1
|
Wu Z, Hu J, Li Y, Yao X, Ouyang S, Ren K. Assessment of renal pathophysiological processes and protective effect of quercetin on contrast-induced acute kidney injury in type 1 diabetic mice using diffusion tensor imaging. Redox Rep 2024; 29:2398380. [PMID: 39284588 PMCID: PMC11407404 DOI: 10.1080/13510002.2024.2398380] [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] [Indexed: 09/19/2024] Open
Abstract
Purpose: To investigate the renal pathophysiological processes and protective effect of quercetin on contrast-induced acute kidney injury (CI-AKI) in mice with type 1 diabetic mellitus(DM) using diffusion tensor imaging(DTI).Methods: Mice with DM were divided into two groups. In the diabetic + contrast medium(DCA) group, the changes of the mice kidneys were monitored at 1, 24, 48, and 72 h after the injection of iodixanol(4gI/kg). The mice in the diabetic + contrast medium + quercetin(DCA + QE) group were orally given different concentrations of quercetin for seven days before injection of iodixanol. In vitro experiments, renal tubular epithelial (HK-2) cells exposed to high glucose conditions were treated with various quercetin concentrations before treatment with iodixanol(250 mgI/mL).Results: DTI-derived mean diffusivity(MD) and fractional anisotropy(FA) values can be used to evaluate CI-AKI effectively. Quercetin significantly increased the expression of Sirt 1 and reduced oxidative stress by increasing Nrf 2/HO-1/SOD1. The antiapoptotic effect of quercetin on CI-AKI was revealed by decreasing proteins level and by reducing the number of apoptosis-positive cells. In addition, flow cytometry indicated quercetin-mediated inhibition of M1 macrophage polarization in the CI-AKI.Conclusions: DTI will be an effective noninvasive tool in diagnosing CI-AKI. Quercetin attenuates CI-AKI on the basis of DM through anti-oxidative stress, apoptosis, and inflammation.
Collapse
Affiliation(s)
- Ziqian Wu
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen Radiological Control Center, Xiamen, People's Republic of China
| | - Jingyi Hu
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen Radiological Control Center, Xiamen, People's Republic of China
| | - Yanfei Li
- Cell Therapy Research Center, Xiamen Humanity Hospital, Xiamen, People's Republic of China
| | - Xiang Yao
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Siyu Ouyang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ke Ren
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen Radiological Control Center, Xiamen, People's Republic of China
| |
Collapse
|
2
|
Chen X, Ge C, Zhang Y, Ma Y, Zhang Y, Li B, Chu Z, Ji Q. Evaluation of Early Renal Changes in Type 2 Diabetes Mellitus Using Multiparametric MR Imaging. Magn Reson Med Sci 2024:mp.2023-0148. [PMID: 39370295 DOI: 10.2463/mrms.mp.2023-0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024] Open
Abstract
PURPOSE To evaluate the clinical value of early renal changes in type 2 diabetes mellitus (T2DM) using multiparameter MRI. METHODS The study included 41 diabetics (normoalbuminuria: n = 23; microalbuminuria: n = 18) and 30 healthy controls. All subjects underwent intravoxel incoherent motion diffusion-weighted imaging (IVIM), blood oxygen level dependent (BOLD) and arterial spin labeling (ASL) examinations. One-way analysis of variance was used to compare MRI parameters among the three groups. Pearson correlation analysis was used to evaluate the relationship between MRI parameters and estimated glomerular filtration rate (eGFR) and albumin-creatinine ratio (ACR). Receiver operating characteristic analysis was performed to assess the diagnostic performance. RESULTS There were statistical differences in cortical D, D*, f, renal blood flow (RBF) and medulla D, D*, f, R2* among the three groups (P < 0.05). The cortical or medullary D, cortical f, and RBF were significantly positively correlated with eGFR (all P < 0.01). The cortical or medullary D, D*, f, cortical RBF were negatively correlated with ACR (all P < 0.05).To evaluate early kidney changes and degree of diabetes, cortical combined D and RBF (AUC [area under the curve] = 0.796 and 0.947, respectively) was better than single D or RBF (all P > 0.05); medullary combined D and R2* (AUC = 0.899 and 0.923, respectively) was better than single D or R2* (all P > 0.05), except single D (P = 0.005) in differentiating normoalbuminuria group from control group. CONCLUSION The early changes of renal diffusion and perfusion, oxygenation level, and blood flow in T2DM could be evaluated noninvasively and quantitatively using IVIM, BOLD and ASL. Renal medullary combined IVIM-derived D and BOLD-derived R2* and cortical combined IVIM-derived D and ASL-derived RBF were better for evaluating early renal changes in T2DM.
Collapse
Affiliation(s)
- Xinyi Chen
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Department of Radiology, Tianjin First Central Hospital, Tianjin, China
| | - Chao Ge
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yuling Zhang
- Department of Radiology, Traditional Chinese Medicine Hospital of Gaoling District, Xi'an, Shaanxi, China
| | - Yajie Ma
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Department of Radiology, Tianjin First Central Hospital, Tianjin, China
| | - Yuling Zhang
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Department of Radiology, Tianjin First Central Hospital, Tianjin, China
| | - Bei Li
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Department of Radiology, Tianjin First Central Hospital, Tianjin, China
| | - Zhiqiang Chu
- Department of Nephrology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Qian Ji
- Department of Radiology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
3
|
Hillaert A, Sanmiguel Serpa LC, Bogaert S, Broeckx BJG, Hesta M, Vandermeulen E, Germonpré J, Stock E, Pullens P, Vanderperren K. Assessment of pharmacologically induced changes in canine kidney function by multiparametric magnetic resonance imaging and contrast enhanced ultrasound. Front Vet Sci 2024; 11:1406343. [PMID: 38966564 PMCID: PMC11223176 DOI: 10.3389/fvets.2024.1406343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024] Open
Abstract
IntroductionDynamic contrast-enhanced (DCE) MRI and arterial spin labeling (ASL) MRI enable non-invasive measurement of renal blood flow (RBF), whereas blood oxygenation level-dependent (BOLD) MRI enables non-invasive measurement of the apparent relaxation rate (R2*), an indicator of oxygenation. This study was conducted to evaluate the potential role of these MRI modalities in assessing RBF and oxygenation in dogs. The correlation between contrast-enhanced ultrasound (CEUS) and the MRI modalities was examined and also the ability of the MRI modalities to detect pharmacologically induced changes.MethodsRBF, using CEUS, ASL- and DCE-MRI, as well as renal oxygenation, using BOLD-MRI of eight adult beagles were assessed at two time-points, 2–3 weeks apart. During each time point, the anesthetized dogs received either a control (0.9% sodium chloride) or a dopamine treatment. For each time point, measurements were carried out over 2 days. An MRI scan at 3 T was performed on day one, followed by CEUS on day two.ResultsUsing the model-free model with caudal placement of the arterial input function (AIF) region of interest (ROI) in the aorta, the DCE results showed a significant correlation with ASL measured RBF and detected significant changes in blood flow during dopamine infusion. Additionally, R2* negatively correlated with ASL measured RBF at the cortex and medulla, as well as with medullary wash-in rate (WiR) and peak intensity (PI). ASL measured RBF, in its turn, showed a positive correlation with cortical WiR, PI, area under the curve (AUC) and fall time (FT), and with medullary WiR and PI, but a negative correlation with medullary rise time (RT). During dopamine infusion, BOLD-MRI observed a significant decrease in R2* at the medulla and entire kidney, while ASL-MRI demonstrated a significant increase in RBF at the cortex, medulla and the entire kidney.ConclusionASL- and BOLD-MRI can measure pharmacologically induced changes in renal blood flow and renal oxygenation in dogs and might allow detection of changes that cannot be observed with CEUS. However, further research is needed to confirm the potential of ASL- and BOLD-MRI in dogs and to clarify which analysis method is most suitable for DCE-MRI in dogs.
Collapse
Affiliation(s)
- Amber Hillaert
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Luis Carlos Sanmiguel Serpa
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Stephanie Bogaert
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, Ghent, Belgium
| | - Bart J. G. Broeckx
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Myriam Hesta
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eva Vandermeulen
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jolien Germonpré
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Emmelie Stock
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Pim Pullens
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, Ghent, Belgium
- Institute of Biomedical Engineering and Technology, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Katrien Vanderperren
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
4
|
Chae SY, Kim Y, Park CW. Oxidative Stress Induced by Lipotoxicity and Renal Hypoxia in Diabetic Kidney Disease and Possible Therapeutic Interventions: Targeting the Lipid Metabolism and Hypoxia. Antioxidants (Basel) 2023; 12:2083. [PMID: 38136203 PMCID: PMC10740440 DOI: 10.3390/antiox12122083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Oxidative stress, a hallmark pathophysiological feature in diabetic kidney disease (DKD), arises from the intricate interplay between pro-oxidants and anti-oxidants. While hyperglycemia has been well established as a key contributor, lipotoxicity emerges as a significant instigator of oxidative stress. Lipotoxicity encompasses the accumulation of lipid intermediates, culminating in cellular dysfunction and cell death. However, the mechanisms underlying lipotoxic kidney injury in DKD still require further investigation. The key role of cell metabolism in the maintenance of cell viability and integrity in the kidney is of paramount importance to maintain proper renal function. Recently, dysfunction in energy metabolism, resulting from an imbalance in oxygen levels in the diabetic condition, may be the primary pathophysiologic pathway driving DKD. Therefore, we aim to shed light on the pivotal role of oxidative stress related to lipotoxicity and renal hypoxia in the initiation and progression of DKD. Multifaceted mechanisms underlying lipotoxicity, including oxidative stress with mitochondrial dysfunction, endoplasmic reticulum stress activated by the unfolded protein response pathway, pro-inflammation, and impaired autophagy, are delineated here. Also, we explore potential therapeutic interventions for DKD, targeting lipotoxicity- and hypoxia-induced oxidative stress. These interventions focus on ameliorating the molecular pathways of lipid accumulation within the kidney and enhancing renal metabolism in the face of lipid overload or ameliorating subsequent oxidative stress. This review highlights the significance of lipotoxicity, renal hypoxia-induced oxidative stress, and its potential for therapeutic intervention in DKD.
Collapse
Affiliation(s)
- Seung Yun Chae
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (S.Y.C.); (Y.K.)
| | - Yaeni Kim
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (S.Y.C.); (Y.K.)
| | - Cheol Whee Park
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (S.Y.C.); (Y.K.)
- Institute for Aging and Metabolic Disease, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
5
|
Inada A, Fukatsu A. Persistence and expansion of hypoxia detected by pimonidazole adduct immunostaining during progression of diabetic nephropathy in diabetic mice. Am J Physiol Renal Physiol 2023; 325:F527-F535. [PMID: 37615048 DOI: 10.1152/ajprenal.00160.2023] [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: 06/12/2023] [Revised: 07/25/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Hypoxia and oxidative stress are considered to be underlying factors in the deterioration of renal function and pathogenesis in acute kidney injury (AKI) and chronic kidney disease, including diabetic nephropathy (DN). However, the long-term role of hypoxia in DN is unknown. Here, we investigated the distribution, severity, and time course of hypoxia during DN development in our well-established severely diabetic transgenic (Tg) DN mouse model that mimics human DN up to 80 wk of age, using pimonidazole adduct immunohistochemistry. The relationship between pimonidazole adduct distribution and hypoxia-inducible factor (HIF) expression was also examined. We found 1) persistent pimonidazole immunostaining mainly in the outer zone of the outer medulla, extending into the inner zone, 2) significant expansion of area and intensity up to 40 wk of age, and 3) characteristic subcellular localization mainly at apical sites in vesicular form by laser scanning microscopy of thin slices. The distribution of pimonidazole adducts was different from that of HIF reported previously, indicating that hypoxia does not directly contribute to persistent abnormal HIF expression. These results suggest that pimonidazole adducts produced under low [Formula: see text] conditions are sustained by a mechanism distinct from direct ischemia. We propose that in the long course of DN development, persistent hyperfiltration and hyperexcretion of glucose, albumin, and water increase metabolism and energy expenditure in the tubules, and such chronic stimulation leads to relative ischemia and local hypoxia, which may contribute in part to the loss of nephrons.NEW & NOTEWORTHY This study provides new insights into hypoxia during the long course of diabetic nephropathy development. Hypoxia was persistently localized only in limited areas and its distribution differed significantly from that of hypoxia-inducible factors. These findings suggests that in the long course of diabetic nephropathy development, increased energy requirements and limited blood supply may lead to relative ischemia and induction of local and persistent hypoxia, which may contribute in part to the loss of nephrons.
Collapse
Affiliation(s)
- Akari Inada
- Clinical Research Department, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Diabetes and Genes, Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
6
|
Liu F, Hu W, Sun Y, Shen Y, Zhou W, Dai Y, Gu L, Zhang M, Zhou Y. Exploration of Interstitial Fibrosis in Chronic Kidney Disease by Diffusion‐Relaxation Correlation Spectrum
MR
Imaging: A Preliminary Study. J Magn Reson Imaging 2022. [DOI: 10.1002/jmri.28535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Fang Liu
- Department of Radiology Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Wentao Hu
- Central Research Institute, United Imaging Healthcare Shanghai China
| | - Yawen Sun
- Department of Radiology Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Yiwei Shen
- Department of Nephrology Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Wenyan Zhou
- Department of Nephrology Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Yongming Dai
- Central Research Institute, United Imaging Healthcare Shanghai China
| | - Leyi Gu
- Department of Nephrology Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Minfang Zhang
- Department of Nephrology Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Yan Zhou
- Department of Radiology Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
7
|
Gonzalez J, Jatem E, Roig J, Valtierra N, Ostos E, Abo A, Santacana M, Garcia A, Segarra A. Usefulness of urinary biomarkers to estimate the interstitial fibrosis surface in diabetic nephropathy with normal kidney function. Nephrol Dial Transplant 2022; 37:2102-2110. [PMID: 35583251 DOI: 10.1093/ndt/gfac185] [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: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Kidney biopsies of patients with diabetic nephropathy (DN) and normal kidney function may exhibit interstitial fibrosis (IF) without reduction of glomerular filtration rate because of hyperfiltration. The aim of our study was to analyze the performance of a set of biomarkers of tubular injury to estimate the extent of IF in patients with DN and normal kidney function. PATIENTS AND METHODS This cross-sectional study included 118 adults with DN diagnosed by kidney biopsy and GFR ≥ 90 mL/min/1.73 m2 and a control group of healthy subjects. We measured the urinary excretion of MCP-1, NGAL, KIM-1, L-FABP, β2-microglobulin and DKK-3 at the time of kidney biopsy. GFR was measured by Cr-EDTA (mGFR). IF was quantified using a quantitative morphometric procedure. Predictive multivariate models were developed to estimate the IF surface. RESULTS patients with DN showed significantly higher levels of DKK-3, MCP-1 and L-FABP and significantly lower levels of EGF than healthy controls. There were no significant between-group differences in the levels of β2-microglobulin, KIM-1 or NGAL. IF was negatively associated with EGF and a positively with age, proteinuria, MCP-1, DKK-3 and L-FABP but no with β2-microglobulin, KIM-1, NGAL or GFR. The best model to predict IF surface accounted for 59% of its variability and included age, proteinuria, EGF, DKK-3 and MCP-1. CONCLUSIONS our study provides a model to estimate the IF in DN that can be useful to assess the progression of IF in patients with normal kidney function.
Collapse
Affiliation(s)
- Jorge Gonzalez
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida
| | - Elias Jatem
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida
| | - Jordi Roig
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida
| | | | - Elena Ostos
- Vall d'Hebrón Institut de Recerca, Barcelona
| | - Anabel Abo
- Servicio de Anatomia Patológica, Hospital Arnau de Vilanova, Lleida
| | - Maria Santacana
- Servicio de Anatomia Patológica, Hospital Arnau de Vilanova, Lleida.,Institut de Recerca Biomèdica August Pi i Sunyer, Lleida
| | - Alicia Garcia
- Institut de Recerca Biomèdica August Pi i Sunyer, Lleida
| | - Alfons Segarra
- Servicio de Nefrologia Hospital Arnau de Vilanova, Lleida.,Vall d'Hebrón Institut de Recerca, Barcelona.,Institut de Recerca Biomèdica August Pi i Sunyer, Lleida.,Servicio de Nefrologia Hospital Vall d'Hebrón, Barcelona, Spain
| |
Collapse
|
8
|
Copur S, Yavuz F, Sag AA, Tuttle KR, Kanbay M. Future of kidney imaging: Functional magnetic resonance imaging and kidney disease progression. Eur J Clin Invest 2022; 52:e13765. [PMID: 35267195 DOI: 10.1111/eci.13765] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Chronic kidney disease (CKD) which is a common cause of death has an increasing trend, but there is no established approach for predicting CKD progression yet. Functional magnetic resonance imaging (fMRI) studies such as blood oxygenation level-dependent MRI (BOLD-MRI), diffusion-weighted MRI (DWI-MRI), diffusion-tensor MRI (DTI-MRI) and arterial spin labelling MRI (ASL-MRI) are rising methods for the assessment of kidney functions in native and transplanted kidneys as well as the estimation of CKD progression. METHODS Systematic literature review was performed through the Embase (Elsevier), Cochrane Central Register of Controlled Trials (Wiley), PubMed/Medline and Web of Science databases, and studies investigating the role of fMRI methods assessing kidney functions in native and transplanted kidneys, as well as the value of fMRI methods to predict CKD progression, were included. Working mechanisms, advantages and limitations of the fMRI modalities were reviewed, and three studies investigating the role of fMRI studies in kidney functions were analysed. RESULTS AND CONCLUSION BOLD-MRI signal was found to be inversely correlated with annual eGFR change, and DWI/ADC (apparent diffusion coefficient map) values were shown to be correlated with annual eGFR decline. fMRI methods which are currently used for other systems can be utilized to provide more detailed information about kidney functions, and doctors should be ready to interpret kidney MRIs.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Furkan Yavuz
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alan A Sag
- Department of Radiology, Division of Vascular and Interventional Radiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Kathherine R Tuttle
- Division of Nephrology, University of Washington, Seattle, Washington, USA.,Providence Medical Research Center, Providence Health Care, Washington, District of Columbia, USA
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
9
|
Sivertsson E, Friederich-Persson M, Persson P, Nangaku M, Hansell P, Palm F. Thyroid hormone increases oxygen metabolism causing intrarenal tissue hypoxia; a pathway to kidney disease. PLoS One 2022; 17:e0264524. [PMID: 35239685 PMCID: PMC8893624 DOI: 10.1371/journal.pone.0264524] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 02/11/2022] [Indexed: 01/10/2023] Open
Abstract
The proposed mechanisms for the development of nephropathy are many, complex and often overlapping. Although recent literature strongly supports a role of kidney hypoxia as an independent pathway to nephropathy, the evidence remains inconclusive since the role of hypoxia is difficult to differentiate from confounding factors such as hyperglycemia, hypertension and oxidative stress. By increasing kidney oxygen consumption using triiodothyronine (T3) and, thus, avoiding these confounding factors, the aim of the present study was to investigate renal hypoxia per se as a causal pathway for the development of nephropathy. Healthy Sprague-Dawley rats were treated with T3 (10 μg/kg/day) and the angiotensin II AT1-receptor antagonist candesartan (1 mg/kg in drinking water) to eliminate effects of T3-induced renin release; and compared to a candesartan treated control group. After 7 weeks of treatment in vivo kidney function, oxygen metabolism and mitochondrial function were evaluated. T3 did not affect glomerular filtration rate or renal blood flow, but increased total kidney oxygen consumption resulting in cortical hypoxia. Nephropathy, demonstrated as albuminuria and tubulointerstitial fibrosis, developed in T3-treated animals. Mitochondria uncoupling mediated by uncoupling protein 2 and the adenosine nucleotide transporter was demonstrated as a mechanism causing the increased kidney oxygen consumption. Importantly, blood glucose levels, mean arterial blood pressure and oxidative stress levels were not affected by T3. In conclusion, the present study provides further evidence for increased kidney oxygen consumption causing intrarenal tissue hypoxia, as a causal pathway for development of nephropathy.
Collapse
Affiliation(s)
- Ebba Sivertsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Patrik Persson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Peter Hansell
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
10
|
Seah JM, Botterill E, MacIsaac RJ, Milne M, Ekinci EI, Lim RP. Functional MRI in assessment of diabetic kidney disease in people with type 1 diabetes. J Diabetes Complications 2022; 36:108076. [PMID: 34802902 DOI: 10.1016/j.jdiacomp.2021.108076] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022]
Abstract
AIMS To compare levels of renal hypoxia measured by Blood Oxygen Level Dependent (BOLD) magnetic resonance imaging (MRI) with measured transverse relaxation rate (R2*) and renal structural changes including apparent diffusion coefficient (ADC) and fractional anisotropy (FA) in patients with type 1 diabetes and healthy controls. METHODS Cohort study comparing MRI metrics in type 1 diabetes (n = 32, GFR 105 (77, 120) ml/min.1.73m2) and controls (n = 10). Renal function and selected inflammatory renal biomarkers were also measured. RESULTS For BOLD, we found reduced cortical [14.7 (13.7,15.8) (1/s) vs 15.7 (15.1,16.6) (1/s), p < 0.001] and medullary [24.8 (21.8,28.2) (1/s) vs. 29.3 (24.3,32.4) (1/s), p < 0.001] R2*, indicating more oxygenated parenchyma, in type 1 diabetes vs. controls, respectively. We observed reduced cortical FA, indicating decreased structural integrity in type 1 diabetes -0.04 (-0.07, -0.01), (p = 0.02). We found reduced cortical ADC, reflecting reduced water diffusion, in non-hyperfiltering [2.40 (2.29,2.53) (103mm2/s)] versus hyperfiltering [2.61 (2.53,2.74) (103mm2/s)] type 1 diabetes patients. MRI parameters correlated with renal function and inflammatory renal biomarkers. CONCLUSIONS MRI derived indices of renal function and structure differed between (i) type 1 diabetes and healthy controls, and (ii) between non-hyperfiltering and hyperfiltering type 1 diabetes patients, providing insight into the role of hypoxia and renal structural, and functional changes in DKD.
Collapse
Affiliation(s)
- Jas-Mine Seah
- Department of Endocrinology and Diabetes, Austin Health, Heidelberg, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Elissa Botterill
- Department of Radiology and Surgery, Austin Health, Heidelberg, VIC, Australia
| | - Richard J MacIsaac
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Fitzroy, VIC, Australia
| | - Michele Milne
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Elif I Ekinci
- Department of Endocrinology and Diabetes, Austin Health, Heidelberg, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia.
| | - Ruth P Lim
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Department of Radiology and Surgery, Austin Health, Heidelberg, VIC, Australia
| |
Collapse
|
11
|
Wahba EN, Elsharkawy A, Awad MH, Abdel Rahman A, Sarhan A. Role of magnetic resonance diffusion weighted imaging in diagnosis of diabetic nephropathy in children living with type 1 diabetes mellitus. J Pediatr Endocrinol Metab 2021; 34:1585-1591. [PMID: 34496164 DOI: 10.1515/jpem-2021-0379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/23/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Diabetic nephropathy is a serious and a common complication of diabetes that can lead to end stage renal disease among children living with type 1 diabetes, thus an early and accurate method of diagnosis that allows timely intervention is of high importance. This study aimed to evaluate the role of magnetic resonance diffusion weighted imaging in diagnosis of diabetic nephropathy in children with type 1 diabetes. METHODS This prospective, observational, case control study included 30 children with type 1 diabetes and 30 matched healthy controls attending the outpatient clinics in Mansoura University Children's Hospital. All were subjected to magnetic resonance DWI of the renal parenchyma and their glomerular filtration rate (GFR) was estimated, along with micro albumin in 24 h urine collection and HbA1c in patients with diabetes. RESULTS Children with diabetes who were positive for microalbuminuria had significantly lower apparent diffusion coefficient value compared to Children with diabetes who were negative for microalbuminuria (p = 0.034) as well as controls (p = 0.001). Among children with type 1 diabetes, apparent diffusion coefficient had significant positive correlation with estimated glomerular filtration rate (r = 0.491, p = 0.006) and negative correlation with microalbuminuria (r = -0.437, p = 0.016). CONCLUSION Magnetic resonance DWI of the renal parenchyma is correlated with estimated glomerular filtration rate (eGFR) in children with type 1 diabetes and can detect GFR deterioration even in presence of normal albumin excretion.
Collapse
Affiliation(s)
| | - Ashraf Elsharkawy
- Department of Pediatrics, Mansoura University Children's Hospital, Mansoura, Egypt
| | - Mohammad Hosny Awad
- Department of Pediatrics, Mansoura University Children's Hospital, Mansoura, Egypt
| | - Ashraf Abdel Rahman
- Diagnostic Radiology Department, Mansoura Faculty of Medicine, Mansoura University Children Hospital, Mansoura, Egypt
| | - Amr Sarhan
- Department of Pediatrics, Mansoura University Children's Hospital, Mansoura, Egypt
| |
Collapse
|
12
|
Hanai S, Uchimura K, Takahashi K, Ishii T, Mitsui T, Furuya F. Hypoxia-induced thyroid hormone receptor expression regulates cell-cycle progression in renal tubule epithelial cells. Endocr J 2021; 68:1309-1320. [PMID: 34108302 DOI: 10.1507/endocrj.ej21-0245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Hypoxia occurs in the kidneys of chronic kidney disease (CKD) patients, inducing interstitial fibrosis and tubule cell death. Renal tubule cell death is an important determinant of mortality in CKD. We focused on the regulation of cell-cycle-mediated protein expression to prevent cell death under chronic hypoxia in the kidneys of CKD patients. Paraffin-embedded kidney sections from patients with CKD (diabetes nephropathy, nephrosclerosis, or IgA nephropathy) were analyzed for the expression of hypoxia-inducible factor (HIF), thyroid hormone receptor (TR) β, or p21 and levels of interstitial fibrosis. Human renal proximal tubule cells were exposed to hypoxia and analyzed for the expression of HIF, TRβ, or p21 and the cell-cycle stage. TRβ expression was enhanced early on when fibrosis was not fully developed in the tubule cells of CKD patients. HIF1α bound to the TRβ promoter and directly induced its transcription. Further, HIF1α expression induced the expression of TRβ and inhibited cell-cycle progression. In the early stage of kidney injury, TRβ might act as a guardian to prepare and organize cell-cycle proliferation and prevent cell death. While the molecular mechanism that regulates the expression of cell-cycle regulators in renal tubule cells remains controversial, TRβ has strong potential as a new therapeutic target.
Collapse
Affiliation(s)
- Shunichiro Hanai
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kohei Uchimura
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kazuya Takahashi
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Toshihisa Ishii
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Takahiko Mitsui
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Fumihiko Furuya
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
13
|
Feng YZ, Chen XQ, Cheng ZY, Lin QT, Chen PK, Si-Tu DK, Cao R, Qian L, Heng B, Cai XR. Non-invasive investigation of early kidney damage in streptozotocin-induced diabetic rats by intravoxel incoherent motion diffusion-weighted (IVIM) MRI. BMC Nephrol 2021; 22:321. [PMID: 34565330 PMCID: PMC8474753 DOI: 10.1186/s12882-021-02530-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 09/10/2021] [Indexed: 11/21/2022] Open
Abstract
Background The current study investigated the performance of intravoxel incoherent motion diffusion (IVIM) technology in monitoring early renal injury in streptozotocin rats. Methods Forty-eight Sprague-Dawley (SD) rats were divided into a control group and a diabetic mellitus (DM) group. Six rats in each group were randomly selected for MR scans at four different time points (0, 4, 8, and 12 weeks). The IVIM-derived parameters (D, D*, f and ADC values) of the renal cortex (CO), outer and inner stripe of the outer medulla (OS, IS), and internal medulla (IM) were acquired. Changes in each IVIM-derived parameter over time were analyzed, and differences between the two groups at each point were assessed. The associations between the IVIM parameters and IV collagen expression, urine volume (UV), blood urea nitrogen (BUN), and serum creatinine (Scr) were investigated. Results The D and D* values of CO and the ADC values of CO, OS, IS and IM displayed significantly different trends between the two groups over time (P<0.05). In addition, significant correlations were discovered between the D* value of CO and UV and BUN (r=0.527, P=0.033; r=0.617, P=0.005), between the ADC value of IM and BUN (r=0.557, P=0.019) and between the f value of IM and BUN (r=0.527, P=0.033). No correlation was found between IVIM parameters and IV collagen expression and Scr. Conclusions IVIM is a potential sensitive and noninvasive technology for the simultaneous assessment of early renal cortical and medullary injuries induced by diabetes. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-021-02530-8.
Collapse
Affiliation(s)
- You-Zhen Feng
- Medical Imaging Center, Jinan University First Affiliated Hospital, No.613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Xiao-Qiao Chen
- Medical Imaging Center, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhong-Yuan Cheng
- Medical Imaging Center, Jinan University First Affiliated Hospital, No.613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Qi-Ting Lin
- Medical Imaging Center, Jinan University First Affiliated Hospital, No.613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Ping-Kang Chen
- Medical Imaging Center, Jinan University First Affiliated Hospital, No.613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Ding-Kun Si-Tu
- Medical Imaging Center, Jinan University First Affiliated Hospital, No.613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Rui Cao
- Nephrology Department, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Long Qian
- GE Healthcare, Beijing, China.,Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Baoli Heng
- Yingde Base, Institute of Kidney Surgery, Jinan University, Guangzhou, Guangdong, China.,Department of Urology, Jinan University First Affiliated Hospital, Guangzhou, China
| | - Xiang-Ran Cai
- Medical Imaging Center, Jinan University First Affiliated Hospital, No.613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
14
|
Spencer S, Wheeler-Jones C, Elliott J. Hypoxia and chronic kidney disease: Possible mechanisms, therapeutic targets, and relevance to cats. Vet J 2021; 274:105714. [PMID: 34252550 DOI: 10.1016/j.tvjl.2021.105714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
There is mounting evidence that kidney ischaemia/hypoxia plays an important role in feline chronic kidney disease (CKD) development and progression, as well as in human disease and laboratory animal models. Ischaemic acute kidney injury is widely accepted as a cause of CKD in people and data from laboratory species has identified some of the pathways underlying this continuum. Experimental kidney ischaemia in cats results in morphological changes, namely chronic tubulointerstitial inflammation, tubulointerstitial fibrosis, and tubular atrophy, akin to those observed in naturally-occurring CKD. Multiple situations are envisaged that could result in acute or chronic episodes of kidney hypoxia in cats, while risk factors identified in epidemiological studies provide further support that kidney hypoxia contributes to spontaneously occurring feline CKD. This review evaluates the evidence for the role of kidney ischaemia/hypoxia in feline CKD and the proposed mechanisms and consequences of kidney hypoxia. As no effective treatments exist that substantially slow or prevent feline CKD progression, there is a need for novel therapeutic strategies. Targeting kidney hypoxia is one such promising approach, with therapies including those that attenuate the hypoxia-inducible factor (HIF) pathway already being utilised in human CKD.
Collapse
Affiliation(s)
- Sarah Spencer
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| | - Caroline Wheeler-Jones
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Jonathan Elliott
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| |
Collapse
|
15
|
Zhang D, Li R, Chen M, Vu T, Sheng H, Yang W, Hoffmann U, Luo J, Yao J. Photoacoustic imaging of in vivo hemodynamic responses to sodium nitroprusside. JOURNAL OF BIOPHOTONICS 2021; 14:e202000478. [PMID: 33768709 PMCID: PMC8263508 DOI: 10.1002/jbio.202000478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/17/2021] [Accepted: 03/12/2021] [Indexed: 05/25/2023]
Abstract
The in vivo hemodynamic impact of sodium nitroprusside (SNP), a widely used antihypertensive agent, has not been well studied. Here, we applied functional optical-resolution photoacoustic microscopy (OR-PAM) to study the hemodynamic responses to SNP in mice in vivo. As expected, after the application of SNP, the systemic blood pressure (BP) was reduced by 53%. The OR-PAM results show that SNP induced an arterial vasodilation of 24% and 23% in the brain and skin, respectively. A weaker venous vasodilation of 9% and 5% was also observed in the brain and skin, respectively. The results show two different types of blood oxygenation response. In mice with decreased blood oxygenation, the arterial and venous oxygenation was respectively reduced by 6% and 13% in the brain, as well as by 7% and 18% in the skin. In mice with increased blood oxygenation, arterial and venous oxygenation was raised by 4% and 22% in the brain, as well as by 1% and 9% in the skin. We observed venous change clearly lagged the arterial change in the skin, but not in the brain. Our results collectively show a correlation among SNP induced changes in systemic BP, vessel size and blood oxygenation.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ran Li
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Maomao Chen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Tri Vu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Huaxin Sheng
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Wei Yang
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Ulrike Hoffmann
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Jianwen Luo
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
16
|
Wang R, Lin Z, Yang X, Zhao K, Wang S, Sui X, Su T, Wang X. Noninvasive Evaluation of Renal Hypoxia by Multiparametric Functional MRI in Early Diabetic Kidney Disease. J Magn Reson Imaging 2021; 55:518-527. [PMID: 34184356 DOI: 10.1002/jmri.27814] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Renal hypoxia, which caused by a mismatch between oxygen delivery and oxygen demand, may be the primary pathophysiological pathway driving diabetic kidney disease (DKD). Blood oxygenation level-dependent (BOLD) magnetic resonance imaging (MRI) could detect hypoxia, but can be limited in distinguishing increased oxygen consumption or decreased blood supply. PURPOSE To explore multiparametric functional MRI in evaluating mechanism of the hypoxia changes in early stage of DKD. STUDY TYPE Prospective. ANIMAL MODEL Thirty-five New Zealand White rabbits were divided into control group (n = 5) and alloxan-induced diabetes mellitus (DM) groups (DM3 group: n = 15, DM7 group: n = 15). FIELD STRENGTH/SEQUENCE 3 T MRI/BOLD, arterial spin labeling (ASL), and asymmetric spin-echo (ASE). ASSESSMENT The renal oxygenation level (R2*), renal blood flow (RBF), and oxygen extraction fraction (OEF) were evaluated by BOLD, ASL, and ASE MRI, respectively. The regions of interest were manually drawn including cortex, outer stripes of outer medulla (OS), and inner stripes of outer medulla (IS). STATISTICAL TESTS Analysis of variance, independent-sample t-test, and paired-sample t-test were applied for comparisons among groups, between groups, and within the same group. P < 0.05 was considered statistically significant. RESULTS All renal regions of DM3 group at Day 3 after DM induction showed significantly higher R2* and OEF values compared to baseline. The RBF values showed no statistically significant difference (P = 0.62, 0.76, 0.09 in cortex, OS, and IS, respectively). For DM7 group at Day 7, R2*, OEF, and RBF values showed no statistically significant difference compared to baseline (P = 0.06, 0.05, 0.06 of R2*; 0.70, 0.64, 0.68 of OEF; and 0.33, 0.58, 0.48 of RBF in cortex, OS, and IS, respectively). DATA CONCLUSION BOLD MRI could detect renal hypoxia in early stage of DKD rabbit model, which was mainly revealed by increased oxygen consumption, but not affected by renal blood flow change. LEVEL OF EVIDENCE 2 Technical Efficacy Stage: 1.
Collapse
Affiliation(s)
- Rui Wang
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Zhiyong Lin
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Xuedong Yang
- Department of Radiology, China Academy of Chinese Medical Sciences Guanganmen Hospital, Beijing, China
| | - Kai Zhao
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Suxia Wang
- Renal Pathology Center, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Xueqing Sui
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tao Su
- Department of Nephrology, Peking University First Hospital, Beijing, China
| | - Xiaoying Wang
- Department of Radiology, Peking University First Hospital, Beijing, China
| |
Collapse
|
17
|
Perspectives on the Role of Magnetic Resonance Imaging (MRI) for Noninvasive Evaluation of Diabetic Kidney Disease. J Clin Med 2021; 10:jcm10112461. [PMID: 34199385 PMCID: PMC8199575 DOI: 10.3390/jcm10112461] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/09/2023] Open
Abstract
Renal magnetic resonance imaging (MRI) techniques are currently in vogue, as they provide in vivo information on renal volume, function, metabolism, perfusion, oxygenation, and microstructural alterations, without the need for exogenous contrast media. New imaging biomarkers can be identified using these tools, which represent a major advance in the understanding and study of the different pathologies affecting the kidney. Diabetic kidney disease (DKD) is one of the most important diseases worldwide due to its high prevalence and impact on public health. However, its multifactorial etiology poses a challenge for both basic and clinical research. Therefore, the use of novel renal MRI techniques is an attractive step forward in the comprehension of DKD, both in its pathogenesis and in its detection and surveillance in the clinical practice. This review article outlines the most promising MRI techniques in the study of DKD, with the purpose of stimulating their clinical translation as possible tools for the diagnosis, follow-up, and monitoring of the clinical impacts of new DKD treatments.
Collapse
|
18
|
Zheng SS, He YM, Lu J. Noninvasive evaluation of diabetic patients with high fasting blood glucose using DWI and BOLD MRI. Abdom Radiol (NY) 2021; 46:1659-1669. [PMID: 32997155 DOI: 10.1007/s00261-020-02780-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To investigate the renal microstructure changes and hypoxia changes in type 2 diabetic patients and the relationship between them and glucose using both diffusion-weighted imaging (DWI) and blood oxygenation level-dependent magnetic resonance imaging (BOLD MRI). METHODS After measuring morning fasting blood glucose, DWI and BOLD MRI were performed in 57 patients with type 2 diabetes mellitus (DM group) and 14 healthy volunteers (NC group). According to the fasting blood glucose levels, diabetic patients were divided into a normoglycemic diabetes group (group A), a less hyperglycemic diabetes group (group B) and a more hyperglycemic diabetes group (group C). The renal parenchymal apparent diffusion coefficient (ADC), renal cortical R2* (CR2*), and medullary R2* (MR2*) were measured, and the R2* ratio between the medulla and cortex (MCR) was calculated. To test for differences in ADC, R2*, and MCR among the four groups, the data were analyzed by separate one-way ANOVAs. The correlations between ADC, R2*, and MCR and the clinical index of renal function were analyzed. RESULTS Groups B and C had significantly lower ADC values in the renal parenchyma (P = 0.048, 0.002) and significantly higher MR2* and MCR values (P < 0.000, P = 0.001, 0.001, and 0.005, respectively) than the NC group. ADC was negatively correlated with glucose, and MR2*, MCR and glucose showed a weak positive correlation. CONCLUSION DWI and BOLD may indirectly and qualitatively reflect the kidney microstructure status and hypoxia level of diabetic patients at different blood glucose levels to a certain extent, and possibly guide the clinical treatment of diabetic patients with different blood glucose levels.
Collapse
Affiliation(s)
- Shuang-Shuang Zheng
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Department of Radiology, Fuxing Hospital, Capital Medical University, Beijing, 100038, China
| | - Yue-Ming He
- Department of Radiology, Fuxing Hospital, Capital Medical University, Beijing, 100038, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Department of Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, 100053, China.
| |
Collapse
|
19
|
Chang J, Yan J, Li X, Liu N, Zheng R, Zhong Y. Update on the Mechanisms of Tubular Cell Injury in Diabetic Kidney Disease. Front Med (Lausanne) 2021; 8:661076. [PMID: 33859992 PMCID: PMC8042139 DOI: 10.3389/fmed.2021.661076] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence supports a role of proximal tubular (PT) injury in the progression of diabetic kidney disease (DKD), in patients with or without proteinuria. Research on the mechanisms of the PT injury in DKD could help us to identify potential new biomarkers and drug targets for DKD. A high glucose transport state and mismatched local hypoxia in the PT of diabetes patients may be the initiating factors causing PT injury. Other mechanism such as mitochondrial dysfunction, reactive oxygen species (ROS) overproduction, ER stress, and deficiency of autophagy interact with each other leading to more PT injury by forming a vicious circle. PT injury eventually leads to the development of tubulointerstitial inflammation and fibrosis in DKD. Many downstream signaling pathways have been demonstrated to mediate these diseased processes. This review focuses mostly on the novel mechanisms of proximal renal tubular injury in DKD and we believe such review could help us to better understand the pathogenesis of DKD and identify potential new therapies for this disease.
Collapse
Affiliation(s)
- Jingsheng Chang
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiayi Yan
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xueling Li
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ni Liu
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Zheng
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Zhong
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Abstract
Hypoxia can be defined as a relative deficiency in the amount of oxygen reaching the tissues. Hypoxia-inducible factors (HIFs) are critical regulators of the mammalian response to hypoxia. In normal circumstances, HIF-1α protein turnover is rapid, and hyperglycemia further destabilizes the protein. In addition to their role in diabetes pathogenesis, HIFs are implicated in development of the microvascular and macrovascular complications of diabetes. Improving glucose control in people with diabetes increases HIF-1α protein and has wide-ranging benefits, some of which are at least partially mediated by HIF-1α. Nevertheless, most strategies to improve diabetes or its complications via regulation of HIF-1α have not currently proven to be clinically useful. The intersection of HIF biology with diabetes is a complex area in which many further questions remain, especially regarding the well-conducted studies clearly describing discrepant effects of different methods of increasing HIF-1α, even within the same tissues. This Review presents a brief overview of HIFs; discusses the range of evidence implicating HIFs in β cell dysfunction, diabetes pathogenesis, and diabetes complications; and examines the differing outcomes of HIF-targeting approaches in these conditions.
Collapse
Affiliation(s)
- Jenny E Gunton
- Centre for Diabetes, Obesity and Endocrinology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Westmead Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| |
Collapse
|
21
|
Baraghithy S, Soae Y, Assaf D, Hinden L, Udi S, Drori A, Gabet Y, Tam J. Renal Proximal Tubule Cell Cannabinoid-1 Receptor Regulates Bone Remodeling and Mass via a Kidney-to-Bone Axis. Cells 2021; 10:414. [PMID: 33671138 PMCID: PMC7922053 DOI: 10.3390/cells10020414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/07/2021] [Accepted: 02/13/2021] [Indexed: 12/20/2022] Open
Abstract
The renal proximal tubule cells (RPTCs), well-known for maintaining glucose and mineral homeostasis, play a critical role in the regulation of kidney function and bone remodeling. Deterioration in RPTC function may therefore lead to the development of diabetic kidney disease (DKD) and osteoporosis. Previously, we have shown that the cannabinoid-1 receptor (CB1R) modulates both kidney function as well as bone remodeling and mass via its direct role in RPTCs and bone cells, respectively. Here we employed genetic and pharmacological approaches that target CB1R, and found that its specific nullification in RPTCs preserves bone mass and remodeling both under normo- and hyper-glycemic conditions, and that its chronic blockade prevents the development of diabetes-induced bone loss. These protective effects of negatively targeting CB1R specifically in RPTCs were associated with its ability to modulate erythropoietin (EPO) synthesis, a hormone known to affect bone mass and remodeling. Our findings highlight a novel molecular mechanism by which CB1R in RPTCs remotely regulates skeletal homeostasis via a kidney-to-bone axis that involves EPO.
Collapse
Affiliation(s)
- Saja Baraghithy
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (Y.S.); (D.A.); (L.H.); (S.U.); (A.D.)
| | - Yael Soae
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (Y.S.); (D.A.); (L.H.); (S.U.); (A.D.)
| | - Dekel Assaf
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (Y.S.); (D.A.); (L.H.); (S.U.); (A.D.)
| | - Liad Hinden
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (Y.S.); (D.A.); (L.H.); (S.U.); (A.D.)
| | - Shiran Udi
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (Y.S.); (D.A.); (L.H.); (S.U.); (A.D.)
| | - Adi Drori
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (Y.S.); (D.A.); (L.H.); (S.U.); (A.D.)
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel;
| | - Joseph Tam
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (Y.S.); (D.A.); (L.H.); (S.U.); (A.D.)
| |
Collapse
|
22
|
Waiczies S, Prinz C, Starke L, Millward JM, Delgado PR, Rosenberg J, Nazaré M, Waiczies H, Pohlmann A, Niendorf T. Functional Imaging Using Fluorine ( 19F) MR Methods: Basic Concepts. Methods Mol Biol 2021; 2216:279-299. [PMID: 33476007 PMCID: PMC9703275 DOI: 10.1007/978-1-0716-0978-1_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Kidney-associated pathologies would greatly benefit from noninvasive and robust methods that can objectively quantify changes in renal function. In the past years there has been a growing incentive to develop new applications for fluorine (19F) MRI in biomedical research to study functional changes during disease states. 19F MRI represents an instrumental tool for the quantification of exogenous 19F substances in vivo. One of the major benefits of 19F MRI is that fluorine in its organic form is absent in eukaryotic cells. Therefore, the introduction of exogenous 19F signals in vivo will yield background-free images, thus providing highly selective detection with absolute specificity in vivo. Here we introduce the concept of 19F MRI, describe existing challenges, especially those pertaining to signal sensitivity, and give an overview of preclinical applications to illustrate the utility and applicability of this technique for measuring renal function in animal models.This chapter is based upon work from the COST Action PARENCHIMA, a community-driven network funded by the European Cooperation in Science and Technology (COST) program of the European Union, which aims to improve the reproducibility and standardization of renal MRI biomarkers. This introduction chapter is complemented by two separate chapters describing the experimental procedure and data analysis.
Collapse
Affiliation(s)
- Sonia Waiczies
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.
| | - Christian Prinz
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Ludger Starke
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Jason M Millward
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Paula Ramos Delgado
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Jens Rosenberg
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA
| | - Marc Nazaré
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- Siemens Healthcare, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
23
|
Mrđanin T, Nikolić O, Molnar U, Mitrović M, Till V. Diffusion-weighted imaging in the assessment of renal function in patients with diabetes mellitus type 2. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2020; 34:273-283. [DOI: 10.1007/s10334-020-00869-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/15/2022]
|
24
|
Ullah MM, Basile DP. Role of Renal Hypoxia in the Progression From Acute Kidney Injury to Chronic Kidney Disease. Semin Nephrol 2020; 39:567-580. [PMID: 31836039 DOI: 10.1016/j.semnephrol.2019.10.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past 20 years, there has been an increased appreciation of the long-term sequelae of acute kidney injury (AKI) and the potential development of chronic kidney disease (CKD). Several pathophysiologic features have been proposed to mediate AKI to CKD progression including maladaptive alterations in tubular, interstitial, inflammatory, and vascular cells. These alterations likely interact to culminate in the progression to CKD. In this article we focus primarily on evidence of vascular rarefaction secondary to AKI, and the potential mechanisms by which rarefaction occurs in relation to other alterations in tubular and interstitial compartments. We further focus on the potential that rarefaction contributes to renal hypoxia. Consideration of the role of hypoxia in AKI to CKD transition focuses on experimental evidence of persistent renal hypoxia after AKI and experimental maneuvers to evaluate the influence of hypoxia, per se, in progressive disease. Finally, consideration of methods to evaluate hypoxia in patients is provided with the suggestion that noninvasive measurement of renal hypoxia may provide insight into progression in post-AKI patients.
Collapse
Affiliation(s)
- Md Mahbub Ullah
- Department of Anatomy, Cell Biology and Physiology, Indiana University, Indianapolis, IN
| | - David P Basile
- Department of Medicine, Division of Nephrology, Indiana University, Indianapolis, IN.
| |
Collapse
|
25
|
Gooding KM, Lienczewski C, Papale M, Koivuviita N, Maziarz M, Dutius Andersson AM, Sharma K, Pontrelli P, Garcia Hernandez A, Bailey J, Tobin K, Saunavaara V, Zetterqvist A, Shelley D, Teh I, Ball C, Puppala S, Ibberson M, Karihaloo A, Metsärinne K, Banks RE, Gilmour PS, Mansfield M, Gilchrist M, de Zeeuw D, Heerspink HJL, Nuutila P, Kretzler M, Welberry Smith M, Gesualdo L, Andress D, Grenier N, Shore AC, Gomez MF, Sourbron S. Prognostic imaging biomarkers for diabetic kidney disease (iBEAt): study protocol. BMC Nephrol 2020; 21:242. [PMID: 32600374 PMCID: PMC7323369 DOI: 10.1186/s12882-020-01901-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/19/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) remains one of the leading causes of premature death in diabetes. DKD is classified on albuminuria and reduced kidney function (estimated glomerular filtration rate (eGFR)) but these have modest value for predicting future renal status. There is an unmet need for biomarkers that can be used in clinical settings which also improve prediction of renal decline on top of routinely available data, particularly in the early stages. The iBEAt study of the BEAt-DKD project aims to determine whether renal imaging biomarkers (magnetic resonance imaging (MRI) and ultrasound (US)) provide insight into the pathogenesis and heterogeneity of DKD (primary aim) and whether they have potential as prognostic biomarkers in DKD (secondary aim). METHODS iBEAt is a prospective multi-centre observational cohort study recruiting 500 patients with type 2 diabetes (T2D) and eGFR ≥30 ml/min/1.73m2. At baseline, blood and urine will be collected, clinical examinations will be performed, and medical history will be obtained. These assessments will be repeated annually for 3 years. At baseline each participant will also undergo quantitative renal MRI and US with central processing of MRI images. Biological samples will be stored in a central laboratory for biomarker and validation studies, and data in a central data depository. Data analysis will explore the potential associations between imaging biomarkers and renal function, and whether the imaging biomarkers improve the prediction of DKD progression. Ancillary substudies will: (1) validate imaging biomarkers against renal histopathology; (2) validate MRI based renal blood flow measurements against H2O15 positron-emission tomography (PET); (3) validate methods for (semi-)automated processing of renal MRI; (4) examine longitudinal changes in imaging biomarkers; (5) examine whether glycocalyx and microvascular measures are associated with imaging biomarkers and eGFR decline; (6) explore whether the findings in T2D can be extrapolated to type 1 diabetes. DISCUSSION iBEAt is the largest DKD imaging study to date and will provide valuable insights into the progression and heterogeneity of DKD. The results may contribute to a more personalised approach to DKD management in patients with T2D. TRIAL REGISTRATION Clinicaltrials.gov ( NCT03716401 ).
Collapse
Affiliation(s)
- Kim M Gooding
- Diabetes and Vascular Medicine, University of Exeter Medical School, Barrack Road, Exeter, EX2 5AX, UK. .,NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK.
| | | | - Massimo Papale
- Department of Emergency and Organ Transplantation, Nephrology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Niina Koivuviita
- Department of Medicine, Division of Nephrology, Turku University Hospital, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - Marlena Maziarz
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmo, Sweden
| | | | - Kanishka Sharma
- Department of Imaging, Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Paola Pontrelli
- Department of Emergency and Organ Transplantation, Nephrology Unit, University of Bari Aldo Moro, Bari, Italy
| | | | | | - Kay Tobin
- Department of Renal Medicine and Renal Transplantation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Virva Saunavaara
- Department of Medical Physics, Division of Medical Imaging, Turku University Hospital, Turku, Finland
| | - Anna Zetterqvist
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmo, Sweden
| | - David Shelley
- Leeds Teaching Hospitals NHS Trust, Leeds, UK.,Advanced Imaging Centre, University of Leeds, Leeds, UK
| | - Irvin Teh
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Claire Ball
- NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | | | - Mark Ibberson
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Anil Karihaloo
- Novo Nordisk Research Center Seattle, Inc., Seattle, USA
| | - Kaj Metsärinne
- Department of Medicine, Division of Nephrology, Turku University Hospital, Turku, Finland
| | - Rosamonde E Banks
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | | | | - Mark Gilchrist
- Diabetes and Vascular Medicine, University of Exeter Medical School, Barrack Road, Exeter, EX2 5AX, UK
| | - Dick de Zeeuw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands.,University of Groningen, Groningen, The Netherlands
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands.,University of Groningen, Groningen, The Netherlands
| | - Pirjo Nuutila
- Department of Medicine, Division of Nephrology, Turku University Hospital, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - Matthias Kretzler
- Department of Nephrology, University of Michigan, Ann Arbor, USA.,Computational Medicine and Bioinformatics, University of Michigan, Ann Arbour, USA
| | - Matthew Welberry Smith
- Department of Renal Medicine and Renal Transplantation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Nephrology Unit, University of Bari Aldo Moro, Bari, Italy
| | | | - Nicolas Grenier
- Service de Radiologie, CHU de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - Angela C Shore
- Diabetes and Vascular Medicine, University of Exeter Medical School, Barrack Road, Exeter, EX2 5AX, UK.,NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Maria F Gomez
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmo, Sweden
| | - Steven Sourbron
- Department of Imaging, Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | | |
Collapse
|
26
|
Matoba K, Takeda Y, Nagai Y, Kanazawa Y, Kawanami D, Yokota T, Utsunomiya K, Nishimura R. ROCK Inhibition May Stop Diabetic Kidney Disease. JMA J 2020; 3:154-163. [PMID: 33150249 PMCID: PMC7590381 DOI: 10.31662/jmaj.2020-0014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease and is strongly associated with cardiovascular mortality. Given the pandemic of obesity and diabetes, the elucidation of the molecular underpinnings of DKD and establishment of effective therapy are urgently required. Studies over the past decade have identified the activated renin-angiotensin system (RAS) and hemodynamic changes as important therapeutic targets. However, given the residual risk observed in patients treated with RAS inhibitors and/or sodium glucose co-transporter 2 inhibitors, the involvement of other molecular machinery is likely, and the elucidation of such pathways represents fertile ground for the development of novel strategies. Rho-kinase (ROCK) is a serine/threonine kinase that is under the control of small GTPase protein Rho. Many fundamental cellular processes, including migration, proliferation, and survival are orchestrated by ROCK through a mechanism involving cytoskeletal reorganization. From a pathological standpoint, several analyses provide compelling evidence supporting the hypothesis that ROCK is an important regulator of DKD that is highly pertinent to cardiovascular disease. In cell-based studies, ROCK is activated in response to a diverse array of external stimuli associated with diabetes, and renal ROCK activity is elevated in the context of type 1 and 2 diabetes. Experimental studies have demonstrated the efficacy of pharmacological or genetic inhibition of ROCK in the prevention of diabetes-related histological and functional abnormalities in the kidney. Through a bird’s eye view of ROCK in renal biology, the present review provides a conceptual framework that may be widely applicable to the pathological processes of multiple organs and illustrate novel therapeutic promise in diabetology.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasushi Kanazawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Daiji Kawanami
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Hesp AC, Schaub JA, Prasad PV, Vallon V, Laverman GD, Bjornstad P, van Raalte DH. The role of renal hypoxia in the pathogenesis of diabetic kidney disease: a promising target for newer renoprotective agents including SGLT2 inhibitors? Kidney Int 2020; 98:579-589. [PMID: 32739206 DOI: 10.1016/j.kint.2020.02.041] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/06/2020] [Accepted: 02/26/2020] [Indexed: 12/17/2022]
Abstract
Diabetic kidney disease is the most common cause of end-stage kidney disease and poses a major global health problem. Finding new, safe, and effective strategies to halt this disease has proven to be challenging. In part that is because the underlying mechanisms are complex and not fully understood. However, in recent years, evidence has accumulated suggesting that chronic hypoxia may be the primary pathophysiological pathway driving diabetic kidney disease and chronic kidney disease of other etiologies and was called the chronic hypoxia hypothesis. Hypoxia is the result of a mismatch between oxygen delivery and oxygen demand. The primary determinant of oxygen delivery is renal perfusion (blood flow per tissue mass), whereas the main driver of oxygen demand is active sodium reabsorption. Diabetes mellitus is thought to compromise the oxygen balance by impairing oxygen delivery owing to hyperglycemia-associated microvascular damage and exacerbate oxygen demand owing to increased sodium reabsorption as a result of sodium-glucose cotransporter upregulation and glomerular hyperfiltration. The resultant hypoxic injury creates a vicious cycle of capillary damage, inflammation, deposition of the extracellular matrix, and, ultimately, fibrosis and nephron loss. This review will frame the role of chronic hypoxia in the pathogenesis of diabetic kidney disease and its prospect as a promising therapeutic target. We will outline the cellular mechanisms of hypoxia and evidence for renal hypoxia in animal and human studies. In addition, we will highlight the promise of newer imaging modalities including blood oxygenation level-dependent magnetic resonance imaging and discuss salutary interventions such as sodium-glucose cotransporter 2 inhibition that (may) protect the kidney through amelioration of renal hypoxia.
Collapse
Affiliation(s)
- Anne C Hesp
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands.
| | - Jennifer A Schaub
- Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Pottumarthi V Prasad
- Department of Radiology, NorthShore University Health System, Evanston, Illinois, USA; Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Volker Vallon
- Department of Medicine, University of California San Diego and Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Gozewijn D Laverman
- Department of Internal Medicine, Ziekenhuis Groep Twente, Almelo, The Netherlands
| | - Petter Bjornstad
- Department of Medicine, Division of Nephrology, and Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Baker ML, Perazella MA. SGLT2 inhibitor therapy in patients with type-2 diabetes mellitus: is acute kidney injury a concern? J Nephrol 2020; 33:985-994. [DOI: 10.1007/s40620-020-00712-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 02/11/2020] [Indexed: 12/11/2022]
|
29
|
Bane O, Mendichovszky IA, Milani B, Dekkers IA, Deux JF, Eckerbom P, Grenier N, Hall ME, Inoue T, Laustsen C, Lerman LO, Liu C, Morrell G, Pedersen M, Pruijm M, Sadowski EA, Seeliger E, Sharma K, Thoeny H, Vermathen P, Wang ZJ, Serafin Z, Zhang JL, Francis ST, Sourbron S, Pohlmann A, Fain SB, Prasad PV. Consensus-based technical recommendations for clinical translation of renal BOLD MRI. MAGMA (NEW YORK, N.Y.) 2020; 33:199-215. [PMID: 31768797 PMCID: PMC7021747 DOI: 10.1007/s10334-019-00802-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 01/08/2023]
Abstract
Harmonization of acquisition and analysis protocols is an important step in the validation of BOLD MRI as a renal biomarker. This harmonization initiative provides technical recommendations based on a consensus report with the aim to move towards standardized protocols that facilitate clinical translation and comparison of data across sites. We used a recently published systematic review paper, which included a detailed summary of renal BOLD MRI technical parameters and areas of investigation in its supplementary material, as the starting point in developing the survey questionnaires for seeking consensus. Survey data were collected via the Delphi consensus process from 24 researchers on renal BOLD MRI exam preparation, data acquisition, data analysis, and interpretation. Consensus was defined as ≥ 75% unanimity in response. Among 31 survey questions, 14 achieved consensus resolution, 12 showed clear respondent preference (65-74% agreement), and 5 showed equal (50/50%) split in opinion among respondents. Recommendations for subject preparation, data acquisition, processing and reporting are given based on the survey results and review of the literature. These technical recommendations are aimed towards increased inter-site harmonization, a first step towards standardization of renal BOLD MRI protocols across sites. We expect this to be an iterative process updated dynamically based on progress in the field.
Collapse
Affiliation(s)
- Octavia Bane
- BioMedical Engineering and Imaging Institute and Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Iosif A Mendichovszky
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Bastien Milani
- Center for BioMedical Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ilona A Dekkers
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean-Francois Deux
- Department of Radiology, Groupe Hospitalier Henri Mondor, Créteil, France
| | - Per Eckerbom
- Department of Radiology, Institution for Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Nicolas Grenier
- Department of Radiology, Université de Bordeaux, CHU de Bordeaux, Bordeaux, France
| | - Michael E Hall
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Tsutomu Inoue
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Christoffer Laustsen
- The MR Research Center Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Chunlei Liu
- Electrical Engineering and Computer Science, and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Glen Morrell
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Michael Pedersen
- Department of Clinical Medicine-Comparative Medicine Lab, Aarhus University Hospital, Aarhus, Denmark
| | - Menno Pruijm
- Nephrology and Hypertension Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Elizabeth A Sadowski
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Erdmann Seeliger
- Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Kanishka Sharma
- Imaging Biomarkers Group, Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Harriet Thoeny
- Department of Radiology, Hôpital Cantonal Fribourgois, University of Fribourg, Fribourg, Switzerland
| | - Peter Vermathen
- Departments for BioMedical Research and Radiology, Inselspital, Universitaetspital Bern, Bern, Switzerland
| | - Zhen J Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco Medical Center, San Francisco, CA, USA
| | - Zbigniew Serafin
- Department of Radiology, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Jeff L Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Susan T Francis
- Sir Peter Mansfield Centre, University of Notthingham, Notthingham, UK
| | - Steven Sourbron
- Imaging Biomarkers Group, Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sean B Fain
- Departments of Biomedical Engineering, Radiology, and Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Pottumarthi V Prasad
- Department of Radiology, Center for Advanced Imaging, NorthShore University Health System, Evanston, IL, USA.
| |
Collapse
|
30
|
Wang Y, Zhang X, Wang B, Xie Y, Wang Y, Jiang X, Wang R, Ren K. Evaluation of Renal Pathophysiological Processes Induced by an Iodinated Contrast Agent in a Diabetic Rabbit Model Using Intravoxel Incoherent Motion and Blood Oxygenation Level-Dependent Magnetic Resonance Imaging. Korean J Radiol 2020; 20:830-843. [PMID: 30993934 PMCID: PMC6470079 DOI: 10.3348/kjr.2018.0757] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To examine the potential of intravoxel incoherent motion (IVIM) and blood oxygen level-dependent (BOLD) magnetic resonance imaging for detecting renal changes after iodinated contrast-induced acute kidney injury (CI-AKI) development in a diabetic rabbit model. MATERIALS AND METHODS Sixty-two rabbits were randomized into 2 groups: diabetic rabbits with the contrast agent (DCA) and healthy rabbits with the contrast agent (NCA). In each group, 6 rabbits underwent IVIM and BOLD imaging at 1 hour, 1 day, 2 days, 3 days, and 4 days after an iohexol injection while 5 rabbits were selected to undergo blood and histological examinations at these specific time points. Iohexol was administrated at a dose of 2.5 g I/kg of body weight. Further, the apparent transverse relaxation rate (R2*), average pure molecular diffusion coefficient (D), pseudo-diffusion coefficient (D*), and perfusion fraction (f) were calculated. RESULTS The D and f values of the renal cortex (CO) and outer medulla (OM) were significantly decreased compared to baseline values in the 2 groups 1 day after the iohexol injection (p < 0.05). A marked reduction in the D* values for both the CO and OM was also observed after 1 hour in each group (p < 0.05). In the OM, a persistent elevation of the R2* was detected for 4 days in the DCA group (p < 0.05). Histopathological changes were prominent, and the pathological features of CI-AKI aggravated in the DCA group until day 4. The D, f, and R2* values significantly correlated with the histological damage scores, hypoxia-inducible transcription factor-1α expression scores, and serum creatinine levels. CONCLUSION A combination of IVIM and BOLD imaging may serve as a noninvasive method for detecting and monitoring CI-AKI in the early stages in the diabetic kidney.
Collapse
Affiliation(s)
- Yongfang Wang
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Xin Zhang
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Bin Wang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yang Xie
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Yi Wang
- Department of Radiology, The Qianfoshan Hospital of Shandong Province, Jinan, China
| | - Xuan Jiang
- Cardiac Surgery, First Hospital of China Medical University, Shenyang, China
| | - Rongjia Wang
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Ke Ren
- Department of Radiology, First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
31
|
Bane O, Mendichovszky IA, Milani B, Dekkers IA, Deux JF, Eckerbom P, Grenier N, Hall ME, Inoue T, Laustsen C, Lerman LO, Liu C, Morrell G, Pedersen M, Pruijm M, Sadowski EA, Seeliger E, Sharma K, Thoeny H, Vermathen P, Wang ZJ, Serafin Z, Zhang JL, Francis ST, Sourbron S, Pohlmann A, Fain SB, Prasad PV. Consensus-based technical recommendations for clinical translation of renal BOLD MRI. MAGMA (NEW YORK, N.Y.) 2019. [PMID: 31768797 DOI: 10.1007/s10334‐019‐00802‐x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Harmonization of acquisition and analysis protocols is an important step in the validation of BOLD MRI as a renal biomarker. This harmonization initiative provides technical recommendations based on a consensus report with the aim to move towards standardized protocols that facilitate clinical translation and comparison of data across sites. We used a recently published systematic review paper, which included a detailed summary of renal BOLD MRI technical parameters and areas of investigation in its supplementary material, as the starting point in developing the survey questionnaires for seeking consensus. Survey data were collected via the Delphi consensus process from 24 researchers on renal BOLD MRI exam preparation, data acquisition, data analysis, and interpretation. Consensus was defined as ≥ 75% unanimity in response. Among 31 survey questions, 14 achieved consensus resolution, 12 showed clear respondent preference (65-74% agreement), and 5 showed equal (50/50%) split in opinion among respondents. Recommendations for subject preparation, data acquisition, processing and reporting are given based on the survey results and review of the literature. These technical recommendations are aimed towards increased inter-site harmonization, a first step towards standardization of renal BOLD MRI protocols across sites. We expect this to be an iterative process updated dynamically based on progress in the field.
Collapse
Affiliation(s)
- Octavia Bane
- BioMedical Engineering and Imaging Institute and Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Iosif A Mendichovszky
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Bastien Milani
- Center for BioMedical Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ilona A Dekkers
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean-Francois Deux
- Department of Radiology, Groupe Hospitalier Henri Mondor, Créteil, France
| | - Per Eckerbom
- Department of Radiology, Institution for Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Nicolas Grenier
- Department of Radiology, Université de Bordeaux, CHU de Bordeaux, Bordeaux, France
| | - Michael E Hall
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Tsutomu Inoue
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Christoffer Laustsen
- The MR Research Center Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Chunlei Liu
- Electrical Engineering and Computer Science, and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Glen Morrell
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Michael Pedersen
- Department of Clinical Medicine-Comparative Medicine Lab, Aarhus University Hospital, Aarhus, Denmark
| | - Menno Pruijm
- Nephrology and Hypertension Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Elizabeth A Sadowski
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Erdmann Seeliger
- Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Kanishka Sharma
- Imaging Biomarkers Group, Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Harriet Thoeny
- Department of Radiology, Hôpital Cantonal Fribourgois, University of Fribourg, Fribourg, Switzerland
| | - Peter Vermathen
- Departments for BioMedical Research and Radiology, Inselspital, Universitaetspital Bern, Bern, Switzerland
| | - Zhen J Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco Medical Center, San Francisco, CA, USA
| | - Zbigniew Serafin
- Department of Radiology, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Jeff L Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Susan T Francis
- Sir Peter Mansfield Centre, University of Notthingham, Notthingham, UK
| | - Steven Sourbron
- Imaging Biomarkers Group, Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sean B Fain
- Departments of Biomedical Engineering, Radiology, and Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Pottumarthi V Prasad
- Department of Radiology, Center for Advanced Imaging, NorthShore University Health System, Evanston, IL, USA.
| |
Collapse
|
32
|
Tanabe J, Ogura Y, Nakabayashi M, Nagai Y, Watanabe S, Sugaya T, Ohata K, Ichikawa D, Inoue K, Hoshino S, Kimura K, Shibagaki Y, Ono Y, Kamijo-Ikemori A. The Possibility of Urinary Liver-Type Fatty Acid-Binding Protein as a Biomarker of Renal Hypoxia in Spontaneously Diabetic Torii Fatty Rats. Kidney Blood Press Res 2019; 44:1476-1492. [PMID: 31734667 DOI: 10.1159/000503926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/04/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Renal hypoxia is an aggravating factor for tubulointerstitial damage, which is strongly associated with renal prognosis in diabetic kidney disease (DKD). Therefore, urinary markers that can detect renal hypoxia are useful for monitoring DKD. OBJECTIVE To determine the correlation between urinary liver-type fatty acid-binding protein (L-FABP) and renal hypoxia using a novel animal model of type 2 diabetes. METHODS Male spontaneously diabetic Torii (SDT) fatty rats (n = 6) were used as an animal model of type 2 diabetes. Age- and sex-matched Sprague-Dawley (SD) rats (n = 8) were used as controls. Body weight, systolic blood pressure, and blood glucose levels were measured at 8, 12, 16, and 24 weeks of age. Urine samples and serum and kidney tissues were collected at 24 weeks of age. Microvascular blood flow index (BFI) was measured using diffuse correlation spectroscopy before sampling both the serum and kidneys for the evaluation of renal microcirculation at the corticomedullary junction. RESULTS Obesity, hyperglycemia, and hypertension were observed in the SDT fatty rats. Focal glomerular sclerosis, moderate interstitial inflammation, and fibrosis were significantly more frequent in SDT fatty rats than in SD rats. While the frequency of peritubular endothelial cells and phosphoendothelial nitric oxide synthase levels were similar in both types of rats, the degree of renal hypoxia-inducible factor-1α (HIF-1α) expression was significantly higher (and with no change in renal vascular endothelial growth factor expression levels) in the SDT fatty rats. Urinary L-FABP levels were significantly higher and renal microvascular BFI was significantly lower in the SDT fatty rats than in the SD rats. Urinary L-FABP levels exhibited a significant positive correlation with renal HIF-1α expression and a significant negative correlation with renal microvascular BFI. CONCLUSIONS Urinary L-FABP levels reflect the degree of renal hypoxia in DKD in a type 2 diabetic animal model. Urinary L-FABP may thus prove useful as a renal hypoxia marker for monitoring DKD in patients with type 2 diabetes in clinical practice.
Collapse
Affiliation(s)
- Jun Tanabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Mikie Nakabayashi
- Department of Electronics and Bioinformatics, School of Science and Technology, Meiji University, Kanagawa, Japan
| | - Yoshio Nagai
- Division of Metabolism and Endocrinology, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Shiika Watanabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Keiichi Ohata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazuho Inoue
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yumie Ono
- Department of Electronics and Bioinformatics, School of Science and Technology, Meiji University, Kanagawa, Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan, .,Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan,
| |
Collapse
|
33
|
Feng YZ, Ye YJ, Cheng ZY, Hu JJ, Zhang CB, Qian L, Lu XH, Cai XR. Non-invasive assessment of early stage diabetic nephropathy by DTI and BOLD MRI. Br J Radiol 2019; 93:20190562. [PMID: 31603347 DOI: 10.1259/bjr.20190562] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Patients with diabetes mellitus, diabetic nephropathy (DN) and healthy donor were analyzed to test whether the early DN patients can be detected using both blood oxygenation level dependent (BOLD) and diffusion tensor imaging. METHODS This study was approved by the Ethics Committee of our hospital. MR images were acquired on a 3.0-Tesla MR system (Discovery MR750, General Electric, Milwaukee, WI). 30 diabetic patients were divided into NAU (normal to mildly increased albuminuria, N = 15) and MAU (moderately increased albuminuria, N = 15) group based on the absence or presence of microalbuminuria. 15 controls with sex- and age-matched were enrolled in the study. Prior to MRI scan, all participants were instructed to collect their fresh morning urine samples for quantitative measurement of urinary microalbumin and urinary creatinine. Then, the estimations of serum creatinine, serum uric acid, HbAlc and fasting plasma glucose as well as fundus examinations were performed in all subjects. Then, the values of albumin-creatinine ratio (ACR) and estimated glomerular filtration rate were also calculated. All subjects underwent renal diffusion tensor imaging (DTI) and BOLD acquisition after fasting for 4 h. Regions of interest were placed in renal medulla and cortex for evaluating apparent diffusion coefficient (ADC), fractional anisotropy (FA) and R2* values by two experienced radiologists. The consistency between the two observations was estimated using intragroup correlation coefficients. To test differences in ADC, FA and R2* values across the three groups, the data were analyzed using separate one-way ANOVAs. Post-hoc pair wise comparisons were then performed using t-test. To investigate the clinical relevance of imaging parameters in both regions across the three groups, the correlations of values of the ACR/estimated glomerular filtration rate and of the ADC/FA/R2* were calculated. RESULTS There was a high level of consistency of those ADC, FA and R2* values across the three groups on both renal cortex and medulla measured by the two doctors. The FA value of medulla in MAU group was lower than that in control (p < 0.01). The R2* value of medulla in the NAU group was higher than that in the control (p < 0.01), and the R2* value of medulla in the MAU group was lower than that in the control (p = 0.009) . Moreover, the current study revealed a decreasing trend in FA values of the renal medulla from the control group to NAU and MAU groups. Finally, a weak negatively correlation between medullary R2* and ACR was found in current study. CONCLUSION Medullary R2* value might be a new more sensitive predictor of early DN. Meanwhile, BOLD imaging detected the medullary hypoxia at the simply diabetic stage, while DTI didn't identify the medullary directional diffusion changes at this stage. Based on our assumption mentioned above, it's presumable that BOLD imaging may be more sensitive for assessment of the early renal function changes than DTI. These imaging techniques are more accurate and practical than conventional tests. ADVANCES IN KNOWLEDGE Non-invasive MRI was used to detect renal function changes at early DN stage.
Collapse
Affiliation(s)
- You-Zhen Feng
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Yao-Jiang Ye
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Zhong-Yuan Cheng
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Jun-Jiao Hu
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Chuang-Biao Zhang
- Endocrinology department, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | | | - Xiao-Hua Lu
- Endocrinology department, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Xiang-Ran Cai
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Bessho R, Takiyama Y, Takiyama T, Kitsunai H, Takeda Y, Sakagami H, Ota T. Hypoxia-inducible factor-1α is the therapeutic target of the SGLT2 inhibitor for diabetic nephropathy. Sci Rep 2019; 9:14754. [PMID: 31611596 PMCID: PMC6791873 DOI: 10.1038/s41598-019-51343-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/23/2019] [Indexed: 01/06/2023] Open
Abstract
Previous studies have demonstrated intrarenal hypoxia in patients with diabetes. Hypoxia-inducible factor (HIF)-1 plays an important role in hypoxia-induced tubulointerstitial fibrosis. Recent clinical trials have confirmed the renoprotective action of SGLT2 inhibitors in diabetic nephropathy. We explored the effects of an SGLT2 inhibitor, luseogliflozin on HIF-1α expression in human renal proximal tubular epithelial cells (HRPTECs). Luseogliflozin significantly inhibited hypoxia-induced HIF-1α protein expression in HRPTECs. In addition, luseogliflozin inhibited hypoxia-induced the expression of the HIF-1α target genes PAI-1, VEGF, GLUT1, HK2 and PKM. Although luseogliflozin increased phosphorylated-AMP-activated protein kinase α (p-AMPKα) levels, the AMPK activator AICAR did not changed hypoxia-induced HIF-1α expression. Luseogliflozin suppressed the oxygen consumption rate in HRPTECs, and subsequently decreased hypoxia-sensitive dye, pimonidazole staining under hypoxia, suggesting that luseogliflozin promoted the degradation of HIF-1α protein by redistribution of intracellular oxygen. To confirm the inhibitory effect of luseogliflozin on hypoxia-induced HIF-1α protein in vivo, we treated male diabetic db/db mice with luseogliflozin for 8 to 16 weeks. Luseogliflozin attenuated cortical tubular HIF-1α expression, tubular injury and interstitial fibronectin in db/db mice. Together, luseogliflozin inhibits hypoxia-induced HIF-1α accumulation by suppressing mitochondrial oxygen consumption. The SGLT2 inhibitors may protect diabetic kidneys by therapeutically targeting HIF-1α protein.
Collapse
Affiliation(s)
- Ryoichi Bessho
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Yumi Takiyama
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan.
| | - Takao Takiyama
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Hiroya Kitsunai
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Yasutaka Takeda
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Hidemitsu Sakagami
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Tsuguhito Ota
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan.
| |
Collapse
|
35
|
Nakamoto H, Yu XQ, Kim S, Origasa H, Zheng H, Chen J, Joo KW, Sritippayawan S, Chen Q, Chen HC, Tsubakihara Y, Tamai H, Song SH, Vaithilingam I, Lee KW, Shu KH, Hok-King Lo S, Isono M, Kurumatani H, Okada K, Kanoh H, Kiriyama T, Yamada S, Fujita T. Effects of Sustained-Release Beraprost in Patients With Primary Glomerular Disease or Nephrosclerosis: CASSIOPEIR Study Results. Ther Apher Dial 2019; 24:42-55. [PMID: 31119846 DOI: 10.1111/1744-9987.12840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/26/2019] [Accepted: 05/21/2019] [Indexed: 12/23/2022]
Abstract
TRK-100STP, a sustained-release preparation of the orally active prostacyclin analogue beraprost sodium, targets renal hypoxia. This study aimed to show the superiority of TRK-100STP over placebos in patients with chronic kidney disease (with either primary glomerular disease or nephrosclerosis) to determine the recommended dose. CASSIOPEIR (Chronic Renal Failure Asian Study with Oral PGI2 Derivative for Evaluating Improvement of Renal Function) was a randomized, double-blind, placebo-controlled study conducted at 160 sites in seven Asia-Pacific countries and regions. Eligible patients (n = 892) were randomized to TRK-100STP 120, 240 μg, or placebo for a treatment period of up to 4 years. The primary efficacy endpoint was time to first occurrence of a renal composite: doubling of serum creatinine or occurrence of end-stage renal disease. No significant differences were observed in composite endpoints between TRK-100STP and placebo (P = 0.5674). Hazard ratios (95% CI) in the TRK-100STP 120 and 240 μg vs. placebo groups were 0.98 (0.78, 1.22) and 0.91 (0.72, 1.14), respectively. The overall incidence of adverse events and adverse drug reactions was comparable between treatment arms.
Collapse
Affiliation(s)
| | - Xue-Qing Yu
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou City, China
| | - Suhnggwon Kim
- Seoul National University Hospital, Seoul K Clinic, Seoul, South Korea
| | | | - Hongguang Zheng
- The General Hospital of Shenyang Military Command, Shenyang City, China
| | - Jianghua Chen
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kwon Wook Joo
- Seoul National University Hospital, Seoul, South Korea
| | | | - Qinkai Chen
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hung-Chun Chen
- Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | | | | | | | | | - Kang Wook Lee
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, South Korea
| | | | - Stanley Hok-King Lo
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Jiang K, Ferguson CM, Lerman LO. Noninvasive assessment of renal fibrosis by magnetic resonance imaging and ultrasound techniques. Transl Res 2019; 209:105-120. [PMID: 31082371 PMCID: PMC6553637 DOI: 10.1016/j.trsl.2019.02.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is a useful biomarker for diagnosis and guidance of therapeutic interventions of chronic kidney disease (CKD), a worldwide disease that affects more than 10% of the population and is one of the major causes of death. Currently, tissue biopsy is the gold standard for assessment of renal fibrosis. However, it is invasive, and prone to sampling error and observer variability, and may also result in complications. Recent advances in diagnostic imaging techniques, including magnetic resonance imaging (MRI) and ultrasonography, have shown promise for noninvasive assessment of renal fibrosis. These imaging techniques measure renal fibrosis by evaluating its impacts on the functional, mechanical, and molecular properties of the kidney, such as water mobility by diffusion MRI, tissue hypoxia by blood oxygenation level dependent MRI, renal stiffness by MR and ultrasound elastography, and macromolecule content by magnetization transfer imaging. Other MR techniques, such as T1/T2 mapping and susceptibility-weighted imaging have also been explored for measuring renal fibrosis. Promising findings have been reported in both preclinical and clinical studies using these techniques. Nevertheless, limited specificity, sensitivity, and practicality in these techniques may hinder their immediate application in clinical routine. In this review, we will introduce methodologies of these techniques, outline their applications in fibrosis imaging, and discuss their limitations and pitfalls.
Collapse
Affiliation(s)
- Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
37
|
Hassan AED, Shaat EA, Deif MM, El Azhary NM, Omar EM. Effect of erythropoietin hormone supplementation on renal functions and the level of hypoxia-inducible factor-1α in rat kidneys with experimentally induced diabetic nephropathy. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2013.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Alaa El Din Hassan
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Eman A. Shaat
- Department of Biochemistry, Faculty of Medicine, Alexandria University, Egypt
| | - Maha M. Deif
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | | | - Eman M. Omar
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| |
Collapse
|
38
|
Morrell GR, Jeong EK, Shi X, Zhang L, Lee VSC. Continuous prospectively navigated multi-echo GRE for improved BOLD imaging of the kidneys. NMR IN BIOMEDICINE 2019; 32:e4078. [PMID: 30811061 PMCID: PMC6476650 DOI: 10.1002/nbm.4078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 06/09/2023]
Abstract
The objective of this study is to develop improved methods for renal blood oxygenation level dependent (BOLD) imaging. T2* mapping of the kidneys, or renal BOLD imaging, may depict renal oxygen levels and may be valuable as a noninvasive means of following the progression of renal disease. Current renal BOLD data is limited by imaging in a single breath hold, which results in low resolution and low signal-to-noise ratio (SNR). We compare a new free-breathing renal BOLD method with conventional breath-hold BOLD (BH-BOLD). A multi-echo GRE sequence with continuous prospective respiratory navigation and real-time feedback was developed that allows high resolution and high SNR renal BOLD imaging with constant sequence repetition time (TR) during free-breathing BOLD (FB-BOLD). The sequence was evaluated in 10 normal volunteers and compared with conventional BH-BOLD. Scan time for the FB-BOLD sequence was approximately three minutes, compared with 15 seconds for the BH-BOLD sequence. SNR of source images and residual error of T2* fitting were compared between the two methods. The FB-BOLD sequence produced motion-free T2* maps of the kidneys with SNR 1.9 times higher than BH-BOLD images. Residual error of T2* fitting was consistently lower in the right kidney with FB-BOLD (30% less than BH-BOLD) but higher in the left kidney (80% more than BH-BOLD), likely related to placement of the navigator on the right hemidiaphragm. A free-breathing prospectively navigated renal BOLD sequence allows flexible tradeoff between scan time, resolution, and SNR.
Collapse
Affiliation(s)
- Glen Robert Morrell
- Utah Center for Advanced Imaging Research, University of Utah Radiology Department, Salt Lake City, Utah, USA
| | - Eun-Kee Jeong
- Utah Center for Advanced Imaging Research, University of Utah Radiology Department, Salt Lake City, Utah, USA
| | - Xianfeng Shi
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, Utah, 84108, USA
| | - Lei Zhang
- Utah Center for Advanced Imaging Research, University of Utah Radiology Department, Salt Lake City, Utah, USA
| | | |
Collapse
|
39
|
Berchtold L, Friedli I, Crowe LA, Martinez C, Moll S, Hadaya K, de Perrot T, Combescure C, Martin PY, Vallée JP, de Seigneux S. Validation of the corticomedullary difference in magnetic resonance imaging-derived apparent diffusion coefficient for kidney fibrosis detection: a cross-sectional study. Nephrol Dial Transplant 2019; 35:937-945. [DOI: 10.1093/ndt/gfy389] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
Kidney cortical interstitial fibrosis (IF) is highly predictive of renal prognosis and is currently assessed by the evaluation of a biopsy. Diffusion magnetic resonance imaging (MRI) is a promising tool to evaluate kidney fibrosis via the apparent diffusion coefficient (ADC), but suffers from inter-individual variability. We recently applied a novel MRI protocol to allow calculation of the corticomedullary ADC difference (ΔADC). We here present the validation of ΔADC for fibrosis assessment in a cohort of 164 patients undergoing biopsy and compare it with estimated glomerular filtration rate (eGFR) and other plasmatic parameters for the detection of fibrosis.
Methods
This monocentric cross-sectional study included 164 patients undergoing renal biopsy at the Nephrology Department of the University Hospital of Geneva between October 2014 and May 2018. Patients underwent diffusion-weighted imaging, and T1 and T2 mappings, within 1 week after biopsy. MRI results were compared with gold standard histology for fibrosis assessment.
Results
Absolute cortical ADC or cortical T1 values correlated poorly to IF assessed by the biopsy, whereas ΔADC was highly correlated to IF (r=−0.52, P < 0.001) and eGFR (r = 0.37, P < 0.01), in both native and allograft patients. ΔT1 displayed a lower, but significant, correlation to IF and eGFR, whereas T2 did not correlate to IF nor to eGFR. ΔADC, ΔT1 and eGFR were independently associated with kidney fibrosis, and their combination allowed detection of extensive fibrosis with good specificity.
Conclusion
ΔADC is better correlated to IF than absolute cortical or medullary ADC values. ΔADC, ΔT1 and eGFR are independently associated to IF and allow the identification of patients with extensive IF.
Collapse
Affiliation(s)
- Lena Berchtold
- Service and Laboratory of Nephrology, Department for Statistics, Department of Internal Medicine Specialties and of Physiology and Metabolism, University Hospital and University of Geneva, Geneva, Switzerland
| | - Iris Friedli
- Service of Radiology, Department for Statistics, Department of Radiology and Medical Informatics, University Hospital and University of Geneva, Geneva, Switzerland
| | - Lindsey A Crowe
- Service of Radiology, Department for Statistics, Department of Radiology and Medical Informatics, University Hospital and University of Geneva, Geneva, Switzerland
| | - Chantal Martinez
- Service and Laboratory of Nephrology, Department for Statistics, Department of Internal Medicine Specialties and of Physiology and Metabolism, University Hospital and University of Geneva, Geneva, Switzerland
| | - Solange Moll
- Department of Clinical Pathology, Institute of Clinical Pathology, University Hospital of Geneva, Geneva, Switzerland
| | - Karine Hadaya
- Service and Laboratory of Nephrology, Department for Statistics, Department of Internal Medicine Specialties and of Physiology and Metabolism, University Hospital and University of Geneva, Geneva, Switzerland
| | - Thomas de Perrot
- Service of Radiology, Department for Statistics, Department of Radiology and Medical Informatics, University Hospital and University of Geneva, Geneva, Switzerland
| | - Christophe Combescure
- CRC & Division of Clinical-Epidemiology, Department of Health and Community Medicine, University of Geneva and University Hospitals of Geneva, Geneva, Switzerland
| | - Pierre-Yves Martin
- Service and Laboratory of Nephrology, Department for Statistics, Department of Internal Medicine Specialties and of Physiology and Metabolism, University Hospital and University of Geneva, Geneva, Switzerland
| | - Jean-Paul Vallée
- Service of Radiology, Department for Statistics, Department of Radiology and Medical Informatics, University Hospital and University of Geneva, Geneva, Switzerland
| | - Sophie de Seigneux
- Service and Laboratory of Nephrology, Department for Statistics, Department of Internal Medicine Specialties and of Physiology and Metabolism, University Hospital and University of Geneva, Geneva, Switzerland
| |
Collapse
|
40
|
Szalat A, Perlman A, Muszkat M, Khamaisi M, Abassi Z, Heyman SN. Can SGLT2 Inhibitors Cause Acute Renal Failure? Plausible Role for Altered Glomerular Hemodynamics and Medullary Hypoxia. Drug Saf 2018; 41:239-252. [PMID: 28952138 DOI: 10.1007/s40264-017-0602-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sodium-glucose co-transporter-2 inhibitors (SGLT2i) provide outstanding long-term cardiovascular and renal protection in high-risk patients with type 2 diabetes mellitus. Yet, despite encouraging renal safety outcomes reported in the EMPA-REG study, scattered reports suggest that there might be a risk for acute kidney injury (AKI), which may occasionally be fatal or might require renal replacement therapy. Reduced trans-glomerular pressure with a modest decline in kidney function, an inherent characteristic of SGLT2i therapy, conceivably forms the basis for the long-term renal protection, resembling agents that block the renin-angiotensin-aldosterone (RAAS) axis. Yet, a major decline in kidney function occasionally occurs, often associated with an acute illness or with specific co-administered medications. SGLT2i may lead to AKI by (a) effective volume depletion, due to excessive diuresis, particularly in hemodynamically unstable and volume-depleted patients; (b) excessive decline in trans-glomerular pressure, specifically in patients on RAAS blockade; and (c) induction of renal medullary hypoxic injury, related to enhanced distal tubular transport, especially with concomitant use of agents impairing medullary oxygenation, such as non-steroidal anti-inflammatory drugs and radiocontrast agents. The risk of developing renal impairment with SGLT2i and the role of these suggested mechanisms are yet to be defined, as there are conflicting data and inconsistent reporting with the various agents currently in use.
Collapse
Affiliation(s)
- Auryan Szalat
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus, P.O. Box 24035, 91240, Jerusalem, Israel
| | - Amichai Perlman
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus, P.O. Box 24035, 91240, Jerusalem, Israel
| | - Mordechai Muszkat
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus, P.O. Box 24035, 91240, Jerusalem, Israel
| | - Mogher Khamaisi
- Department of Internal Medicine, Rambam Medical Center, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology and Systems Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Laboratory Medicine, Rambam Health Campus, Haifa, Israel
| | - Samuel N Heyman
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus, P.O. Box 24035, 91240, Jerusalem, Israel.
| |
Collapse
|
41
|
Friederich-Persson M, Persson P, Hansell P, Palm F. Deletion of Uncoupling Protein-2 reduces renal mitochondrial leak respiration, intrarenal hypoxia and proteinuria in a mouse model of type 1 diabetes. Acta Physiol (Oxf) 2018; 223:e13058. [PMID: 29480974 DOI: 10.1111/apha.13058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/19/2018] [Accepted: 02/19/2018] [Indexed: 12/26/2022]
Abstract
AIM Uncoupling protein-2 (UCP-2) can induce mitochondrial uncoupling in the diabetic kidney. Although mitochondrial uncoupling reduces oxidative stress originating from the mitochondria and can be regarded as a protective mechanism, the increased oxygen consumption occurring secondarily to increased mitochondria uncoupling, that is leak respiration, may contribute to kidney tissue hypoxia. Using UCP-2-/- mice, we tested the hypothesis that UCP-2-mediated leak respiration is important for the development of diabetes-induced intrarenal hypoxia and proteinuria. METHODS Kidney function, in vivo oxygen metabolism, urinary protein leakage and mitochondrial function were determined in wild-type and UCP-2-/- mice during normoglycaemia and 2 weeks after diabetes induction. RESULTS Diabetic wild-type mice displayed mitochondrial leak respiration, pronounced intrarenal hypoxia, proteinuria and increased urinary KIM-1 excretion. However, diabetic UCP-2-/- mice did not develop increased mitochondrial leak respiration and presented with normal intrarenal oxygen levels, urinary protein and KIM-1 excretion. CONCLUSION Although functioning as an antioxidant system, mitochondria uncoupling is always in co-occurrence with increased oxygen consumption, that is leak respiration; a potentially detrimental side effect as it can result in kidney tissue hypoxia; an acknowledged unifying pathway to nephropathy. Indeed, this study demonstrates a novel mechanism in which UCP-2-mediated mitochondrial leak respiration is necessary for the development of diabetes-induced intrarenal tissue hypoxia and proteinuria.
Collapse
Affiliation(s)
- M. Friederich-Persson
- Division of Integrative Physiology; Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - P. Persson
- Division of Integrative Physiology; Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - P. Hansell
- Division of Integrative Physiology; Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - F. Palm
- Division of Integrative Physiology; Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| |
Collapse
|
42
|
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: 1.8] [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
|
43
|
The value of blood oxygen level dependent (BOLD) imaging in evaluating post-operative renal function outcomes after laparoscopic partial nephrectomy. Eur Radiol 2018; 28:5035-5043. [PMID: 29948077 DOI: 10.1007/s00330-018-5525-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/22/2018] [Accepted: 05/03/2018] [Indexed: 10/14/2022]
Abstract
OBJECTIVES To assess the ability of renal blood oxygen level dependent (BOLD) MRI metrics to predict post-operative renal function. METHODS We studied 152 patients who underwent laparoscopic partial nephrectomy (LPN) and renal MRI examination including BOLD. Short-term and long-term renal function was evaluated using the glomerular filtration rate (GFR) derived from renal scintigraphy. Renal function decline was assessed as the absolute decline (AD), percentage decline (PD) and optimal renal function preservation (OP). T2* values were analysed in the renal cortex and medulla ipsilateral and contralateral to the tumour. Clinical characteristics and imaging metrics were evaluated using univariate and multivariate linear regression analyses. Risk factors obtained using BOLD metrics (determined by multivariate regression) were then combined and compared with RENAL scores to predict OP. RESULTS Increasing warm ischaemia time (WIT), resected and ischaemic volume (RAIV), larger tumour size, higher RENAL score and lower preoperative GFR were short-term risk factors for AD, while increasing WIT and lower preoperative GFR were significant for long-term outcomes. Increasing WIT, RAIV, lower T2* value in the cortex and higher T2* value in the medulla on the ipsilateral side were short-term risk factors for PD, while all of the above factors (except WIT and RAIV) were significant for long-term outcomes. The performance of the combination of T2* values in the cortex and medulla on the ipsilateral side to tumour in predicting OP was better than RENAL score (AUC 0.762 vs 0.634, p = 0.013). CONCLUSIONS Renal BOLD-MRI metrics could provide useful information to the clinician in predicting post-operative renal function outcomes. KEY POINTS • Renal fMRI metrics may be useful for prediction of renal functional outcomes and merit further study. • Renal fMRI metrics may reflect degree of baseline disease and ability to tolerate warm ischaemia. • Combination of T2* values was better than RENAL score for predicting OP.
Collapse
|
44
|
Sun J, Lv J, Zhang W, Li L, Lv J, Geng Y, Yin A. Combination with miR-124a improves the protective action of BMSCs in rescuing injured rat podocytes from abnormal apoptosis and autophagy. J Cell Biochem 2018; 119:7166-7176. [PMID: 29904949 DOI: 10.1002/jcb.26771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 02/02/2018] [Indexed: 12/13/2022]
Abstract
This in vitro study was performed to identify the role of miR-124a in bone marrow stromal stem cells (BMSCs) therapy for H2 O2 -induced rat podocyte injury, and determine whether combination treatment with miR-124a could improve the protective effect of BMSCs. Cell viability of podocytes was detected by CCK-8 assay. Detection of ROS level, apoptotic rate, and autophagy rate was carried out using flow cytometry assays. Oxidative stress parameters were analyzed using the ELISA assays. MiR-124a and mRNA levels were determined using real-time PCR. Protein expression was detected using Western blotting. Our study revealed a pivotal role of miR-124a in the protective action of BMSCs on podocyte injury driven by oxidative stress. BMSCs could rescue injured podocytes from aberrant apoptosis and autophagy by regulating cleaved caspase-3, Bax, Bcl-2, LC3-II/I, and p62. Suppression of the PI3 K/Akt/mTOR signaling pathway is likely one of the main mechanisms underlying the protective action of BMSCs transfected with miR-124a. Our study revealed that miR-124a further improves the protective effect of BMSCs in injured podocytes. Thus, the combination of BMSCs and microRNAs could be a beneficial treatment for renal diseases in the near future.
Collapse
Affiliation(s)
- Jiping Sun
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Lv
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenjing Zhang
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lili Li
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jia Lv
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yingzhou Geng
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Aiping Yin
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
45
|
Li ZC, Cai YZ, Tang ZG, Zuo PL, Liu RB, Liu F. Lipo-prostaglandin E1 improves renal hypoxia evaluated by BOLD-MRI in patients with diabetic kidney disease. Clin Imaging 2018; 50:239-242. [PMID: 29689479 DOI: 10.1016/j.clinimag.2018.04.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/12/2018] [Accepted: 04/16/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To evaluate the effect of lipo-PGE1 on renal hypoxia in patients with DKD by BOLD-MRI. MATERIALS AND METHODS All patients were divided into DKD group and CKD-without-diabetes group. All patients received intravenous 10 μg lipo-PGE1 once daily for 14 days. BOLD-MRI was performed before and after lipo-PGE1 administration to acquire renal CR2* and MR2* values. RESULTS Renal MR2* value in DKD group after lipo-PGE1 treatment were significantly decreased compared with the baseline. However, no significant differences in MR2* values were found in the CKD-without-diabetes group. CONCLUSIONS Lipo-PGE1 was shown to improve kidney medullary oxygenation in patients with DKD.
Collapse
Affiliation(s)
- Zhi-Cheng Li
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Department of Nephrology, Zigong First People's Hospital, Zigong, Sichuan 643000, China
| | - Yu-Zhe Cai
- Department of Radiology, West China Hospital of Sichuan University, No. 37 Guoxue Street, Wuhou District, Chengdu, Sichuan 610041, China
| | - Zhi-Gang Tang
- Department of Nephrology, Zigong First People's Hospital, Zigong, Sichuan 643000, China
| | - Pan-Li Zuo
- Siemens Healthcare, MR Collaboration NE Asia, Beijing, China
| | - Rong-Bo Liu
- Department of Radiology, West China Hospital of Sichuan University, No. 37 Guoxue Street, Wuhou District, Chengdu, Sichuan 610041, China
| | - Fang Liu
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
46
|
Yu SMW, Bonventre JV. Acute Kidney Injury and Progression of Diabetic Kidney Disease. Adv Chronic Kidney Dis 2018; 25:166-180. [PMID: 29580581 DOI: 10.1053/j.ackd.2017.12.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 12/23/2022]
Abstract
Diabetic kidney disease, commonly termed diabetic nephropathy (DN), is the most common cause of end-stage kidney disease (ESKD) worldwide. The characteristic histopathology of DN includes glomerular basement membrane thickening, mesangial expansion, nodular glomerular sclerosis, and tubulointerstitial fibrosis. Diabetes is associated with a number of metabolic derangements, such as reactive oxygen species overproduction, hypoxic state, mitochondrial dysfunction, and inflammation. In the past few decades, our knowledge of DN has advanced considerably although much needs to be learned. The traditional paradigm of glomerulus-centered pathophysiology has expanded to the tubule-interstitium, the immune response and inflammation. Biomarkers of proximal tubule injury have been shown to correlate with DN progression, independent of traditional glomerular injury biomarkers such as albuminuria. In this review, we summarize mechanisms of increased susceptibility to acute kidney injury in diabetes mellitus and the roles played by many kidney cell types to facilitate maladaptive responses leading to chronic and end-stage kidney disease.
Collapse
|
47
|
Wang Q, Guo C, Zhang L, Zhang R, Wang Z, Xu Y, Xiao W. BOLD MRI to evaluate early development of renal injury in a rat model of diabetes. J Int Med Res 2018; 46:1391-1403. [PMID: 29446322 PMCID: PMC6091826 DOI: 10.1177/0300060517743826] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective To investigate changes in renal oxygenation levels by blood-oxygenation-level dependent (BOLD)-magnetic resonance imaging (MRI), and to evaluate BOLD-MRI for detecting early diabetic renal injury. Methods Seventy-five rats, with unilateral nephrectomy, were randomly divided into streptozotocin-induced diabetes mellitus (DM, n = 65) and normal control (NC, n = 10) groups. BOLD-MRI scans were performed at baseline (both groups) and at 3, 7, 14, 21, 28, 35, 42, 49, 56, 63 and 70 days (DM only). Renal cortical (C) and medullary (M) R2* signals were measured and R2* medulla/cortex ratio (MCR) was calculated. Results DM-group CR2* and MR2* values were significantly higher than NC values following diabetes induction. R2* values increased gradually and peaked at day 35 (CR2*, 33.95 ± 0.34 s–1; MR2*, 43.79 ± 1.46 s–1), then dropped gradually (CR2*, 33.17 ± 0.69 s–1; MR2*, 41.61 ± 0.95 s–1 at day 70). DM-group MCR rose gradually from 1.12 to 1.32 at day 42, then decreased to 1.25 by day 70. Conclusions BOLD-MRI can be used to non-invasively evaluate renal hypoxia and early diabetic renal injury in diabetic rats. MCR may be adopted to reflect dynamic changes in renal hypoxia.
Collapse
Affiliation(s)
- Qidong Wang
- 1 Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,*These authors contributed equally to this work
| | - Chuangen Guo
- 1 Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,*These authors contributed equally to this work
| | - Lan Zhang
- 2 Department of Radiology, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Rui Zhang
- 1 Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhaoming Wang
- 3 Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Xu
- 4 Department of Nephrology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenbo Xiao
- 1 Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,2 Department of Radiology, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
48
|
Mariager CØ, Nielsen PM, Qi H, Ringgaard S, Laustsen C. Hyperpolarized 13
C,15
N2
-urea T2
relaxation changes in acute kidney injury. Magn Reson Med 2017; 80:696-702. [DOI: 10.1002/mrm.27050] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 01/18/2023]
Affiliation(s)
| | - Per Mose Nielsen
- MR Research Centre, Department of Clinical Medicine; Aarhus University; Aarhus Denmark
| | - Haiyun Qi
- MR Research Centre, Department of Clinical Medicine; Aarhus University; Aarhus Denmark
| | - Steffen Ringgaard
- MR Research Centre, Department of Clinical Medicine; Aarhus University; Aarhus Denmark
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine; Aarhus University; Aarhus Denmark
| |
Collapse
|
49
|
Wang Y, Zhang H, Zhang R, Zhao Z, Xu Z, Wang L, Liu R, Gao F. Investigation of aquaporins and apparent diffusion coefficient from ultra-high b-values in a rat model of diabetic nephropathy. Eur Radiol Exp 2017; 1:13. [PMID: 29708187 PMCID: PMC5909346 DOI: 10.1186/s41747-017-0016-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/12/2017] [Indexed: 02/05/2023] Open
Abstract
Background To assess kidney damage in a rat model of type-2 diabetic nephropathy based on apparent diffusion coefficient (ADC) data obtained from ultra-high b-values and discuss its relationship to the expression of aquaporins (AQPs). Methods This study was approved by the institutional Animal Care and Use Committee. Thirty male Sprague-Dawley rats were randomised into two groups: (1) untreated controls and (2) diabetes mellitus (DM). All rats underwent diffusion-weighted imaging (DWI) with 18 b-values (0–4500 s/mm2). Maps of low ADC (ADClow), standard ADC (ADCst) and ultra-high ADC (ADCuh) were calculated from low b-values (0–200 s/mm2), standard b-values (300–1500 s/mm2) and ultra-high b-values (1700–4500 s/mm2), respectively. The expression of AQPs in the kidneys was studied using immunohistochemistry. Laboratory parameters of diabetic and kidney functions, ADClow, ADCst, ADCuh, and the optical density (OD) of AQP expression in the two groups were compared using an independent t test. Correlations between ADCs and the OD of AQP expression were evaluated by Pearson’s correlation analysis. Results ADCuh were significantly higher in the cortex (CO), outer stripe of the outer medulla (OS) and inner stripe of the outer medulla (IS), and the OD values of AQ-2 were significantly higher in the OS, IS and inner medulla (IM) in DM animals compared with control animals. ADCuh and OD values of AQP-2 expression were positively correlated in the OS, IS and IM of the kidney. Conclusions ADCuh may work as useful metrics for early detection of kidney damage in diabetic nephropathy and may be associated with AQP-2 expression.
Collapse
Affiliation(s)
- Yu Wang
- 1Department of Radiology, West China Hospital of Sichuan University, No. 37, Guoxue Lane outside the south, Wuhou District Chengdu, China
| | - Heng Zhang
- 1Department of Radiology, West China Hospital of Sichuan University, No. 37, Guoxue Lane outside the south, Wuhou District Chengdu, China
| | - Ruzhi Zhang
- 1Department of Radiology, West China Hospital of Sichuan University, No. 37, Guoxue Lane outside the south, Wuhou District Chengdu, China
| | - Zhoushe Zhao
- General Electronic Company Healthcare (China), Beijing, China
| | - Ziqian Xu
- 1Department of Radiology, West China Hospital of Sichuan University, No. 37, Guoxue Lane outside the south, Wuhou District Chengdu, China
| | - Lei Wang
- 1Department of Radiology, West China Hospital of Sichuan University, No. 37, Guoxue Lane outside the south, Wuhou District Chengdu, China
| | - Rongbo Liu
- 1Department of Radiology, West China Hospital of Sichuan University, No. 37, Guoxue Lane outside the south, Wuhou District Chengdu, China
| | - Fabao Gao
- 1Department of Radiology, West China Hospital of Sichuan University, No. 37, Guoxue Lane outside the south, Wuhou District Chengdu, China
| |
Collapse
|
50
|
GYY4137, a Hydrogen Sulfide Donor Modulates miR194-Dependent Collagen Realignment in Diabetic Kidney. Sci Rep 2017; 7:10924. [PMID: 28883608 PMCID: PMC5589897 DOI: 10.1038/s41598-017-11256-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/22/2017] [Indexed: 12/22/2022] Open
Abstract
The relationship between hydrogen sulfide (H2S), microRNAs (miRs), matrix metalloproteinases (MMPs) and poly-ADP-ribose-polymerase-1 (PARP-1) in diabetic kidney remodeling remains mostly obscured. We aimed at investigating whether alteration of miR-194-dependent MMPs and PARP-1 causes renal fibrosis in diabetes kidney, and whether H2S ameliorates fibrosis. Wild type, diabetic Akita mice as well as mouse glomerular endothelial cells (MGECs) were used as experimental models, and GYY4137 as H2S donor. In diabetic mice, plasma H2S levels were decreased while ROS and expression of its modulator (ROMO1) were increased. In addition, alteration of MMPs-9, −13 and −14 expression, PARP-1, HIF1α, and increased collagen biosynthesis as well as collagen cross-linking protein, P4HA1 and PLOD2 were observed along with diminished vascular density in diabetic kidney. These changes were ameliorated by GYY4137. Further, downregulated miRNA-194 was normalized by GYY4137 in diabetic kidney. Similar results were obtained in in vitro condition. Interestingly, miR-194 mimic also diminished ROS production, and normalized ROMO1, MMPs-9, −13 and −14, and PARP-1 along with collagen biosynthesis and cross-linking protein in HG condition. We conclude that decrease H2S diminishes miR-194, induces collagen deposition and realignment leading to fibrosis and renovascular constriction in diabetes. GYY4137 mitigates renal fibrosis in diabetes through miR-194-dependent pathway.
Collapse
|