collapse
|
10
|
Matar AJ, Crepeau RL, Duran-Struuck R. Non-invasive imaging for the diagnosis of acute rejection in transplantation: The next frontier. Transpl Immunol 2021; 68:101431. [PMID: 34157374 DOI: 10.1016/j.trim.2021.101431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
Acute rejection is a leading cause of organ transplant failure and the most common indication for re-transplantation. Clinically, suspicion of acute rejection is often dependent upon serum laboratory values which may only manifest after organ injury. The gold standard for diagnosis requires an invasive biopsy which can carry serious clinical risks including bleeding and graft loss as well as the possibility of sampling error. The use of noninvasive imaging modalities to monitor transplanted organs is of great clinical value, particularly as a tool for early detection of graft dysfunction or acute rejection. Herein, we provide an overview of the existing literature evaluating noninvasive imaging modalities of solid organ and cellular allografts after transplantation, including both preclinical and clinical studies.
Collapse
Affiliation(s)
- Abraham J Matar
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca L Crepeau
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Raimon Duran-Struuck
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Liposomal Delivery Improves the Efficacy of Prednisolone to Attenuate Renal Inflammation in a Mouse Model of Acute Renal Allograft Rejection. Transplantation 2020; 104:744-753. [PMID: 31929419 PMCID: PMC7147400 DOI: 10.1097/tp.0000000000003060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Supplemental Digital Content is available in the text. Background. Systemic exposure to high-dose corticosteroids effectively combats acute rejection after kidney transplantation, but at the cost of substantial side effects. In this study, a murine acute renal allograft rejection model was used to investigate whether liposomal-encapsulated prednisolone (LP) facilitates local exposure to enhance its therapeutic effect. Methods. Male BalbC recipients received renal allografts from male C57BL/6J donors. Recipients were injected daily with 5 mg/kg cyclosporine A and received either 10 mg/kg prednisolone (P), or LP intravenously on day 0, 3, and 6, or no additional treatment. Functional magnetic resonance imaging (fMRI) was performed on day 6 to study allograft perfusion and organs were retrieved on day 7 for further analysis. Results. Staining of polyethylene-glycol-labeled liposomes and high performance liquid chromatography analysis revealed accumulation in the LP treated allograft. LP treatment induced the expression of glucocorticoid responsive gene Fkbp5 in the allograft. Flow-cytometry of allografts revealed liposome presence in CD45+ cells, and reduced numbers of F4/80+ macrophages, and CD3+ T-lymphocytes upon LP treatment. Banff scoring showed reduced interstitial inflammation and tubulitis and fMRI analysis revealed improved allograft perfusion in LP versus NA mice. Conclusions. Liposomal delivery of prednisolone improved renal bio-availability, increased perfusion and reduced cellular infiltrate in the allograft, when compared with conventional prednisolone. Clinical studies should reveal if treatment with LP results in improved efficacy and reduced side effects in patients with renal allograft rejection.
Collapse
|
12
|
Zhao K, Pohlmann A, Feng Q, Mei Y, Yang G, Yi P, Feng Q, Chen W, Zhou L, Wu EX, Seeliger E, Niendorf T, Feng Y. Physiological system analysis of the kidney by high-temporal-resolution T 2 ∗ monitoring of an oxygenation step response. Magn Reson Med 2020; 85:334-345. [PMID: 32710578 DOI: 10.1002/mrm.28399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/21/2020] [Accepted: 06/08/2020] [Indexed: 11/11/2022]
Abstract
PURPOSE Examine the feasibility of characterizing the regulation of renal oxygenation using high-temporal-resolution monitoring of the T 2 ∗ response to a step-like oxygenation stimulus. METHODS For T 2 ∗ mapping, multi-echo gradient-echo imaging was used (temporal resolution = 9 seconds). A step-like renal oxygenation challenge was applied involving sequential exposure to hyperoxia (100% O2 ), hypoxia (10% O2 + 90% N2 ), and hyperoxia (100% O2 ). In vivo experiments were performed in healthy rats (N = 10) and in rats with bilateral ischemia-reperfusion injury (N = 4). To assess the step response of renal oxygenation, a second-order exponential model was used (model parameters: amplitude [A], time delay [Δt], damping constant [D], and period of the oscillation [T]) for renal cortex, outer stripe of the outer medulla, inner stripe of the outer medulla, and inner medulla. RESULTS The second-order exponential model permitted us to model the exponential T 2 ∗ recovery and the superimposed T 2 ∗ oscillation following renal oxygenation stimulus. The in vivo experiments revealed a difference in Douter medulla between healthy controls (D < 1, indicating oscillatory recovery) and ischemia-reperfusion injury (D > 1, reflecting aperiodic recovery). The increase in Douter medulla by a factor of 3.7 (outer stripe of the outer medulla) and 10.0 (inner stripe of the outer medulla) suggests that this parameter might be rather sensitive to (patho)physiological oxygenation changes. CONCLUSION This study demonstrates the feasibility of monitoring the dynamic oxygenation response of renal tissues to a step-like oxygenation challenge using high-temporal-resolution T 2 ∗ mapping. Our results suggest that the implemented system analysis approach may help to unlock questions regarding regulation of renal oxygenation, with the ultimate goal of providing imaging means for diagnostics and therapy of renal diseases.
Collapse
Affiliation(s)
- Kaixuan Zhao
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Qijian Feng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingjie Mei
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Philips Healthcare, Guangzhou, China
| | - Guixiang Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| | - Peiwei Yi
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| | - Qianjin Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| | - Wufang Chen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ed X Wu
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Hong Kong, China
| | - Erdmann Seeliger
- Center for Cardiovascular Research, Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Ni X, Wang W, Li X, Li Y, Chen J, Shi D, Wen J. Utility of Diffusion-Weighted Imaging for Guiding Clinical Management of Patients With Kidney Transplant: A Prospective Study. J Magn Reson Imaging 2020; 52:565-574. [PMID: 32030832 DOI: 10.1002/jmri.27071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Although biopsy is essential for the diagnosis and management of kidney transplant recipients, it is invasive. Intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) is a noninvasive technique that can assess both capillary perfusion and tissue diffusion. PURPOSE To evaluate the capability of IVIM-DWI as a differentiation of kidney transplant patients who need clinical intervention from those who need not. STUDY TYPE Prospective. SUBJECTS In all, 33 kidney transplant patients who needed clinical intervention and 19 who need not. FIELD STRENGTH/SEQUENCE 3.0T; IVIM-DWI with a single-shot echo planar imaging sequence. ASSESSMENT All patients underwent kidney transplant biopsy and IVIM-DWI scans. Patients were dichotomized into those who needed clinical intervention (CHANGE group) and those who need not (Non-CHANGE group) based on biopsy results. The values of total apparent diffusion coefficient (ADCT ), diffusion coefficient (D), and perfusion fraction (f) were acquired from renal cortex and medulla, respectively. The area under the curve (AUC) was calculated and compared. STATISTICAL TESTS Independent Student's t-test, receiver-operating characteristic curve, and Spearman correlation analysis. RESULTS All the cortical and medullary DWI parameters in the CHANGE group were significantly lower than those in the Non-CHANGE group (all P ≤ 0.012). Except for medullary fp, all DWI parameters in both the cortex and the medulla were inversely correlated with both the chronic (ρ ranging from -0.33 to -0.54, all P ≤ 0.02) and acute (ρ ranging from -0.35 to -0.60, all P ≤ 0.01) composite scores. Cortical ADCT and D had the largest AUC and specificity of 0.84 and 75.8%, respectively. Combined use of cortical D and medullary fp at each optimal cutoff point yielded a specificity of 90.9%. DATA CONCLUSION DWI demonstrated potential as a noninvasive biomarker to allow the stratification of patients into categories in which kidney allograft biopsy results are or are not likely to change clinical management. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY STAGE: 5 J. Magn. Reson. Imaging 2020;52:565-574.
Collapse
Affiliation(s)
- Xuefeng Ni
- National Clinical Research Center of Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wei Wang
- National Clinical Research Center of Kidney Disease, Jinling Hospital, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Li
- National Clinical Research Center of Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yanjun Li
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jinsong Chen
- National Clinical Research Center of Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Donghong Shi
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiqiu Wen
- National Clinical Research Center of Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Schutter R, Lantinga VA, Borra RJH, Moers C. MRI for diagnosis of post-renal transplant complications: current state-of-the-art and future perspectives. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2019; 33:49-61. [PMID: 31879853 DOI: 10.1007/s10334-019-00813-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/27/2019] [Accepted: 11/30/2019] [Indexed: 02/07/2023]
Abstract
Kidney transplantation has developed into a widespread procedure to treat end stage renal failure, with transplantation results improving over the years. Postoperative complications have decreased over the past decades, but are still an important cause of morbidity and mortality. Early accurate diagnosis and treatment is the key to prevent renal allograft impairment or even graft loss. Ideally, a diagnostic tool should be able to detect post-transplant renal dysfunction, differentiate between the different causes and monitor renal function during and after therapeutic interventions. Non-invasive imaging modalities for diagnostic purposes show promising results. Magnetic resonance imaging (MRI) techniques have a number of advantages, such as the lack of ionizing radiation and the possibility to obtain relevant tissue information without contrast, reducing the risk of contrast-induced nephrotoxicity. However, most techniques still lack the specificity to distinguish different types of parenchymal diseases. Despite some promising outcomes, MRI is still barely used in the post-transplantation diagnostic process. The aim of this review is to survey the current literature on the relevance and clinical applicability of diagnostic MRI modalities for the detection of various types of complications after kidney transplantation.
Collapse
Affiliation(s)
- Rianne Schutter
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - Veerle A Lantinga
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ronald J H Borra
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Cyril Moers
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Rund KM, Peng S, Greite R, Claaßen C, Nolte F, Oger C, Galano JM, Balas L, Durand T, Chen R, Gueler F, Schebb NH. Dietary omega-3 PUFA improved tubular function after ischemia induced acute kidney injury in mice but did not attenuate impairment of renal function. Prostaglandins Other Lipid Mediat 2019; 146:106386. [PMID: 31698142 DOI: 10.1016/j.prostaglandins.2019.106386] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/09/2019] [Accepted: 08/28/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) is an important complication after major surgery and solid organ transplantation. Here, we present a dietary omega-3 polyunsaturated fatty acid (n3-PUFA) supplementation study to investigate whether pre-treatment can reduce ischemia induced AKI in mice. METHODS Male 12-14 week old C57BL/6 J mice received a linoleic acid rich sunflower oil based standard diet containing 10 % fat (STD) or the same diet enriched with n3-PUFA (containing 1 % EPA and 1 % DHA) (STD + n3). After 14 days of feeding bilateral 30 min renal ischemia reperfusion injury (IRI) was conducted to induce AKI and mice were sacrificed at 24 h. Serum creatinine and blood urea nitrogen (BUN) as well as liver enzyme elevation were measured. Kidney damage was analyzed by histology and immunohistochemistry. Furthermore, pro-inflammatory cytokines (IL-6, MCP-1) were determined by qPCR. FA and oxylipin pattern were quantified in blood and kidneys by GC-FID and LC-MS/MS, respectively. RESULTS n3-PUFA supplementation prior to renal IRI increased systemic and renal levels of n3-PUFA. Consistently, eicosanoids and other oxylipins derived from n3-PUFA including precursors of specialized pro-resolving mediators were elevated while n6-PUFA derived mediators such as pro-inflammatory prostaglandins were decreased. Feeding of n3-PUFA did not attenuate renal function impairment, morphological renal damage and inflammation characterized by IL-6 and MCP-1 elevation or neutrophil infiltration. However, the tubular transport marker alpha-1 microglobulin (A1M) was significantly higher expressed in proximal tubular epithelial cells of STD + n3 compared to STD fed mice. This indicates a better integrity of proximal tubular epithelial cells and thus significant protection of tubular function. In addition, heme oxygenase-1 (HO-1) which protects tubular function was also up-regulated in the treatment group receiving n3-PUFA supplemented chow. DISCUSSION We showed that n3-PUFA pre-treatment did not affect overall renal function or renal inflammation in a mouse model of moderate ischemia induced AKI, but tubular transport was improved. In conclusion, dietary n3-PUFA supplementation altered the oxylipin levels significantly but did not protect from renal function deterioration or attenuate ischemia induced renal inflammation.
Collapse
Affiliation(s)
- Katharina M Rund
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Shu Peng
- Nephrology, Hannover Medical School, Hannover, Germany; Department of Thoracic surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Robert Greite
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Cornelius Claaßen
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Fabian Nolte
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Laurence Balas
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Rongjun Chen
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Faikah Gueler
- Nephrology, Hannover Medical School, Hannover, Germany.
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany.
| |
Collapse
|
16
|
Schiffer L, Wiehler F, Bräsen JH, Gwinner W, Greite R, Kreimann K, Thorenz A, Derlin K, Teng B, Rong S, von Vietinghoff S, Haller H, Mengel M, Pape L, Lerch C, Schiffer M, Gueler F. Chemokine CXCL13 as a New Systemic Biomarker for B-Cell Involvement in Acute T Cell-Mediated Kidney Allograft Rejection. Int J Mol Sci 2019; 20:ijms20102552. [PMID: 31137652 PMCID: PMC6567305 DOI: 10.3390/ijms20102552] [Citation(s) in RCA: 14] [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: 03/26/2019] [Revised: 05/13/2019] [Accepted: 05/19/2019] [Indexed: 12/31/2022] Open
Abstract
The presence of B-cell clusters in allogenic T cell-mediated rejection (TCMR) of kidney allografts is linked to more severe disease entities. In this study we characterized B-cell infiltrates in patients with TCMR and examined the role of serum CXCL-13 in these patients and experimentally. CXCL-13 serum levels were analyzed in 73 kidney allograft recipients at the time of allograft biopsy. In addition, four patients were evaluated for CXCL13 levels during the first week after transplantation. ELISA was done to measure CXCL-13 serum levels. For further mechanistic understanding, a translational allogenic kidney transplant (ktx) mouse model for TCMR was studied in BalbC recipients of fully mismatched transplants with C57BL/6 donor kidneys. CXCL-13 serum levels were measured longitudinally, CD20 and CD3 composition and CXCL13 mRNA in tissue were examined by flow cytometry and kidneys were examined by histology and immunohistochemistry. We found significantly higher serum levels of the B-cell chemoattractant CXCL13 in patients with TCMR compared to controls and patients with borderline TCMR. Moreover, in patients with acute rejection within the first week after ktx, a >5-fold CXCL13 increase was measured and correlated with B-cell infiltrates in the biopsies. In line with the clinical findings, TCMR in mice correlated with increased systemic serum-CXCL13 levels. Moreover, renal allografts had significantly higher CXCL13 mRNA expression than isogenic controls and showed interstitial CD20+ B-cell clusters and CD3+ cell infiltrates accumulating in the vicinity of renal vessels. CXCL13 blood levels correlate with B-cell involvement in TCMR and might help to identify patients at risk of a more severe clinical course of rejection.
Collapse
Affiliation(s)
- Lena Schiffer
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
- Pediatric Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Flavia Wiehler
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | | | | | - Robert Greite
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Kirill Kreimann
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Anja Thorenz
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Katja Derlin
- Radiology, Hannover Medical School, 30625 Hannover, Germany.
| | - Beina Teng
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Song Rong
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Hermann Haller
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Michael Mengel
- Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| | - Lars Pape
- Pediatric Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Christian Lerch
- Pediatric Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| | - Mario Schiffer
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
- Nephrology and Hypertension, University Hospital Erlangen, 91054 Erlangen, Gerrmany.
| | - Faikah Gueler
- Nephrology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
17
|
Köhnke R, Kentrup D, Schütte-Nütgen K, Schäfers M, Schnöckel U, Hoerr V, Reuter S. Update on imaging-based diagnosis of acute renal allograft rejection. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2019; 9:110-126. [PMID: 31139495 PMCID: PMC6526365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 04/12/2019] [Indexed: 06/09/2023]
Abstract
Kidney transplantation is the preferred treatment for patients with end-stage renal disease. Despite effective immunosuppressants, acute allograft rejections pose a major threat to graft survival. In early stages, acute rejections are still potentially reversible, and early detection is crucial to initiate the necessary treatment options and to prevent further graft dysfunction or even loss of the complete graft. Currently, invasive core needle biopsy is the reference standard to diagnose acute rejection. However, biopsies carry the risk of graft injuries and cannot be immediately performed on patients receiving anticoagulation drugs. Therefore, non-invasive assessment of the whole organ for specific and rapid detection of acute allograft rejection is desirable. We herein provide a review summarizing current imaging-based approaches for non-invasive diagnosis of acute renal allograft rejection.
Collapse
Affiliation(s)
- Richard Köhnke
- Department of Medicine, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster48149 Muenster, Germany
| | - Dominik Kentrup
- Department of Medicine, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster48149 Muenster, Germany
- Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham (UAB)35294 Birmingham Alabama, US
| | - Katharina Schütte-Nütgen
- Department of Medicine, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster48149 Muenster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital of Muenster48149 Muenster, Germany
- European Institute for Molecular Imaging, University of Muenster48140 Muenster, Germany
| | - Uta Schnöckel
- Department of Nuclear Medicine, University Hospital of Muenster48149 Muenster, Germany
| | - Verena Hoerr
- Department of Clinical Radiology, University Hospital of Muenster48149 Muenster, Germany
- Institute of Medical Microbiology, Jena University HospitalAm Klinikum 1, 07747 Jena, Germany
| | - Stefan Reuter
- Department of Medicine, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Muenster48149 Muenster, Germany
| |
Collapse
|
18
|
Renal Allograft Rejection: Noninvasive Ultrasound- and MRI-Based Diagnostics. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:3568067. [PMID: 31093027 PMCID: PMC6481101 DOI: 10.1155/2019/3568067] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
To date, allogeneic kidney transplantation remains the best available therapeutic option for patients with end-stage renal disease regarding overall survival and quality of life. Despite the advancements in immunosuppressive drugs and protocols, episodes of acute allograft rejection, a sterile inflammatory process, continue to endanger allograft survival. Since effective treatment for acute rejection episodes is available, instant diagnosis of this potentially reversible graft injury is imperative. Although histological examination by invasive core needle biopsy of the graft remains the gold standard for the diagnosis of ongoing rejection, it is always associated with the risk of causing substantial graft injury as a result of the biopsy procedure itself. At the same time, biopsies are not immediately feasible for a considerable number of patients taking anticoagulants due to the high risk of complications such as bleeding and uneven distribution of pathological changes within the graft. This can result in the wrong diagnosis due to the small size of the tissue sample taken. Therefore, there is a need for a tool that overcomes these problems by being noninvasive and capable of assessing the whole organ at the same time for specific and fast detection of acute allograft rejection. In this article, we review current state-of-the-art approaches for noninvasive diagnostics of acute renal transplant inflammation, i.e., rejection. We especially focus on nonradiation-based methods using magnetic resonance imaging (MRI) and ultrasound.
Collapse
|
19
|
Waiczies S, Rosenberg JT, Kuehne A, Starke L, Delgado PR, Millward JM, Prinz C, Dos Santos Periquito J, Pohlmann A, Waiczies H, Niendorf T. Fluorine-19 MRI at 21.1 T: enhanced spin-lattice relaxation of perfluoro-15-crown-5-ether and sensitivity as demonstrated in ex vivo murine neuroinflammation. MAGMA (NEW YORK, N.Y.) 2019; 32:37-49. [PMID: 30421250 PMCID: PMC6514110 DOI: 10.1007/s10334-018-0710-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/16/2018] [Accepted: 10/18/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Fluorine MR would benefit greatly from enhancements in signal-to-noise ratio (SNR). This study examines the sensitivity gain of 19F MR that can be practically achieved when moving from 9.4 to 21.1 T. MATERIALS AND METHODS We studied perfluoro-15-crown-5-ether (PFCE) at both field strengths (B0), as a pure compound, in the form of nanoparticles (NP) as employed to study inflammation in vivo, as well as in inflamed tissue. Brains, lymph nodes (LNs) and spleens were obtained from mice with experimental autoimmune encephalomyelitis (EAE) that had been administered PFCE NPs. All samples were measured at both B0 with 2D-RARE and 2D-FLASH using 19F volume radiofrequency resonators together. T1 and T2 of PFCE were measured at both B0 strengths. RESULTS Compared to 9.4 T, an SNR gain of > 3 was observed for pure PFCE and > 2 for PFCE NPs at 21.1 T using 2D-FLASH. A dependency of 19F T1 and T2 relaxation on B0 was demonstrated. High spatially resolved 19F MRI of EAE brains and LNs at 21.1 T revealed signals not seen at 9.4 T. DISCUSSION Enhanced SNR and T1 shortening indicate the potential benefit of in vivo 19F MR at higher B0 to study inflammatory processes with greater detail.
Collapse
Affiliation(s)
- Sonia Waiczies
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany.
| | - Jens T Rosenberg
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA
| | | | - Ludger Starke
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Paula Ramos Delgado
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Jason M Millward
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Christian Prinz
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Joao Dos Santos Periquito
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Andreas Pohlmann
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | | | - Thoralf Niendorf
- Experimental Ultrahigh Field MRI, Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, 13125, Berlin, Germany
- MRI TOOLS GmbH, Berlin, Germany
- Experimental and Clinical Research Center, A Joint Cooperation Between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
20
|
Using functional magnetic resonance imaging to evaluate an acute allograft rejection model in rats. Magn Reson Imaging 2019; 58:24-31. [PMID: 30630071 DOI: 10.1016/j.mri.2019.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/03/2018] [Accepted: 01/06/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE To assess the longitudinal changes of allograft pathophysiology by intravoxel incoherent motion (IVIM) and blood oxygen level-dependent (BOLD) MRI in a rat model of acute renal allograft rejection. MATERIALS AND METHODS Acute rejection (AR) was induced by transplantation of Dark Agouti donor kidneys into Lewis recipients (n = 18). A Lewis-Lewis rat syngeneically transplanted (sTX) model served as the control (n = 6). Acute tubular necrosis (n = 6) and acute calcineurin inhibitor toxicity (n = 6) groups were established using Lewis rats. MRI was performed on postoperative day (POD) 1, 4 and 7 in the allogeneically transplanted (aTX) group and on POD4 in the other groups. Histological evaluation and PCR were performed. RESULTS After the allogenic transplantation, all MRI parameters of allograft further decreased until POD7, and the D and ADC values in the cortex were significantly lower than that in the sTX group (1.03 ± 0.09 vs 1.52 ± 0.09 × 10-3 mm2/s, Padj < 0.05; 1.21 ± 0.03 vs 1.78 ± 0.07 × 10-3 mm2/s, Padj < 0.05). The D*, f and R2* values of the aTX group in the cortex and medulla were significantly lower than those in the sTX group on POD7 (cortex, D*: 25.60 ± 4.78 vs 69.32 ± 9.79 × 10-3 mm2/s, Padj < 0.05; f: 7.84 ± 1.83 vs 20.34 ± 3.08%, Padj < 0.05; R2*: 16.61 ± 4.18 vs 31.48 ± 6.43 1/s, Padj < 0.05; medulla, D*: 13.59 ± 6.08 vs 62.75 ± 9.20 × 10-3 mm2/s, Padj < 0.05; f: 7.46 ± 1.62 vs 14.68 ± 2.05%, Padj < 0.05; R2*: 21.59 ± 3.45 vs 39.53 ± 4.34 1/s, Padj < 0.05). AR grafts presented serve interstitial inflammation, tubulitis and infiltration of T-lymphocytes and macrophages. The MRI parameters, including D, ADC, D*, f and R2*, were significantly correlated with the histological changes, cell infiltration and inflammatory cytokine mRNA levels. CONCLUSIONS IVIM coupled with BOLD MRI allows longitudinal assessment of allograft diffusion, perfusion and oxygen consumption impairment caused by acute renal allograft rejection in rat model.
Collapse
|
21
|
Baban B, Marchetti C, Khodadadi H, Malik A, Emami G, Lin PC, Arbab AS, Riccardi C, Mozaffari MS. Glucocorticoid-Induced Leucine Zipper Promotes Neutrophil and T-Cell Polarization with Protective Effects in Acute Kidney Injury. J Pharmacol Exp Ther 2018; 367:483-493. [PMID: 30301736 DOI: 10.1124/jpet.118.251371] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/01/2018] [Indexed: 12/11/2022] Open
Abstract
The glucocorticoid-induced leucine zipper (GILZ) mediates anti-inflammatory effects of glucocorticoids. Acute kidney injury (AKI) mobilizes immune/inflammatory mechanisms, causing tissue injury, but the impact of GILZ in AKI is not known. Neutrophils play context-specific proinflammatory [type 1 neutrophil (N1)] and anti-inflammatory [type 2 neutrophil (N2)] functional roles. Also, regulatory T lymphocytes (Tregs) and regulatory T-17 (Treg17) cells exert counterinflammatory effects, including the suppression of effector T lymphocytes [e.g., T-helper (Th) 17 cells]. Thus, utilizing cell preparations of mice kidneys subjected to AKI or sham operation, we determined the effects of GILZ on T cells and neutrophil subtypes in the context of its renoprotective effect; these studies used the transactivator of transcription (TAT)-GILZ or the TAT peptide. AKI increased N1 and Th-17 cells but reduced N2, Tregs, and Treg17 cells in association with increased interleukin (IL)-17+ but reduced IL-10+ cells accompanied with the disruption of mitochondrial membrane potential (ψ m) and increased apoptosis/necrosis compared with sham kidneys. TAT-GILZ, compared with TAT, treatment reduced N1 and Th-17 cells but increased N2 and Tregs, without affecting Treg17 cells, in association with a reduction in IL-17+ cells but an increase in IL-10+ cells; TAT-GILZ caused less disruption of ψ m and reduced cell death in AKI. Importantly, TAT-GILZ increased perfusion of the ischemic-reperfused kidney but reduced tissue edema compared with TAT. Utilizing splenic T cells and bone marrow-derived neutrophils, we further showed marked reduction in the proliferation of Th cells in response to TAT-GILZ compared with response to TAT. Collectively, the results indicate that GILZ exerts renoprotection accompanied by the upregulation of the regulatory/suppressive arm of immunity in AKI, likely via regulating cross talk between T cells and neutrophils.
Collapse
Affiliation(s)
- Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Cristina Marchetti
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Aneeq Malik
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Golnaz Emami
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Ping-Chang Lin
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Ali S Arbab
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Carlo Riccardi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Mahmood S Mozaffari
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| |
Collapse
|
22
|
Natanov R, Gueler F, Falk CS, Kühn C, Maus U, Boyle EC, Siemeni T, Knoefel AK, Cebotari S, Haverich A, Madrahimov N. Blood cytokine expression correlates with early multi-organ damage in a mouse model of moderate hypothermia with circulatory arrest using cardiopulmonary bypass. PLoS One 2018; 13:e0205437. [PMID: 30308065 PMCID: PMC6181365 DOI: 10.1371/journal.pone.0205437] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiopulmonary bypass (CPB) with moderate hypothermic cardiac arrest (MHCA) is essential for prolonged complex procedures in cardiac surgery and is associated with postoperative complications. Although cytokine release provoked through MHCA under CPB plays a pivotal role in postoperative organ damage, the pathomechanisms are unclear. Here, we investigated the cytokine release pattern and histological organ damage after MHCA using a recently described mouse CPB model. Eight BALB/c mice underwent 60 minutes of circulatory arrest under CPB, were successively rewarmed and reperfused. Blood cytokine concentrations and liver and kidney function parameters were measured and histological changes to these organs were compared to control animals. Our results showed a marked increase in proinflammatory cytokines and histological changes in the kidney, lung, and liver after CPB. Furthermore, clinical chemistry showed signs of hemolysis and acute kidney injury. These results suggest early onset of solid organ injury which correlates with increased leukocyte infiltration. A better understanding of the interplay between pro-inflammatory cytokine activation and solid organ injury in this model of CBP with MHCA will inform strategies to reduce organ damage during cardiac surgeries in the clinic.
Collapse
Affiliation(s)
- Ruslan Natanov
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Faikah Gueler
- Department of Nephrology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Christine S. Falk
- Institute of Transplant Immunology, Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Christian Kühn
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Ulrich Maus
- Department of Pneumology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Erin C. Boyle
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Thierry Siemeni
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Ann-Katrin Knoefel
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Serghei Cebotari
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Nodir Madrahimov
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
- * E-mail:
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW In this article, we describe the basics of how magnetic resonance urography (MRU) is performed in the pediatric population as well as the common indications and relative performance compared to standard imaging modalities. RECENT FINDINGS Although MRU is still largely performed in major academic or specialty imaging centers, more and more applications in the pediatric setting have been described in the literature. MRU is a comprehensive imaging modality for evaluating multiple pediatric urologic conditions combining excellent anatomic detail with functional information previously only available via renal scintigraphy. While generally still reserved for problem solving, MRU should be considered for some conditions as an early imaging technique.
Collapse
|
24
|
Selby NM, Blankestijn PJ, Boor P, Combe C, Eckardt KU, Eikefjord E, Garcia-Fernandez N, Golay X, Gordon I, Grenier N, Hockings PD, Jensen JD, Joles JA, Kalra PA, Krämer BK, Mark PB, Mendichovszky IA, Nikolic O, Odudu A, Ong ACM, Ortiz A, Pruijm M, Remuzzi G, Rørvik J, de Seigneux S, Simms RJ, Slatinska J, Summers P, Taal MW, Thoeny HC, Vallée JP, Wolf M, Caroli A, Sourbron S. Magnetic resonance imaging biomarkers for chronic kidney disease: a position paper from the European Cooperation in Science and Technology Action PARENCHIMA. Nephrol Dial Transplant 2018; 33:ii4-ii14. [PMID: 30137584 PMCID: PMC6106645 DOI: 10.1093/ndt/gfy152] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Indexed: 12/13/2022] Open
Abstract
Functional renal magnetic resonance imaging (MRI) has seen a number of recent advances, and techniques are now available that can generate quantitative imaging biomarkers with the potential to improve the management of kidney disease. Such biomarkers are sensitive to changes in renal blood flow, tissue perfusion, oxygenation and microstructure (including inflammation and fibrosis), processes that are important in a range of renal diseases including chronic kidney disease. However, several challenges remain to move these techniques towards clinical adoption, from technical validation through biological and clinical validation, to demonstration of cost-effectiveness and regulatory qualification. To address these challenges, the European Cooperation in Science and Technology Action PARENCHIMA was initiated in early 2017. PARENCHIMA is a multidisciplinary pan-European network with an overarching aim of eliminating the main barriers to the broader evaluation, commercial exploitation and clinical use of renal MRI biomarkers. This position paper lays out PARENCHIMA's vision on key clinical questions that MRI must address to become more widely used in patients with kidney disease, first within research settings and ultimately in clinical practice. We then present a series of practical recommendations to accelerate the study and translation of these techniques.
Collapse
Affiliation(s)
- Nicholas M Selby
- Centre for Kidney Research and Innovation, University of Nottingham, UK
| | - Peter J Blankestijn
- Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, RWTH University, Aachen, Germany
| | - Christian Combe
- Service de Néphrologie Transplantation Dialyse Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Eli Eikefjord
- Department of Health and Functioning, Western Norway University of Applied Sciences, Norway
| | | | - Xavier Golay
- Institute of Neurology, University College London, Queen Square, London, UK
| | - Isky Gordon
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Nicolas Grenier
- Service d'Imagerie Diagnostique et Interventionnelle de l'Adulte, Centre Hospitalier Universitaire de Bordeaux Place Amelie Raba-Leon, Bordeaux, France
| | | | - Jens D Jensen
- Departments of Renal and Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jaap A Joles
- Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Philip A Kalra
- Department of Renal Medicine, Salford Royal Hospital and Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Bernhard K Krämer
- Vth Department of Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University Heidelberg, Mannheim, Germany
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Iosif A Mendichovszky
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, UK
| | - Olivera Nikolic
- Faculty of Medicine,University of Novi Sad, Center of Radiology, Clinical Centre of Vojvodina, Serbia
| | - Aghogho Odudu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Albert C M Ong
- Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, UK
| | - Alberto Ortiz
- Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
| | - Menno Pruijm
- Service of Nephrology and Hypertension, Department of Medicine, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Giuseppe Remuzzi
- IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Jarle Rørvik
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Sophie de Seigneux
- Service of Nephrology, Department of Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Roslyn J Simms
- Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, UK
| | - Janka Slatinska
- Department of Nephrology, Transplant Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Paul Summers
- Department of Medical Imaging and Radiation Sciences, Radiology Division, European Institute of Oncology (IEO), Milan, Italy
- QMRI Tech iSrl, Piazza dei Martiri Pennesi 20, Pescara, Italy
| | - Maarten W Taal
- Centre for Kidney Research and Innovation, University of Nottingham, UK
| | - Harriet C Thoeny
- University of Bern, Inselspital, Bern, Switzerland
- HFR Fribourg, Hôpital Cantonal, Fribourg, Switzerland
| | - Jean-Paul Vallée
- Radiology Department, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Marcos Wolf
- Center for Medical Physics and Biomedical Engineering, MR-Centre of Excellence, Medical University of Vienna, Vienna, Austria
| | - Anna Caroli
- IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Steven Sourbron
- Leeds Imaging Biomarkers Group, Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
25
|
Schley G, Jordan J, Ellmann S, Rosen S, Eckardt KU, Uder M, Willam C, Bäuerle T. Multiparametric magnetic resonance imaging of experimental chronic kidney disease: A quantitative correlation study with histology. PLoS One 2018; 13:e0200259. [PMID: 30011301 PMCID: PMC6047786 DOI: 10.1371/journal.pone.0200259] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 06/24/2018] [Indexed: 12/27/2022] Open
Abstract
Objectives In human chronic kidney disease (CKD) the extent of renal tubulointerstitial fibrosis correlates with progressive loss of renal function. However, fibrosis can so far only be assessed by histology of kidney biopsies. Magnetic resonance imaging (MRI) can provide information about tissue architecture, but its potential to assess fibrosis and inflammation in diseased kidneys remains poorly defined. Materials and methods We evaluated excised kidneys in a murine adenine-induced nephropathy model for CKD by MRI and correlated quantitative MRI parameters (T1, T2, and T2* relaxation times, apparent diffusion coefficient and fractional anisotropy) with histological hallmarks of progressive CKD, including renal fibrosis, inflammation, and microvascular rarefaction. Furthermore, we analyzed the effects of paraformaldehyde fixation on MRI parameters by comparing kidney samples before and after fixation with paraformaldehyde. Results In diseased kidneys T2 and T2* relaxation times, apparent diffusion coefficient and fractional anisotropy in the renal cortex and/or outer medulla were significantly different from those in control kidneys. In particular, T2 relaxation time was the best parameter to distinguish control and CKD groups and correlated very well with the extent of fibrosis, inflammatory infiltrates, tubular dilation, crystal deposition, and loss of peritubular capillaries and normal tubules in the renal cortex and outer medulla. Fixation with paraformaldehyde had no impact on T2 relaxation time and fractional anisotropy, whereas T1 times significantly decreased and T2* times and apparent diffusion coefficients increased in fixed kidney tissue. Conclusions MRI parameters provide a promising approach to quantitatively assess renal fibrosis and inflammation in CKD. Especially T2 relaxation time correlates well with histological features of CKD and is not influenced by paraformaldehyde fixation of kidney samples. Thus, T2 relaxation time might be a candidate parameter for non-invasive assessment of renal fibrosis in human patients.
Collapse
Affiliation(s)
- Gunnar Schley
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- * E-mail:
| | - Jutta Jordan
- Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Stephan Ellmann
- Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Seymour Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité –Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Uder
- Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Carsten Willam
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
26
|
Abstract
KEY POINTS • The number of publications on imaging and kidney transplantation is low. • These publications are poorly cited, as compared with other fields of imaging. • Conversely, there is a clinical need for evidence-based recommendations. • Innovative advances for the use of imaging and kidney transplantation are essential. • An increased focus and adequate research funding are highly anticipated by clinicians.
Collapse
|
27
|
Zhang B, Dong Y, Guo B, Chen W, Ouyang F, Lian Z, Liu J, Zhang S. Application of noninvasive functional imaging to monitor the progressive changes in kidney diffusion and perfusion in contrast-induced acute kidney injury rats at 3.0 T. Abdom Radiol (NY) 2018; 43:655-662. [PMID: 28677006 DOI: 10.1007/s00261-017-1247-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Contrast-induced acute kidney injury is a prevalent cause of renal failure, and the noninvasive tools to monitor its progress are lacking. We applied intravoxel incoherent motion (IVIM) DWI to measure the progressive changes in kidney diffusion and perfusion of CI-AKI. METHODS Twenty-four rats received Iopromide (370 mg/ml, 1600 mg iodine/kg) to induce CI-AKI. IVIM DWI was performed on rats (n = 6) at 24 h prior to and 12, 24, 48, 72, and 96 h after the injection using a 3.0 T MRI scanner. The progressive changes in the diffusion (D) and perfusion parameters (D* and f) were studied in the cortex (CO), outer medulla (OM), and inner medulla (IM). For the histology group (n = 18), three rats were sacrificed at each time point. RESULTS In the CO, D reduced progressively from 24 to 48 h (P < 0.001) and increased starting from 72 h (P < 0.001). However, D decreased until to 72 h in the medulla (P < 0.001) and increased starting from 96 h (P < 0.001). D* decreased to the bottom at 24 h in the cortex and medulla (P = 0.037) and started to recover at 48 h (P = 0.007). f decreased in the cortex and medulla in an early stage (12 h) (P = 0.035) of CI-AKI and then ascended in the later stage (72 h) (P = 0.017). The H & E staining showed different degrees of serial pathological change including cloudy swelling, atrophy, even necrosis, and interstitial vasodilation of tubule epithelial cells and glomerulus cells. CONCLUSION Our study demonstrates the feasibility of using IVIM DWI to monitor the progress of CI-AKI, implying that IVIM DWI is a useful biomarker in the staging of CI-AKI.
Collapse
|
28
|
Abstract
RELATION Iodide mumps is an uncommon condition, induced by iodide-containing contrast, and is characterized by a rapid, painless enlargement of the bilateral or unilateral salivary gland. At present, the pathogenesis of iodide mumps is not yet clear. It may be related to an idiosyncratic reaction, a toxic accumulation of iodine in the gland duct, or renal function damage leading to an iodine excretion disorder. This paper reports the clinical manifestations and magnetic resonance imaging results of one case of iodide mumps, which occurred after digital subtraction angiography. PATIENT CONCERNS A 66-year-old Chinese man presented to our department with a 1-month speech barrier and 1 day of vomiting. He had the history of high blood sugar, the history of high blood pressure and the history of Vitiligo. He had no history of allergies and had never previously received iodide-containing contrast. His renal function and other laboratory examinations were normal. During the digital subtraction angiography (DSA), the patient received approximately 130 mL of nonionic contrast agent (iodixanol). Five hours postsurgery, the patient experienced bilateral parotid enlargement with no other discomfort, such as pain, fever, skin redness, itching, hives, nausea, vomiting, or respiratory abnormalities. DIAGNOSES We thought the diagnosis was iodide mumps. INTERVENTION Intravenous dexamethasone (5 mg) was administered. OUTCOME 20 hours post-DSA, after which the bilateral parotid shrunk. By 4 days postsurgery, the patient's bilateral parotid had recovered completely. LESSONS We found no obvious abnormal sequence signal in diffusion magnetic resonance imaging or the corresponding apparent diffusion coefficient. Our findings suggest that vasogenic edema may play an important role in the pathogenesis of iodide mumps.
Collapse
|
29
|
Li B, Xu A, Wang N, Min X, Feng Z, Deng M, Li L, Cai J, Kang Z, Jiang K, Kuang D, Wang L. Benign prostatic hyperplasia after prostatic arterial embolization in a canine model: A 3T multiparametric MRI and whole-mount step-section pathology correlated longitudinal study. J Magn Reson Imaging 2017; 46:1220-1229. [PMID: 28182304 DOI: 10.1002/jmri.25654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/17/2017] [Indexed: 01/10/2023] Open
Abstract
PURPOSE To explore the morphological and functional characteristics of prostatic arterial embolization (PAE) in a canine model of benign prostatic hyperplasia (BPH) with 3T multiparametric magnetic resonance imaging (mp-MRI) and whole-mount step-section pathology correlation. MATERIALS AND METHODS Eight adult male beagle dogs with hormone-induced BPH underwent 3T mp-MRI before and 1, 3, and 6 months after PAE, with subsequent whole-mount step-section pathologic assessment. Images were acquired using T1 -weighted images (T1 WI), T2 WI, 3D-SPACE, diffusion-weighted imaging (DWI), susceptibility-weighted imaging (SWI), T2 -mapping, and dynamic contrast-enhanced (DCE) sequences. Variance analysis was performed to assess statistical differences in prostatic volume (PV), apparent diffusion coefficient (ADC), and T2 values. Pearson correlation analysis was performed to correlate ADC, T2 , and PV. RESULTS The PV decreased from baseline to 1, 3, and 6 months after PAE from (25.88 ± 7.09) cm3 to (6.48 ± 2.08) cm3 , (6.48 ± 3.39) cm3 , (6.20 ± 2.88) cm3 . The ADC values sequentially decreased from baseline to 1, 3, and 6 months after PAE from (1497.06 ± 222.72) × 10-6 mm2 /s to (1056.00 ± 189.46) × 10-6 mm2 /s, (950.48 ± 77.85) × 10-6 mm2 /s, (980.98 ± 107.78) × 10-6 mm2 /s. The T2 values decreased from baseline to 1, 3, and 6 months after PAE were (83.74 ± 5.29) msec, (68.72 ± 5.66) msec, (53.96 ± 15.04) msec, (49.81 ± 13.34) msec, respectively. ADC and T2 values were positively correlated with PV (r = 0.823 and 0.744, respectively). Microhemorrhages and hemosiderin were found on SWI after PAE. CONCLUSION 3T mp-MRI may facilitate noninvasive assessment of morphological and functional changes of BPH after PAE. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2017;46:1220-1229.
Collapse
Affiliation(s)
- Basen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Anhui Xu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Nan Wang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiangde Min
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhaoyan Feng
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ming Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Liang Li
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Jie Cai
- Department of Interventional Radiology, First People's Hospital of Jingzhou, Yangtze University, Jingzhou, P.R. China
| | - Zhen Kang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Kehua Jiang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Dong Kuang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Liang Wang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|