1
|
Song J, Chen Y, Chen Y, Qiu M, Xiang W, Ke B, Fang X. DKK3 promotes renal fibrosis by increasing MFF-mediated mitochondrial dysfunction in Wnt/β-catenin pathway-dependent manner. Ren Fail 2024; 46:2343817. [PMID: 38682264 PMCID: PMC11060011 DOI: 10.1080/0886022x.2024.2343817] [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: 06/14/2023] [Accepted: 04/11/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) lacks effective treatments and renal fibrosis (RF) is one of CKD's outcomes. Dickkopf 3 (DKK3) has been identified as an agonist in CKD. However, the underlying mechanisms of DKK3 in CKD are not fully understood. METHODS H2O2-treated HK-2 cells and ureteric obstruction (UUO) mice were used as RF models. Biomarkers, Masson staining, PAS staining, and TUNEL were used to assess kidney function and apoptosis. Oxidative stress and mitochondria function were also evaluated. CCK-8 and flow cytometry were utilized to assess cell viability and apoptosis. Western blotting, IHC, and qRT-PCR were performed to detect molecular expression levels. Immunofluorescence was applied to determine the subcellular localization. Dual luciferase assay, MeRIP, RIP, and ChIP were used to validate the m6A level and the molecule interaction. RESULTS DKK3 was upregulated in UUO mouse kidney tissue and H2O2-treated HK-2 cells. Knockdown of DKK3 inhibited oxidative stress, maintained mitochondrial homeostasis, and alleviated kidney damage and RF in UUO mice. Furthermore, DKK3 silencing suppressed HK-2 cell apoptosis, oxidative stress, and mitochondria fission. Mechanistically, DKK3 upregulation was related to the high m6A level regulated by METTL3. DKK3 activated TCF4/β-catenin and enhanced MFF transcriptional expression by binding to its promoter. Overexpression of MFF reversed in the inhibitory effect of DKK3 knockdown on cell damage. CONCLUSION Upregulation of DKK3 caused by m6A modification activated the Wnt/β-catenin pathway to increase MFF transcriptional expression, leading to mitochondrial dysfunction and oxidative stress, thereby promoting RF progression.
Collapse
Affiliation(s)
- Jianling Song
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Yanxia Chen
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Yan Chen
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Minzi Qiu
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Wenliu Xiang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Ben Ke
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| |
Collapse
|
2
|
Fallois JD, Günzel A, Daniel C, Stumpf J, Busch M, Pein U, Paliege A, Amann K, Wiech T, Hantmann E, Wolf G, Pfeifer F, Girndt M, Lindner TH, Weimann A, Seehofer D, Bachmann A, Budde K, Biemann R, Isermann B, Engel C, Dittrich K, Hugo C, Halbritter J. Deceased donor urinary Dickkopf-3 associates with future allograft function following kidney transplantation. Am J Transplant 2024:S1600-6135(24)00571-9. [PMID: 39303796 DOI: 10.1016/j.ajt.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Predicting future kidney allograft function is challenging. Novel biomarkers, such as urinary Dickkopf-3 (uDKK3), may help guide donor selection and improve allograft outcomes. In this prospective multicenter pilot trial, we investigated whether donor uDKK3 reflects organ quality and is associated with future allograft function. We measured uDKK3/crea ratios (uDKK3/crea) from 95 deceased and 46 living kidney donors. Prenephrectomy uDKK3/crea levels were 100× higher in deceased than in living donors (9888 pg/mg vs 113 pg/mg; P < .001). Among deceased donor transplantations, recipients were stratified by their corresponding uDKK3/crea donor levels ranging below (group A, n = 68) or above (group B, n = 65) median. The primary end point of best estimated glomerular filtration rate (eGFR) within the first 3 months after kidney transplantation was superior in group A (56.3 mL/min/1.73 m2) than that in group B (44.2 mL/min/1.73 m2; P = .0139). Second, the composite clinical end point consisting of death, allograft failure or eGFR decline >50% occurred less frequent in group A. By mixed linear regression modeling, donor uDKK3/crea remained an independent predictor of eGFR after transplantation, with a slope of -4.282 mL/min/1.73 m2 per logarithmic increase in donor uDKK3/crea. In summary, uDKK3 may serve as a noninvasive, donor-dependent biomarker for assessing organ quality and future allograft function.
Collapse
Affiliation(s)
- Jonathan de Fallois
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany.
| | - Anna Günzel
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Christoph Daniel
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Julian Stumpf
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Martin Busch
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | - Ulrich Pein
- Department of Internal Medicine II, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Alexander Paliege
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Kerstin Amann
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Thorsten Wiech
- Nephropathology Section, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Hantmann
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany; Department of Nephrology and Medical Intensive Care, Charité Berlin, Berlin, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | - Felix Pfeifer
- German Organ Procurement Organization (DSO), Region East, Leipzig, Germany
| | - Matthias Girndt
- Department of Internal Medicine II, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Tom H Lindner
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Antje Weimann
- Division of Visceral Surgery and Transplantation Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Daniel Seehofer
- Division of Visceral Surgery and Transplantation Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Anette Bachmann
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Berlin, Berlin, Germany
| | - Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Christoph Engel
- Institute for Medical Informatics, Statistics, and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Katalin Dittrich
- German Organ Procurement Organization (DSO), Region East, Leipzig, Germany; Division of Pediatric Nephrology and Transplantation, Department of Pediatrics, University Medical Center Leipzig, Leipzig, Germany
| | - Christian Hugo
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jan Halbritter
- Division of Nephrology, Department of Internal Medicine, University Medical Center Leipzig, Leipzig, Germany; Department of Nephrology and Medical Intensive Care, Charité Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Peschard VG, Scherzer R, Katz R, Chen TK, Bullen AL, Campos K, Estrella MM, Ix JH, Shlipak MG. Association of Urinary Dickkopf-3 Levels with Cardiovascular Events and Kidney Disease Progression in Systolic Blood Pressure Intervention Trial. KIDNEY360 2024; 5:690-697. [PMID: 38472135 PMCID: PMC11146650 DOI: 10.34067/kid.0000000000000413] [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: 01/21/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024]
Abstract
Key Points In unadjusted analyses, elevated urinary Dickkopf-3 levels were strongly associated with higher risks of cardiovascular disease, ESKD, AKI, and mortality. However, associations were substantially weakened after adjustment for eGFR and albuminuria, suggesting limited prognostic value. Background Urinary Dickkopf-3 (uDKK3) is a tubular epithelial-derived profibrotic protein secreted into the urine under tubular stress. It is associated with kidney disease progression in persons with CKD and diabetes and postoperative and contrast-associated AKI. We explored associations of uDKK3 with cardiovascular disease (CVD), kidney, and mortality outcomes within the subset of Systolic Blood Pressure Intervention Trial participants with nondiabetic CKD. Methods We included 2344 participants with eGFR <60 ml/min per 1.73 m2 at baseline. We used Cox proportional hazards models to evaluate associations of uDKK3 with CVD (acute decompensated heart failure, myocardial infarction, acute coronary syndrome, stroke, or CVD death), kidney outcomes (incident ESKD, incident AKI, and eGFR decline ≥30%), and all-cause mortality. We used linear mixed models to examine the association of uDKK3 with annual percentage change in eGFR. Models were adjusted for demographic and clinical characteristics, eGFR, and albuminuria. Results Over a median follow-up of 3.5 years, there were 292 CVD, 73 ESKD, 183 AKI, 471 eGFR decline, and 228 mortality events. In multivariable models without adjustment for eGFR and albuminuria, uDKK3 was strongly associated with CVD, ESKD, AKI, eGFR decline ≥30%, and mortality. However, after further adjustment for eGFR and albuminuria, uDKK3 was no longer associated with risks for composite CVD (hazard ratio, 1.07; 95% confidence interval, 0.92 to 1.23), ESKD (0.80; 0.62 to 1.02), AKI (1.01; 0.85 to 1.21), eGFR decline ≥30% (0.88; 0.79 to 0.99), or mortality (1.02; 0.87 to 1.20). For the linear eGFR change outcome, higher uDKK3 also had no association in the fully adjusted model (−0.03; −0.41 to 0.36). Conclusions Among individuals with hypertension and nondiabetic CKD, higher uDKK3 appeared to have associations with a greater risk of CVD events, incident ESKD, incident AKI, eGFR decline ≥30%, and mortality but these associations were not independent of eGFR and albuminuria.
Collapse
Affiliation(s)
- Vanessa-Giselle Peschard
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Rebecca Scherzer
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Ronit Katz
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Teresa K. Chen
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Alexander L. Bullen
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, California
| | - Kasey Campos
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Michelle M. Estrella
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Michael G. Shlipak
- University of California, San Francisco, California
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center, San Francisco, California
| |
Collapse
|
4
|
Dziamałek-Macioszczyk P, Winiarska A, Pawłowska A, Wojtacha P, Stompór T. Patterns of Dickkopf-3 Serum and Urine Levels at Different Stages of Chronic Kidney Disease. J Clin Med 2023; 12:4705. [PMID: 37510820 PMCID: PMC10380869 DOI: 10.3390/jcm12144705] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/08/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Dickkopf 3 (Dkk3) is a WNT/β-catenin signaling pathway regulator secreted by tubular epithelial cells upon the influence of different stressors. Recently Dkk3 was described as a biomarker of tubular cell injury and a tool that may estimate the risk of chronic kidney disease (CKD) progression. The data about Dkk3 concentrations at particular stages of CKD are lacking. The aim of this study was to measure serum and urine Dkk3 levels in patients with different 'renal status' and evaluate its role as a biomarker of renal damage. One hundred individuals, aged between 24 and 85 years (mean 53.1 ± 17.1), were enrolled in the study. Five groups of 20 subjects each were recruited based on their kidney function. Serum and urine Dkk3 levels were measured by ELISA. The highest median urinary Dkk3 normalized to urinary creatinine was found in patients with established CKD (7051 pg/mg). It was two times higher in renal transplant patients (5705 pg/mg) than in healthy individuals (2654 pg/mg) and the glomerulonephritis group (2470 pg/mg). Urinary Dkk3 was associated with serum creatinine in participants with established CKD and following transplantation. Our results confirm the potential role of Dkk3 as a biomarker of an ongoing renal injury.
Collapse
Affiliation(s)
- Paulina Dziamałek-Macioszczyk
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Agata Winiarska
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Anna Pawłowska
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Paweł Wojtacha
- Department of Industrial and Food Microbiology, University of Warmia and Mazury in Olsztyn, 10-726 Olsztyn, Poland
| | - Tomasz Stompór
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| |
Collapse
|
5
|
Caffo M, Fusco R, Siracusa R, Caruso G, Barresi V, Di Paola R, Cuzzocrea S, Germanò AF, Cardali SM. Molecular Investigation of DKK3 in Cerebral Ischemic/Reperfusion Injury. Biomedicines 2023; 11:biomedicines11030815. [PMID: 36979794 PMCID: PMC10045463 DOI: 10.3390/biomedicines11030815] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Dickkopf-3 (Dkk3) is an atypical member of the Dkk family of Wnt inhibitors, which has been implicated in the pathophysiology of neurodegenerative disorders. Its role in the mechanisms of cellular degeneration and protection is still unknown. The aim of our work is to investigate the endogenous activation of the DKK3 pathway in a model of transient occlusion of the middle cerebral artery in rats. In particular, the animals were subjected to 1 h of ischemia followed by different reperfusion times (1 h, 6 h, 12 h and 24 h) to evaluate the downstream pathway and the time course of its activation. Western blot analysis showed increased Dkk3 expression in animals with the highest time of reperfusion. The increased levels of Dkk3 were accompanied by reduced Wnt3a, Frz1 and PIWI1a expression in the cytosol while FOXM1 and β-catenin decreased in the nucleus. These molecular changes led to an increase in the apoptotic pathway, as showed by the increased expression of Caspase 3 and Bax and the reduced levels of Bcl-2, and to a decrease in neurogenesis, as shown by the decreased expression of Tbr2, Ngn2 and Pax6. In the second part of the study, we decided to employ curcumin, an activator of the Wnt/β-catenin signaling, to investigate its effect on Dkk3. In particular, curcumin was administered 1 and 6 h after ischemia, and animals were sacrificed 24 h later when the expression of Dkk3 was higher. Our data displayed that curcumin administration decreased Dkk3 expression, and increased Wnt3a, Frz1 and PIWI1a levels. Well in line with these data, curcumin administration increased nuclear β-catenin and FOXM1 expression. The down-regulation of Dkk3 by curcumin led to reduced apoptosis and increased neurogenesis. Summarizing, our results showed that Dkk3 acts as an inhibitor of Wnt/β-catenin signaling during cerebral ischemia. Additionally, its inhibition and the contextual activation of the Wnt/β-catenin pathway are protective against ischemic stroke.
Collapse
Affiliation(s)
- Maria Caffo
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, 98166 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, 98166 Messina, Italy
| | - Gerardo Caruso
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Piazzale Ludovico Antonio Scuro, 37124 Verona, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Viale Annunzita, 98168 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, 98166 Messina, Italy
| | - Antonino Francesco Germanò
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Salvatore Massimo Cardali
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
6
|
Urinary Dickkopf-3 (DKK3) Is Associated with Greater eGFR Loss in Patients with Resistant Hypertension. J Clin Med 2023; 12:jcm12031034. [PMID: 36769689 PMCID: PMC9918035 DOI: 10.3390/jcm12031034] [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: 12/11/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Patients with resistant hypertension (HTN) demonstrate an increased risk of chronic kidney disease and progression to end-stage renal disease; however, the individual course of progression is hard to predict. Assessing the stress-induced, urinary glycoprotein Dickkopf-3 (uDKK3) may indicate ongoing renal damage and consecutive estimated glomerular filtration rate (eGFR) decline. The present study aimed to determine the association between uDKK3 levels and further eGFR changes in patients with resistant HTN. In total, 31 patients with resistant HTN were included. Blood pressure and renal function were measured at baseline and up to 24 months after (at months 12 and 24). uDKK3 levels were determined exclusively from the first available spot urine sample at baseline or up to a period of 6 months after, using a commercial ELISA kit. Distinctions between different patient groups were analyzed using the unpaired t-test or Mann-Whitney test. Correlation analysis was performed using Spearman's correlation. The median uDKK3 level was 303 (interquartile range (IQR) 150-865) pg/mg creatinine. Patients were divided into those with high and low eGFR loss (≥3 vs. <3 mL/min/1.73 m²/year). Patients with high eGFR loss showed a significantly higher median baseline uDKK3 level (646 (IQR 249-2555) (n = 13) vs. 180 (IQR 123-365) pg/mg creatinine (n = 18), p = 0.0412 (Mann-Whitney U)). Alternatively, patients could be classified into those with high and low uDKK3 levels (≥400 vs. <400 pg/mg creatinine). Patients with high uDKK3 levels showed significantly higher eGFR loss (-6.4 ± 4.7 (n = 11) vs. 0.0 ± 7.6 mL/min/1.73 m2/year (n = 20), p = 0.0172 (2-sided, independent t-test)). Within the entire cohort, there was a significant correlation between the uDKK3 levels and change in eGFR at the latest follow-up (Spearman's r = -0.3714, p = 0.0397). In patients with resistant HTN, high levels of uDKK3 are associated with higher eGFR loss up to 24 months later.
Collapse
|
7
|
Nephroprotective Role of Chrysophanol in Hypoxia/Reoxygenation-Induced Renal Cell Damage via Apoptosis, ER Stress, and Ferroptosis. Biomedicines 2021; 9:biomedicines9091283. [PMID: 34572468 PMCID: PMC8467645 DOI: 10.3390/biomedicines9091283] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/11/2021] [Accepted: 09/18/2021] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is caused by hypoxia-reoxygenation (H/R), which is a kidney injury produced by a variety of causes, resulting in the remaining portion of the kidney function being unable to maintain the balance for performing the tasks of waste excretion metabolism, and electrolyte and acid-base balance. Many studies have reported the use of Chinese medicine to slow down the progression and alleviate the complications of chronic renal failure. Chrysophanol is a component of Rheum officinale Baill, a traditional Chinese medicine that has been clinically used to treat renal disease. We aimed to study the nephroprotective effect of chrysophanol on hypoxia/ reoxygenation (H/R)-induced cell damage. The results showed that chrysophanol prevented H/R-induced apoptosis via downregulation of cleaved Caspase-3, p-JNK, and Bax but upregulation of Bcl-2 expression. In contrast, chrysophanol attenuated H/R-induced endoplasmic reticulum (ER) stress via the downregulation of CHOP and p-IRE1α expression. Our data demonstrated that chrysophanol alleviated H/R-induced lipid ROS accumulation and ferroptosis. Therefore, we propose that chrysophanol may have a protective effect against AKI by regulating apoptosis, ER stress, and ferroptosis.
Collapse
|
8
|
Obert LA, Elmore SA, Ennulat D, Frazier KS. A Review of Specific Biomarkers of Chronic Renal Injury and Their Potential Application in Nonclinical Safety Assessment Studies. Toxicol Pathol 2021; 49:996-1023. [PMID: 33576319 DOI: 10.1177/0192623320985045] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A host of novel renal biomarkers have been developed over the past few decades which have enhanced monitoring of renal disease and drug-induced kidney injury in both preclinical studies and in humans. Since chronic kidney disease (CKD) and acute kidney injury (AKI) share similar underlying mechanisms and the tubulointerstitial compartment has a functional role in the progression of CKD, urinary biomarkers of AKI may provide predictive information in chronic renal disease. Numerous studies have explored whether the recent AKI biomarkers could improve upon the standard clinical biomarkers, estimated glomerular filtration rate (eGFR), and urinary albumin to creatinine ratio, for predicting outcomes in CKD patients. This review is an introduction to alternative assays that can be utilized in chronic (>3 months duration) nonclinical safety studies to provide information on renal dysfunction and to demonstrate specific situations where these assays could be utilized in nonclinical drug development. Novel biomarkers such as symmetrical dimethyl arginine, dickkopf homolog 3, and cystatin C predict chronic renal injury in animals, act as surrogates for GFR, and may predict changes in GFR in patients over time, ultimately providing a bridge from preclinical to clinical renal monitoring.
Collapse
Affiliation(s)
- Leslie A Obert
- 549350GlaxoSmithKline (GSK), Nonclinical Safety, Collegeville, PA, USA
| | - Susan A Elmore
- Cellular and Molecular Pathology Branch, National Toxicology Program (NTP), 6857National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Daniela Ennulat
- 549350GlaxoSmithKline (GSK), Nonclinical Safety, Collegeville, PA, USA
| | | |
Collapse
|
9
|
Yiu WH, Li Y, Lok SWY, Chan KW, Chan LYY, Leung JCK, Lai KN, Tsu JHL, Chao J, Huang XR, Lan HY, Tang SCW. Protective role of kallistatin in renal fibrosis via modulation of Wnt/β-catenin signaling. Clin Sci (Lond) 2021; 135:429-446. [PMID: 33458750 DOI: 10.1042/cs20201161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/31/2022]
Abstract
Kallistatin is a multiple functional serine protease inhibitor that protects against vascular injury, organ damage and tumor progression. Kallistatin treatment reduces inflammation and fibrosis in the progression of chronic kidney disease (CKD), but the molecular mechanisms underlying this protective process and whether kallistatin plays an endogenous role are incompletely understood. In the present study, we observed that renal kallistatin levels were significantly lower in patients with CKD. It was also positively correlated with estimated glomerular filtration rate (eGFR) and negatively correlated with serum creatinine level. Unilateral ureteral obstruction (UUO) in animals also led to down-regulation of kallistatin protein in the kidney, and depletion of endogenous kallistatin by antibody injection resulted in aggravated renal fibrosis, which was accompanied by enhanced Wnt/β-catenin activation. Conversely, overexpression of kallistatin attenuated renal inflammation, interstitial fibroblast activation and tubular injury in UUO mice. The protective effect of kallistatin was due to the suppression of TGF-β and β-catenin signaling pathways and subsequent inhibition of epithelial-to-mesenchymal transition (EMT) in cultured tubular cells. In addition, kallistatin could inhibit TGF-β-mediated fibroblast activation via modulation of Wnt4/β-catenin signaling pathway. Therefore, endogenous kallistatin protects against renal fibrosis by modulating Wnt/β-catenin-mediated EMT and fibroblast activation. Down-regulation of kallistatin in the progression of renal fibrosis underlies its potential as a valuable clinical biomarker and therapeutic target in CKD.
Collapse
Affiliation(s)
- Wai Han Yiu
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ye Li
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Sarah W Y Lok
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kam Wa Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Loretta Y Y Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Joseph C K Leung
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kar Neng Lai
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - James H L Tsu
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Julie Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Hui Yao Lan
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Sydney C W Tang
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
10
|
Fang X, Hu J, Chen Y, Shen W, Ke B. Dickkopf-3: Current Knowledge in Kidney Diseases. Front Physiol 2020; 11:533344. [PMID: 33391006 PMCID: PMC7772396 DOI: 10.3389/fphys.2020.533344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Dickkopf-related protein 3 (DKK3) is a secreted glycoprotein that has been implicated in the pathogenesis of a variety of diseases. Recent evidence suggests that urinary DKK3 may serve as a potential biomarker for monitoring kidney disease progression and assessing the effects of interventions. We review the biological role of DKK3 as an agonist in chronic kidney disease (CKD) and autosomal dominant polycystic kidney disease (ADPKD) and as an antagonist in idiopathic membranous nephropathy (IMN). In addition, we present the clinical applications of DKK3 in acute kidney disease and tubulointerstitial fibrosis, suggesting that urine DKK3 may be a potential biomarker for acute kidney disease and CKD. Further research into the mechanism of DKK3 and its use as a diagnostic tool, alone or in combination with other biomarkers, could prove clinically useful for better understanding the pathology of kidney diseases and improving early detection and treatment.
Collapse
Affiliation(s)
- Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Hu
- The Third Hospital of Nanchang, Nanchang, China
| | - Yanxia Chen
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Shen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ben Ke
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
11
|
WNT-β-catenin signalling - a versatile player in kidney injury and repair. Nat Rev Nephrol 2020; 17:172-184. [PMID: 32989282 DOI: 10.1038/s41581-020-00343-w] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
Abstract
The WNT-β-catenin system is an evolutionary conserved signalling pathway that is of particular importance for morphogenesis and cell organization during embryogenesis. The system is usually suppressed in adulthood; however, it can be re-activated in organ injury and regeneration. WNT-deficient mice display severe kidney defects at birth. Transient WNT-β-catenin activation stimulates tissue regeneration after acute kidney injury, whereas sustained (uncontrolled) WNT-β-catenin signalling promotes kidney fibrosis in chronic kidney disease (CKD), podocyte injury and proteinuria, persistent tissue damage during acute kidney injury and cystic kidney diseases. Additionally, WNT-β-catenin signalling is involved in CKD-associated vascular calcification and mineral bone disease. The WNT-β-catenin pathway is tightly regulated, for example, by proteins of the Dickkopf (DKK) family. In particular, DKK3 is released by 'stressed' tubular epithelial cells; DKK3 drives kidney fibrosis and is associated with short-term risk of CKD progression and acute kidney injury. Thus, targeting the WNT-β-catenin pathway might represent a promising therapeutic strategy in kidney injury and associated complications.
Collapse
|
12
|
Zhang J, Jiang T, Liang X, Shu S, Xiang X, Zhang W, Guo T, Xie W, Deng W, Tang X. lncRNA MALAT1 mediated high glucose-induced HK-2 cell epithelial-to-mesenchymal transition and injury. J Physiol Biochem 2019; 75:443-452. [PMID: 31388927 DOI: 10.1007/s13105-019-00688-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/26/2019] [Indexed: 01/10/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) and injury of tubular cells play critical roles in the pathogenesis of diabetic nephropathy (DN). lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been shown to be involved in DN progression. However, whether MALAT1 induces EMT and injury in tubular cells is unclear. Here, we investigated the effects of MALAT1 on human proximal tubular cells (HK-2 cells) and the underlying mechanism. We performed qPCR to detect MALAT1, E-cadherin, α-smooth muscle actin (α-SMA), kidney injury molecule 1 (KIM-1), and neutrophil gelatinase-associated lipocalin (NGAL). Additionally, we conducted Western blot analyses to measure E-cadherin, α-SMA, cyclin D1, c-Myc, and β-catenin in HK-2 cells cultured with normal glucose and high glucose (HG) and in transfected cells or cells treated with LiCl and DKK-1. The β-catenin localization was observed using immunofluorescence, and the protein levels of NGAL and KIM-1 were evaluated by ELISA. We found that HG-upregulated MALAT1 decreased E-cadherin and increased α-SMA, KIM-1, NGAL, cyclin D1, c-Myc, and β-catenin in HK-2 cells. LiCl exposure increased the expression of α-SMA but decreased that of E-cadherin on the base of knocking down MALAT1, and decreased NGAL and KIM-1 expression. DKK-1 showed the opposite effects. Our results suggested that upregulated MALAT1 induced EMT in HG-treated HK-2 cells through activating the Wnt/β-catenin pathway. However, MALAT1-mediated injury in HK-2 cells was not mediated by activation of the Wnt/β-catenin pathway. Our results indicate that MALAT1 might serve as a novel therapeutic target for suppressing the progression of DN.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Guangzhou, 510282, Guangdong, People's Republic of China
| | - Tingting Jiang
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Guangzhou, 510282, Guangdong, People's Republic of China
- Department of Nephrology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Province, 530021, China
| | - Xiujie Liang
- Department of Medicine/Physiology, University of Fribourg, Chemin du Musée 5, 1700, Fribourg, Switzerland
| | - Shuangshuang Shu
- Department of Nephrology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiaohong Xiang
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Wenying Zhang
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Guangzhou, 510282, Guangdong, People's Republic of China
| | - Tingting Guo
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Guangzhou, 510282, Guangdong, People's Republic of China
| | - Wei Xie
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Guangzhou, 510282, Guangdong, People's Republic of China
| | - Weiqian Deng
- Department of Nephrology, Fifth Affiliated Hospital of Southern Medical University, 566 Congcheng Road, Conghua District, Guangzhou, 510900, Guangdong, People's Republic of China.
| | - Xun Tang
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Guangzhou, 510282, Guangdong, People's Republic of China.
| |
Collapse
|
13
|
Tang Y, Shen J, Zhang F, Yang FY, Liu M. Human serum albumin attenuates global cerebral ischemia/reperfusion-induced brain injury in a Wnt/β-Catenin/ROS signaling-dependent manner in rats. Biomed Pharmacother 2019; 115:108871. [PMID: 31026729 DOI: 10.1016/j.biopha.2019.108871] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/06/2019] [Accepted: 04/09/2019] [Indexed: 11/26/2022] Open
Abstract
This study sought to clarify the role and underlying mechanisms of human serum albumin (HSA) therapy in global cerebral ischemia/reperfusion (GCI/R)-induced brain damage in rats. Five groups of adult male Wistar rats (n = 12 per group) were created as follows: sham operation (Sham), global cerebral ischemia/reperfusion (GCI/R), HSA treatment (GCI/R + HSA), Dickkopf-1 (DDK1) treatment (GCI/R + DDK1), and DDK1 plus HSA treatment (GCI/R + DKK1 + HSA). The GCI/R injury model was created using the modified Pusinelli four-vessel occlusion method. After 24 h, rats were evaluated using neurological scoring, Nissl staining, and brain tissue water content. The mRNA expression of Wnt, GSK3β, and β-Catenin in the brain were detected by quantitative real time polymerase chain reaction. The protein expression of β-Catenin and GSK-3β were investigated by western blot and immunohistochemical analysis in the presence and absence of the Wnt/β-Catenin antagonist, DKK-1. Complex I activity and ROS content were also measured. After 24 h of reperfusion, the behavior score and brain tissue water content in the GCI/R + HSA group were lower than that in the GCI/R group. In addition, the degree of neuronal injury was significantly reduced in the GCI/R + HSA group (P < 0.05). The ROS content was significantly decreased and Complex I activity was markedly raised in the GCI/R + HSA group compared to the GCI/R group (P < 0.05). Further, GSK-3β expression in the GCI/R + HSA group was lower than that in the GCI/R group, while the Wnt and β-catenin expression were increased. These effects were reversed by DKK1. Taken together, we showed that HSA attenuates GCI/R-induced brain damage and may be neuroprotective via regulation of the Wnt/β-catenin/ROS signaling pathway.
Collapse
Affiliation(s)
- Yuedong Tang
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China.
| | - Feng Zhang
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China
| | - Fei-Yu Yang
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China
| | - Ming Liu
- Department of Respiration, Shanghai Punan Hospital, Shanghai, China
| |
Collapse
|
14
|
Busceti CL, Di Menna L, Bianchi F, Mastroiacovo F, Di Pietro P, Traficante A, Bozza G, Niehrs C, Battaglia G, Bruno V, Fornai F, Volpe M, Rubattu S, Nicoletti F. Dickkopf-3 Causes Neuroprotection by Inducing Vascular Endothelial Growth Factor. Front Cell Neurosci 2018; 12:292. [PMID: 30258353 PMCID: PMC6143799 DOI: 10.3389/fncel.2018.00292] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022] Open
Abstract
Dickkopf-3 (Dkk3) is an atypical member of the Dkk family of Wnt inhibitors, which has been implicated in the pathophysiology of neurodegenerative disorders. However, the role of Dkk3 in mechanisms of cell degeneration and protection is unknown. We used Dkk3 knockout mice to examine how endogenous Dkk3 influences ischemic brain damage. In addition, we used primary cultures of astrocytes or mixed cultures of astrocytes and neurons to investigate the action of Dkk3 on cell damage and dissect the underlying molecular mechanisms. In a model of focal brain ischemia induced by permanent middle cerebral artery (MCA) occlusion (MCAO) Dkk3−/− mice showed a significantly greater infarct size with respect to their wild-type counterparts at all time points investigated (1, 3 and 7 days after MCAO). Immunohistochemical analysis showed that Dkk3 expression was enhanced at the borders of the ischemic focus, and was predominantly detected in astrocytes. This raised the possibility that Dkk3 produced by astrocytes acted as a protective molecule. We tested this hypothesis using either primary cultures of cortical astrocytes or mixed cortical cultures containing both neurons and astrocytes. Genetic deletion of Dkk3 was permissive to astrocyte damage induced by either oxidative stress or glucose deprivation. In addition, application of human recombinant Dkk3 (hrDkk3) was highly protective against oxidative stress in cultured astrocytes. We tested the hypothesis that the protective activity of Dkk3 was mediated byvascular endothelial growth factor (VEGF). Interestingly, glucose deprivation up-regulated both Dkk3 and VEGF in cultured astrocytes prepared from wild-type mice. VEGF induction was not observed in astrocytes lacking Dkk3 (i.e., in cultures prepared from Dkk3−/− mice). In mixed cultures of cortical cells, excitotoxic neuronal death induced by a brief pulse with N-methyl-D-aspartate (NMDA) was significantly enhanced when Dkk3 was lacking in astrocytes, whereas post-NMDA addition of hrDkk3 was neuroprotective. Neuroprotection by hrDkk3 was significantly reduced by pharmacological blockade of type-2 VEGF receptors and was mimicked by hrVEGF. These data offer the first evidence that Dkk3 protects both neurons and astrocytes against a variety of toxic insults, and at least in culture, protection involves VEGF induction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Allianz, German Cancer Research Center, Heidelberg, Germany.,Institute of Molecular Biology (IMB), Mainz, Germany
| | | | - Valeria Bruno
- IRCCS Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, University Sapienza, Rome, Italy
| | - Francesco Fornai
- IRCCS Neuromed, Pozzilli, Italy.,Department of Human Morphology and Applied Biology, University of Pisa, Pisa, Italy
| | - Massimo Volpe
- IRCCS Neuromed, Pozzilli, Italy.,Clinical and Molecular Medicine, University Sapienza, Rome, Italy
| | - Speranza Rubattu
- IRCCS Neuromed, Pozzilli, Italy.,Clinical and Molecular Medicine, University Sapienza, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, University Sapienza, Rome, Italy
| |
Collapse
|
15
|
Zhu X, Li W, Li H. miR-214 ameliorates acute kidney injury via targeting DKK3 and activating of Wnt/β-catenin signaling pathway. Biol Res 2018; 51:31. [PMID: 30180910 PMCID: PMC6122444 DOI: 10.1186/s40659-018-0179-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
Background miR-214 was demonstrated to be upregulated in models of renal disease and promoted fibrosis in renal injury independent of TGF-β signaling in vivo. However, the detailed role of miR-214 in acute kidney injury (AKI) and its underlying mechanism are still largely unknown. Methods In this study, an I/R-induced rat AKI model and a hypoxia-induced NRK-52E cell model were used to study AKI. The concentrations of kidney injury markers serum creatinine, blood urea nitrogen, and kidney injury molecule-1 were measured. The expressions of miR-214, tumor necrosis factor-α, interleukin (IL)-1β, IL-6, were detected by RT-qPCR. The protein levels of Bcl-2, Bax, Dickkopf-related protein 3, β-catenin, c-myc, and cyclinD1 were determined by western blot. Cell apoptosis and caspase 3 activity were evaluated by flow cytometry analysis and caspase 3 activity assay, respectively. Luciferase reporter assay was used to confirm the interaction between miR-214 and Dkk3. Results miR-214 expression was induced in ischemia–reperfusion (I/R)-induced AKI rat and hypoxic incubation of NRK-52E cells. Overexpression of miR-214 alleviated hypoxia-induced NRK-52E cell apoptosis while inhibition of miR-214 expression exerted the opposite effect. Dkk3 was identified as a target of miR-214. Anti-miR-214 abolished the inhibitory effects of DKK3 knockdown on hypoxia-induced NRK-52E cell apoptosis by inactivation of Wnt/β-catenin signaling. Moreover, miR-214 ameliorated AKI in vivo by inhibiting apoptosis and fibrosis through targeting Dkk3 and activating Wnt/β-catenin pathway. Conclusion miR-214 ameliorates AKI by inhibiting apoptosis through targeting Dkk3 and activating Wnt/β-catenin signaling pathway, offering the possibility of miR-214 in the therapy of ischemic AKI. Electronic supplementary material The online version of this article (10.1186/s40659-018-0179-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoguang Zhu
- Department of Nephrology, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng, 475000, China.
| | - Wenwen Li
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Huicong Li
- Department of Nephrology, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng, 475000, China
| |
Collapse
|
16
|
Activated renal tubular Wnt/β-catenin signaling triggers renal inflammation during overload proteinuria. Kidney Int 2018; 93:1367-1383. [PMID: 29605095 DOI: 10.1016/j.kint.2017.12.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 12/01/2017] [Accepted: 12/21/2017] [Indexed: 01/27/2023]
Abstract
Imbalance of Wnt/β-catenin signaling in renal cells is associated with renal dysfunction, yet the precise mechanism is poorly understood. Previously we observed activated Wnt/β-catenin signaling in renal tubules during proteinuric nephropathy with an unknown net effect. Therefore, to identify the definitive role of tubular Wnt/β-catenin, we generated a novel transgenic "Tubcat" mouse conditionally expressing stabilized β-catenin specifically in renal tubules following tamoxifen administration. Four weeks after tamoxifen injection, uninephrectomized Tubcat mice displayed proteinuria and elevated blood urea nitrogen levels compared to non-transgenic mice, implying a detrimental effect of the activated signaling. This was associated with infiltration of the tubulointerstitium predominantly by M1 macrophages and overexpression of the inflammatory chemocytokines CCL-2 and RANTES. Induction of overload proteinuria by intraperitoneal injection of low-endotoxin bovine serum albumin following uninephrectomy for four weeks aggravated proteinuria and increased blood urea nitrogen levels to a significantly greater extent in Tubcat mice. Renal dysfunction correlated with the degree of M1 macrophage infiltration in the tubulointerstitium and renal cortical up-regulation of CCL-2, IL-17A, IL-1β, CXCL1, and ICAM-1. There was overexpression of cortical TLR-4 and NLRP-3 in Tubcat mice, independent of bovine serum albumin injection. Finally, there was no fibrosis, activation of epithelial-mesenchymal transition or non-canonical Wnt pathways observed in the kidneys of Tubcat mice. Thus, conditional activation of renal tubular Wnt/β-catenin signaling in a novel transgenic mouse model demonstrates that this pathway enhances intrarenal inflammation via the TLR-4/NLRP-3 inflammasome axis in overload proteinuria.
Collapse
|
17
|
Chen D, Xiong XQ, Zang YH, Tong Y, Zhou B, Chen Q, Li YH, Gao XY, Kang YM, Zhu GQ. BCL6 attenuates renal inflammation via negative regulation of NLRP3 transcription. Cell Death Dis 2017; 8:e3156. [PMID: 29072703 PMCID: PMC5680929 DOI: 10.1038/cddis.2017.567] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022]
Abstract
Renal inflammation contributes to the pathogeneses of hypertension. This study was designed to determine whether B-cell lymphoma 6 (BCL6) attenuates renal NLRP3 inflammasome activation and inflammation and its underlying mechanism. Male spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY) were used in the present study. Angiotensin (Ang) II or lipopolysaccharides (LPS) was used to induce inflammation in HK-2 cells, a human renal tubular epithelial (RTE) cell line. NLRP3 inflammasome was activated and BCL6 was downregulated in the kidneys of SHR. Either Ang II or LPS suppressed BCL6 expression in HK-2 cells. BCL6 overexpression in HK-2 cells attenuated Ang II-induced NLRP3 upregulation, inflammation and cell injury. The inhibitory effects of BCL6 overexpression on NLRP3 expression and inflammation were also observed in LPS-treated HK-2 cells. BCL6 inhibited the NLRP3 transcription via binding to the NLRP3 promoter. BCL6 knockdown with shRNA increased NLRP3 and mature IL-1β expression levels in both PBS- or Ang II-treated HK-2 cells but had no significant effects on ASC, pro-caspase-1 and pro-IL-1β expression levels. BCL6 overexpression caused by recombinant lentivirus expressing BCL6 reduced blood pressure in SHR. BCL6 overexpression prevented the upregulation of NLRP3 and mature IL-1β expression levels in the renal cortex of SHR. The results indicate that BCL6 attenuates Ang II- or LPS-induced inflammation in HK-2 cells via negative regulation of NLRP3 transcription. BCL6 overexpression in SHR reduced blood pressure, NLRP3 expression and inflammation in the renal cortex of SHR.
Collapse
Affiliation(s)
- Dan Chen
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiao-Qing Xiong
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying-Hao Zang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying Tong
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bing Zhou
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xing-Ya Gao
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|