1
|
Machado PAB, Lass A, Pilger BI, Fornazari R, de Moraes TP, Pinho RA. SGLT2 inhibitors and NLRP3 inflammasome: potential target in diabetic kidney disease. J Bras Nefrol 2024; 46:e20230187. [PMID: 39412512 PMCID: PMC11539899 DOI: 10.1590/2175-8239-jbn-2023-0187en] [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: 12/11/2023] [Accepted: 05/28/2024] [Indexed: 11/08/2024] Open
Abstract
Diabetic kidney disease (DKD) remains the leading cause of chronic kidney disease (CKD) worldwide. The pathogenesis of DKD is influenced by functional, histopathological, and immune mechanisms, including NLRP3 inflammasome activity and oxidative stress. The sodium-glucose cotransporter 2 inhibitors (SGLT2i) have shown metabolic benefits and the ability to slow the progression of DKD in several clinical studies over the years. Recent studies suggest that the antidiabetic activity also extends to inhibition of the inflammatory response, including modulation of the NLRP3 inflammasome, reduction of pro-inflammatory markers and reduction of oxidative stress. Here we review the efficacy of SGLT2i in the treatment of CKD and discuss the role of the inflammatory response in the development of DKD, including its relationship to the NLRP3 inflammasome and oxidative stress.
Collapse
Affiliation(s)
- Paulo André Bispo Machado
- Pontifícia Universidade Católica do Paraná, Laboratório de Bioquímica do Exercício em Saúde, Curitiba, PR, Brazil
- Pontificia Universidade Católica do Paraná, Pós-graduação em Ciências da Saúde, Curitiba, PR, Brazil
| | - André Lass
- Pontifícia Universidade Católica do Paraná, Laboratório de Bioquímica do Exercício em Saúde, Curitiba, PR, Brazil
- Pontificia Universidade Católica do Paraná, Pós-graduação em Ciências da Saúde, Curitiba, PR, Brazil
| | - Bruna Isadora Pilger
- Pontifícia Universidade Católica do Paraná, Laboratório de Bioquímica do Exercício em Saúde, Curitiba, PR, Brazil
- Pontificia Universidade Católica do Paraná, Pós-graduação em Ciências da Saúde, Curitiba, PR, Brazil
| | - Raphaella Fornazari
- Pontifícia Universidade Católica do Paraná, Laboratório de Bioquímica do Exercício em Saúde, Curitiba, PR, Brazil
- Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil
| | - Thyago Proença de Moraes
- Pontifícia Universidade Católica do Paraná, Laboratório de Bioquímica do Exercício em Saúde, Curitiba, PR, Brazil
- Pontificia Universidade Católica do Paraná, Pós-graduação em Ciências da Saúde, Curitiba, PR, Brazil
| | - Ricardo Aurino Pinho
- Pontifícia Universidade Católica do Paraná, Laboratório de Bioquímica do Exercício em Saúde, Curitiba, PR, Brazil
- Pontificia Universidade Católica do Paraná, Pós-graduação em Ciências da Saúde, Curitiba, PR, Brazil
| |
Collapse
|
2
|
Wang F, Huang X, Wang S, Wu D, Zhang M, Wei W. The main molecular mechanisms of ferroptosis and its role in chronic kidney disease. Cell Signal 2024; 121:111256. [PMID: 38878804 DOI: 10.1016/j.cellsig.2024.111256] [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: 03/28/2024] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
The term ferroptosis, coined in 2012, has been widely applied in various disease research fields. Ferroptosis is a newly regulated form of cell death distinct from apoptosis, necrosis, and autophagy, the mechanisms of which have been extensively studied. Chronic kidney disease, characterized by renal dysfunction, is a common disease severely affecting human health, with its occurrence and development influenced by multiple factors and leading to dysfunction in multiple systems. It often lacks obvious clinical symptoms in the early stages, and thus, diagnosis is typically made in the later stages, complicating treatment. While research on ferroptosis and acute kidney injury has made continuous progress, studies on the association between ferroptosis and chronic kidney disease remain limited. This review aims to summarize chronic kidney disease, investigate the mechanism and regulation of ferroptosis, and attempt to elucidate the role of ferroptosis in the occurrence and development of chronic kidney disease.
Collapse
Affiliation(s)
- Fulin Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xuesong Huang
- Department of Urology, Jilin People's Hospital, Jilin, China
| | - Shaokun Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Dawei Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | | | - Wei Wei
- Department of Urology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
3
|
Ullah S, Rahman W, Ullah F, Ullah A, Jehan R, Iqbal MN, Ali I, Tianshun G. Identification of lead compound screened from the natural products atlas to treat renal inflammasomes using molecular docking and dynamics simulation. J Biomol Struct Dyn 2024; 42:4851-4861. [PMID: 37705295 DOI: 10.1080/07391102.2023.2254397] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/04/2023] [Indexed: 09/15/2023]
Abstract
One of the most prevalent ailments is kidney disease. Effective therapies for chronic renal disease are hard to come by. As a result, there is significant clinical and social interest to predict and develop novel compounds to treat renal disorders. So, specific natural products have been employed in this study because they have protective effects against kidney diseases. When taken orally, natural products can help protect against or lessen the severity of the kidney damage caused by high fructose intake, a high-fat diet, and both Type I and Type 2 diabetes. Reduced podocyte injury, a contributor to albuminuria in diabetic nephropathy, reduces renal endothelial barrier function disruption due to hyperglycemia, as well as urinary microalbumin excretion and glomerular hyperfiltration. Multiple natural products have been shown to protect the kidneys from nephrotoxic chemicals such as LPS, gentamycin, alcohol, nicotine, lead, and cadmium, all of which can persuade acute kidney injury (AKI) or chronic kidney disease (CKD). Natural compounds inhibit regulatory enzymes for controlling inflammation-related diseases. For this, use computational methods such as drug design to identify novel flavonoid compounds against kidney diseases. Drug design via computational methods gaining admiration as a swift and effective technique to identify lead compounds in a shorter time at a low cost. In this in-silico study, we screened The Natural Product Atlas based on a structure-based pharmacophore query. Top hits were analyzed for ADMET analysis followed by molecular docking and docking validation. Finally, the lead compound was simulated for a period of 200 ns and trajectories were studied for stability. We found that NPA024823 showed promising binding and stability with the AIM2. This research work aims to predict novel anti-inflammatory compounds against kidney diseases to inhibit kidney inflammasome by targeting the AIM2 protein. So, in initial preclinical research, there will be lower failure rates that demonstrate safety profiles against predicted compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | | | - Anees Ullah
- S Khan Lab Mardan, Khyber Pakhtunkhwa, Pakistan
| | | | - Muhammad Nasir Iqbal
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology, and Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Iqra Ali
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Gao Tianshun
- Big Data Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
4
|
Lin J, Zhu C, Cui F, Qu H, Zhang Y, Le X, Yin J, Cao Y. Based on functional and histopathological correlations: is diffusion kurtosis imaging valuable for noninvasive assessment of renal damage in early-stage of chronic kidney disease? Int Urol Nephrol 2024; 56:263-273. [PMID: 37326823 DOI: 10.1007/s11255-023-03632-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/10/2023] [Indexed: 06/17/2023]
Abstract
PURPOSE To evaluate the potential of 3 T magnetic resonance diffusion kurtosis imaging (DKI) in assessing the renal damage in early-stage of chronic kidney disease (CKD) patients with normal or slightly changed functional index, using histopathology as reference standard. METHODS 49 CKD patients and 18 healthy volunteers were recruited in this study. CKD patients were divided into two groups based on estimated glomerular filtration rate (eGFR): Study group I (eGFR ≥ 90 ml/min/1.73 m2 [n = 20]) and Study group II (eGFR < 90 ml/min/1.73 m2 [n = 29]). DKI was performed in all participants. The DKI parameters (mean kurtosis [MK], mean diffusivity [MD], fractional anisotropy [FA]) of renal cortex and medulla were measured. The differences of parenchymal MD, MK and FA values among the different groups were compared. The correlations between DKI parameters and clinicopathological characteristics were assessed. Diagnostic performance of DKI to assess renal damage in early-stage of CKD was analyzed. RESULTS The cortex MD and MK showed significant difference among three groups (P < 0.05): trend of cortex MD: Study group II < Study group I < control group; trend of cortex MK: control group < Study group I < Study group II. The cortex MD and MK and medulla FA were correlated with eGFR and Interstitial fibrosis/Tubular atrophy score (0.3 < r < 0.5). Cortex MD and MK yielded an AUC of 0.752 for differentiating healthy volunteers from CKD patients with eGFR ≥ 90 ml/min/1.73 m2. CONCLUSION DKI shows potential in non-invasive and multi-parameter quantitative assessment of renal damage in early-stage of CKD patients and provide additional information for changes in renal function and histopathology.
Collapse
Affiliation(s)
- Jiazhen Lin
- Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China
| | - Caifeng Zhu
- Department of Nephrology, Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China
| | - Feng Cui
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China
| | - Hua Qu
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China
| | - Yongsheng Zhang
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China
| | - Xianjie Le
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China
| | - Jiazhen Yin
- Department of Nephrology, Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China.
| | - Youjun Cao
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Number 453, Road Stadium, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
5
|
Xu Y, Chen Q, Jiang Y, Liang X, Wang T, Xu Y. UMI-77 Modulates the Complement Cascade Pathway and Inhibits Inflammatory Factor Storm in Sepsis Based on TMT Proteomics and Inflammation Array Glass Chip. J Proteome Res 2023; 22:3464-3474. [PMID: 37830896 DOI: 10.1021/acs.jproteome.3c00317] [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: 10/14/2023]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by infection, which has no specific drug at present. UMI-77 can significantly improve the survival rate of septic mice; the detailed role of UMI-77 and its underlying mechanisms in sepsis are not clear. Inflammation array glass chip and proteomic analyses were performed to elucidate the latent mechanism of UMI-77 in the treatment of sepsis. The results showed that 7.0 mg/kg UMI-77 improved the 5 day survival rate in septic mice compared to the LPS group (60.964 vs 9.779%) and ameliorated the pathological conditions. Inflammation array glass chip analysis showed that sepsis treatment with UMI-77 may eventually through the suppression of the characteristic inflammatory storm-related cytokines such as KC, RANTES, LIX, IL-6, eotaxin, TARC, IL-1β, and so on. Proteomics analysis showed that 213 differential expression proteins and complement and coagulation cascades were significantly associated with the process for the UMI-77 treatment of sepsis. The top 10 proteins including Apoa2, Tgfb1, Serpinc1, Vtn, Apoa4, Cat, Hp, Serpinf2, Fgb, and Serpine1 were identified and verified, which play important roles in the mechanism of UMI-77 in the treatment of sepsis. Our findings indicate that UMI-77 exerts an antisepsis effect by modulating the complement cascade pathway and inhibiting inflammatory storm factors.
Collapse
Affiliation(s)
- Yubin Xu
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou 317000, Zhejiang, China
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang, China
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang, China
| | - Yongpo Jiang
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou 317000, Zhejiang, China
| | - Xi Liang
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang, China
| | - Ting Wang
- School of Medicine, Taizhou University, Taizhou 318000, Zhejiang, China
| | - Yinhe Xu
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou 317000, Zhejiang, China
- Taizhou Key Laboratory of Critical Care Medicine, Taizhou 318000, Zhejiang, China
| |
Collapse
|
6
|
Kadatane SP, Satariano M, Massey M, Mongan K, Raina R. The Role of Inflammation in CKD. Cells 2023; 12:1581. [PMID: 37371050 DOI: 10.3390/cells12121581] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic kidney disease (CKD) affects many adults worldwide. Persistent low-grade inflammation is a substantial factor in its development and progression and has correlated with increased mortality and cardiovascular problems. This low-grade inflammation is a product of dysregulation of the normal balance between pro- and anti-inflammatory markers. Various factors such as increased innate immune system activation, reactive oxygen species production, periodontal disease, dysregulation of anti-inflammatory systems and intestinal dysbiosis result in the dysregulation of this balance. Furthermore, this low-grade inflammation has down-effects such as hypertension, renal fibrosis and acceleration of renal function decline. Moreover, low-grade inflammation over time has been linked to malignancy in CKD. As CKD progresses, many patients require dialysis, which has a negative bidirectional relationship with persistent inflammation. Treatment options for inflammation in CKD are vast, including cytokine inhibitors, statins and diets. However, more research is needed to create a standardized management plan. In this review, we will examine the normal physiology of the kidney and its relationship with the immune system. We will then delve into the pathology behind persistent inflammation, the various causes of inflammation, the downstream effects of inflammation, dialysis and potential treatments for inflammation in CKD.
Collapse
Affiliation(s)
| | - Matthew Satariano
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Michael Massey
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Kai Mongan
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Rupesh Raina
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH 44302, USA
- Department of Nephrology, Akron Children's Hospital, Akron, OH 44308, USA
| |
Collapse
|
7
|
Huang Z, Vlasschaert C, Robinson-Cohen C, Pan Y, Sun X, Lash JP, Kestenbaum B, Kelly TN. Emerging evidence on the role of clonal hematopoiesis of indeterminate potential in chronic kidney disease. Transl Res 2023; 256:87-94. [PMID: 36586535 PMCID: PMC10101890 DOI: 10.1016/j.trsl.2022.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Chronic kidney disease (CKD) was responsible for 1.2 million deaths globally in 2016. Despite the large and growing burden of CKD, treatment options are limited and generally only preserve kidney function. Characterizing molecular precursors to incident and progressive CKD could point to critically needed prevention and treatment strategies. Clonal hematopoiesis of indeterminate potential (CHIP) is typically characterized by the clonal expansion of blood cells carrying somatic mutations in specific driver genes. An age-related disorder, CHIP is rare in the young but common in older adults. Recent studies have identified causal associations between CHIP and atherosclerotic cardiovascular disease which are most likely mediated by inflammation, a hallmark of CKD. Animal evidence has supported causal effects of CHIP on kidney injury, inflammation, and fibrosis, providing impetus for human research. Although prospective epidemiologic studies investigating associations of CHIP with development and progression of CKD are few, intriguing findings have been reported. CHIP was significantly associated with kidney function decline and end stage kidney disease in the general population, although effect sizes were modest. Recent work suggests larger associations of CHIP with kidney disease progression in CKD patients, but further investigations in this area are needed. In addition, the accumulating literature has identified some heterogeneity in associations between CHIP and kidney endpoints across study populations, but reasons for these differences remain unclear. The current review provides an in-depth exploration into this nascent area of research, develops a conceptual framework linking CHIP to CKD, and discusses the clinical and public health implications of this work.
Collapse
Affiliation(s)
- Zhijie Huang
- Department of Epidemiology, Tulane University, New Orleans, Louisiana
| | | | - Cassianne Robinson-Cohen
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yang Pan
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Xiao Sun
- Department of Epidemiology, Tulane University, New Orleans, Louisiana; Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - James P Lash
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Bryan Kestenbaum
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, Washington
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
8
|
Evaluating the renal mild tubulointerstitial damage and renal function in IgAN patients: a comparative study based on diffusion kurtosis imaging and diffusion tensor imaging. ABDOMINAL RADIOLOGY (NEW YORK) 2023; 48:1350-1362. [PMID: 36749369 DOI: 10.1007/s00261-023-03822-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To compare the performance of 3.0 T magnetic resonance diffusion kurtosis imaging (DKI) and diffusion tensor imaging (DTI) in evaluation of the degree of tubulointerstitial damage and renal function in Immunoglobulin A Nephropathy (IgAN) patients. METHODS Both DKI and DTI were performed in 40 IgAN patients and 17 healthy volunteers. IgAN patients were divided into two groups according to tubulointerstitial lesion score: Mild injury group, n = 24; Moderate-severe injury group, n = 16. DKI characteristic parameters [mean kurtosis (MK), axial kurtosis (Ka), radial kurtosis (Kr)] and DTI parameters [fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (Da), radial diffusivity (Dr)] of renal cortex and medulla were measured and compared among different groups. Correlations between DKI, DTI parameters and clinicopathological characteristics were assessed. Diagnostic performance of DKI and DTI to evaluate tubulointerstitial damage of IgAN was compared. RESULTS Cortical MK, Kr, Da and parenchymal Ka significantly differed among three groups (P < 0.05). Cortical MK, Kr, Ka were negatively correlated with estimated glomerular filtration rate (eGFR) (MK: r = - 0.613; Kr: r = - 0.539; Ka: r = - 0.664) and positively correlated with tubulointerstitial lesion score (MK: r = 0.655; Kr: r = 0.577; Ka: r = 0.661) (all P < 0.001). Lower correlation coefficient was found among cortical FA, MD, Dr and eGFR, tubulointerstitial lesion score (all|r|< 0.350). The AUCs of DKI and DTI parameters for differentiating Mild injury group from control group were (cortical MK 0.822, cortical Ka 0.816; cortical FA 0.515, cortical MD 0.714) and for differentiating Mild injury group from Moderate-severe injury group were (cortical MK 0.813, cortical Ka 0.831; medulla FA 0.784, medulla MD 0.586). CONCLUSION Compared with DTI, DKI was more sensitive and accurate to probe the renal function and the tubulointerstitial damage of IgAN, especially the mild tubulointerstitial damage.
Collapse
|
9
|
Identification of the Subtypes of Renal Ischemia-Reperfusion Injury Based on Pyroptosis-Related Genes. Biomolecules 2023; 13:biom13020275. [PMID: 36830644 PMCID: PMC9952921 DOI: 10.3390/biom13020275] [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: 10/23/2022] [Revised: 12/29/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) often occurs in the process of kidney transplantation, which significantly impacts the subsequent treatment and prognosis of patients. The prognosis of patients with different subtypes of IRI is quite different. Therefore, in this paper, the gene expression data of multiple IRI samples were downloaded from the GEO database, and a double Laplacian orthogonal non-negative matrix factorization (DL-ONMF) algorithm was proposed to classify them. In this algorithm, various regularization constraints are added based on the non-negative matrix factorization algorithm, and the prior information is fused into the algorithm from different perspectives. The connectivity information between different samples and features is added to the algorithm by Laplacian regularization constraints on samples and features. In addition, orthogonality constraints on the basis matrix and coefficient matrix obtained by the algorithm decomposition are added to reduce the influence of redundant samples and redundant features on the results. Based on the DL-ONMF algorithm for clustering, two PRGs-related IRI isoforms were obtained in this paper. The results of immunoassays showed that the immune microenvironment was different among PRGS-related IRI types. Based on the differentially expressed PRGs between subtypes, we used LASSO and SVM-RFE algorithms to construct a diagnostic model related to renal transplantation. ROC analysis showed that the diagnostic model could predict the outcome of renal transplant patients with high accuracy. In conclusion, this paper presents an algorithm, DL-ONMF, which can identify subtypes with different disease characteristics. Comprehensive bioinformatic analysis showed that pyroptosis might affect the outcome of kidney transplantation by participating in the immune response of IRI.
Collapse
|
10
|
Piperine mitigates aortic vasculopathy in streptozotocin-diabetic rats via targeting TXNIP-NLRP3 signaling. Life Sci 2023; 314:121275. [PMID: 36496033 DOI: 10.1016/j.lfs.2022.121275] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/27/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Several in vivo and in vitro studies reported a favorable effect of piperine (PIP) on vascular function. However, the potential impacts of PIP on macrovasculopathy in streptozotocin (STZ)-diabetic rats have not yet been studied. Thirty-two Sprague Dawley rats were used (n= 8/group). STZ-administered rats (50 mg/kg once, i.p) received PIP (30 mg/kg/day, orally) or its vehicle starting from day 15 till the end of the study (10 weeks). Control groups consisted of age-matched normal rats with or without PIP treatment. Metabolic and oxidative stress parameters were biochemically determined. Aortas were histologically examined. Ex vivo aortic reactivity to phenylephrine and acetylcholine was studied. Components of the TXNIP-NLRP3 pathway were assessed using real-time PCR, ELISA, and immunohistochemistry. Two-way ANOVA was used to compare groups. Statistical significance was set at P < 0.05. PIP treatment of diabetic rats significantly reduced levels of fasting glycemia, HbA1c, and serum AGEs, TGs, TC, and LDL-C compared to control diabetic group. PIP diminished aortic endothelial denudation and fibrous tissue proliferation compared to control STZ aortas. PIP lessened aortic contractility to phenylephrine and improved aortic relaxation to acetylcholine relative to untreated STZ group. PIP administration to diabetic rats elicited significant enhancements in GSH and SOD levels, eNOS expression, and total nitrate/nitrite bioavailability compared to untreated STZ rats. Moreover, PIP attenuated aortic contents of ROS, MDA, TXNIP protein and mRNA, NF-κB p65 mRNA, NLRP3 mRNA, IL-1β protein, and caspase-3 and TNF-α expressions compared to untreated STZ levels. In conclusion, PIP might ameliorate diabetes-associated functional and structural aortic remodeling by targeting TXNIP-NLRP3 signaling.
Collapse
|
11
|
Kandeel M, Iqbal MN, Ali I, Malik S, Malik A, Sehgal SA. Comprehensive in silico analyses of flavonoids elucidating the drug properties against kidney disease by targeting AIM2. PLoS One 2023; 18:e0285965. [PMID: 37200367 DOI: 10.1371/journal.pone.0285965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023] Open
Abstract
Kidney disorders are among the most common diseases and there is a scarcity of effective treatments for chronic kidney disease. There has been a progressive improvement in specific flavonoids for protective effects against kidney diseases. Flavonoids inhibit the regulatory enzymes to control inflammation-related diseases. In the present study, a hybrid approach of molecular docking analyses and molecular dynamic simulation was followed by principal component analyses and a dynamics cross-correlation matrix. In the present study, the top-ranked five flavonoids were reported, and the maximum binding affinity was observed against AIM2. Molecular docking analyses revealed that Glu_186, Phe_187, Lys_245, Glu_248, Ile_263, and Asn_265 are potent residues against AIM2 for ligand-receptor interactions. Extensive in silico analyses suggested that procyanidin is a potential molecule against AIM2. Moreover, the site-directed mutagenesis for the reported interacting residues of AIM2 could be important for further in vitro analyses. The observed novel results based on extensive computational analyses may be significant for potential drug design against renal disorders by targeting AIM2.
Collapse
Affiliation(s)
- Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Hofuf, Al-Ahsa, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelshikh University, Kafrelshikh, Egypt
| | - Muhammad Nasir Iqbal
- Department of Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Iqra Ali
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad, Pakistan
| | - Saima Malik
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Abbeha Malik
- Department of Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Sheikh Arslan Sehgal
- Department of Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
- Department of Bioinformatics, University of Okara, Okara, Pakistan
| |
Collapse
|
12
|
Asgari M, Salehi I, Ranjbar K, Khosravi M, Zarrinkalam E. Interval training and Crataegus persica ameliorate diabetic nephropathy via miR-126/Nrf-2 mediated inhibition of stress oxidative in rats with diabetes after myocardial ischemia-reperfusion injury. Biomed Pharmacother 2022; 153:113411. [PMID: 36076481 DOI: 10.1016/j.biopha.2022.113411] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 12/14/2022] Open
Abstract
Myocardial disorders are the most common cause of renal failure and mortality in diabetic patients, but the molecular mechanism of this process is not yet clear. The reduction of nuclear Erythroid2-related factor-2 (Nrf-2) and positive regulators of Nrf-2 proteins, such as DJ-1 and microRNA-126 (miR-126), after hypoxia and the promotion of reactive oxygen species, might be an intervention indicator in renal failure after myocardial ischemia-reperfusion. Therefore, this study evaluates the renoprotective effect of exercise training and Crataegus persica extract (CE) on myocardial ischemia-reperfusion-induced kidney injury in diabetic rats. Fifty rats were divided into five groups: healthy sedentary control (Con), sedentary diabetic (D), interval trained diabetic (TD), diabetic plus Crataegus persica extract treatment (CD), and interval trained diabetic plus Crataegus persica extract treatment (TCD) groups. The rats in the exercise groups were subjected to moderate-intensity interval training five days per week for ten weeks. The rats in CD and TCD groups received 300 mg/kg of Crataegus persica through gavage for ten weeks. Then, the subjects underwent 30 min of myocardial ischemia and subsequently reperfusion for 24 h. At the end of the experiment, insulin sensitivity, oxidative stress, renal function, histopathology of the kidney, Nrf-2, miR-126, and DJ-1 gene expression levels were evaluated. The results show that the treatments decreased elevated levels of renal oxidative stress, glomerular filtration rate, insulin sensitivity, and pathological score in diabetic rats. Also, the expression of Nrf-2 and miR-126, unlike DJ-1, decreased in diabetic rats due to interval training. Due to the results, diabetes aggravates acute myocardial ischemia-reperfusion-induced kidney injury, while moderate-intensity interval training and Crataegus persica treatment simultaneously ameliorate myocardial ischemia-reperfusion-induced renal injury via miR-126/Nrf-2 pathway and improve insulin sensitivity and renal function in type 1 diabetic rats.
Collapse
Affiliation(s)
- Masoumeh Asgari
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Islamic Republic of Iran
| | - Iraj Salehi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Islamic Republic of Iran; Department of Physiology, School of Medicine, Neurophysiology Research Center, Hamedan University of Medical Sciences, Hamedan, Islamic Republic of Iran.
| | - Kamal Ranjbar
- Department of Physical Education and Sport Science, Bandar Abbas Branch, Islamic Azad University, Bandar Abbas, Islamic Republic of Iran.
| | - Maryam Khosravi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Islamic Republic of Iran
| | - Ebrahim Zarrinkalam
- Department of Physical Education and Sport Science, Hamedan Branch, Islamic Azad University, Hamedan, Islamic Republic of Iran
| |
Collapse
|
13
|
Mertowska P, Mertowski S, Smarz-Widelska I, Grywalska E. Biological Role, Mechanism of Action and the Importance of Interleukins in Kidney Diseases. Int J Mol Sci 2022; 23:ijms23020647. [PMID: 35054831 PMCID: PMC8775480 DOI: 10.3390/ijms23020647] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Each year, the number of patients who are diagnosed with kidney disease too late is increasing, which leads to permanent renal failure. This growing problem affects people of every age, sex and origin, and its full etiopathogenesis is not fully understood, although the involvement of genetic susceptibility, infections, immune disorders or high blood pressure is suggested. Difficulties in making a correct and quick diagnosis are caused by the lack of research on early molecular markers, as well as educational and preventive activities among the public, which leads to the late detection of kidney diseases. An important role in the homeostasis and disease progression, including kidney diseases, is attributed to interleukins, which perform several biological functions and interact with other cells and tissues of the body. The aim of this article was to systematize the knowledge about the biological functions performed by interleukins in humans and their involvement in kidney diseases development. In our work, we took into account the role of interleukins in acute and chronic kidney disease and kidney transplantation.
Collapse
Affiliation(s)
- Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4A Chodzki Street, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4A Chodzki Street, 20-093 Lublin, Poland; (P.M.); (E.G.)
- Correspondence:
| | - Iwona Smarz-Widelska
- Department of Nephrology, Cardinal Stefan Wyszynski Provincial Hospital in Lublin, Al. Kraśnicka Street, 20-718 Lublin, Poland;
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4A Chodzki Street, 20-093 Lublin, Poland; (P.M.); (E.G.)
| |
Collapse
|
14
|
Elsayed MS, Abu-Elsaad NM, Nader MA. The NLRP3 inhibitor dapansutrile attenuates folic acid induced nephrotoxicity via inhibiting inflammasome/caspase-1/IL axis and regulating autophagy/proliferation. Life Sci 2021; 285:119974. [PMID: 34560082 DOI: 10.1016/j.lfs.2021.119974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 11/26/2022]
Abstract
AIMS Chemical renal toxicity is common and has limited therapeutic interventions. The NLRP3 inhibitor dapansutrile (DAPA) undergoes clinical phase II trials and it shows promising beneficial effects in various inflammatory diseases. The current study aims at evaluating the effect of DAPA on folic acid (FA) induced acute kidney injury (AKI) and its possible transition to chronic injury. MATERIALS AND METHODS Two treatment protocols were studied depending on DAPA injection timing. A prophylactic protocol involving the injection of DAPA (0.2 mg/kg) daily for seven days before FA challenge and a therapeutic protocol where DAPA was injected after FA. Each protocol included four groups of rats: control group, DAPA group, FA group and DAPA+FA group. Serum creatinine, urea and uric acid were measured. Also, kidney injury, necrosis and fibrosis percentage in addition to infiltration of CD68 positive cells were evaluated. Activation markers of inflammasome and the expression of Ki-67 and LC-3 were measured. KEY FINDINGS Results showed an improvement in renal tissue integrity and a significant decrease in kidney function biomarkers, caspase-1, IL-1β and IL-18 by DAPA injection (p < 0.05). In addition, DAPA decreased the proliferation marker Ki-67 and the autophagic marker LC-3 (p < 0.01). SIGNIFICANCE DAPA potentially alleviates FA induced nephrotoxicity through targeting inflammasome/caspase-1/IL axis. Moreover, it shows a regulatory effect on renal regeneration and autophagy.
Collapse
Affiliation(s)
- Mohamed S Elsayed
- Pharmacology and Toxicology Dep., Faculty of Pharmacy, Mansoura University, Egypt
| | - Nashwa M Abu-Elsaad
- Pharmacology and Toxicology Dep., Faculty of Pharmacy, Mansoura University, Egypt.
| | - Manar A Nader
- Pharmacology and Toxicology Dep., Faculty of Pharmacy, Mansoura University, Egypt
| |
Collapse
|
15
|
Burke RM, Dale BL, Dholakia S. The NLRP3 Inflammasome: Relevance in Solid Organ Transplantation. Int J Mol Sci 2021; 22:ijms221910721. [PMID: 34639062 PMCID: PMC8509131 DOI: 10.3390/ijms221910721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
The NOD, LRR, and pyrin domain-containing 3 (NLRP3) protein has been established as a central component of the inflammasome and regulates the inflammatory response to a myriad of environmental, microbial, and endogenous danger stimuli. Assembly of the NLRP3 inflammasome results in the cleavage and activation of caspase-1, in turn causing release of the pro-inflammatory interleukins 1-beta and 18. This activation response, while crucial to coordinated innate immune defense, can be aberrantly activated by the likes of cell-free DNA, and cause significant autoimmune pathology. Complications of autoimmunity induced by aberrant NLRP3 inflammasome activation have a great degree of mechanistic crossover with alloimmune injury in solid organ transplant, and stratagems to neutralize NLRP3 inflammasome activation may prove beneficial in solid organ transplant management. This article reviews NLRP3 inflammasome biology and the pathology associated with its hyperactivation, as well as the connections between NLRP3 inflammasome activation and allograft homeostasis.
Collapse
Affiliation(s)
- Ryan M. Burke
- CareDx, Inc., Brisbane, CA 94080, USA; (R.M.B.); (B.L.D.)
| | | | - Shamik Dholakia
- CareDx, Inc., Brisbane, CA 94080, USA; (R.M.B.); (B.L.D.)
- Oxford Transplant Center, Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 7LD, UK
- Correspondence:
| |
Collapse
|
16
|
Zhu Z, Liu J, Yang Y, Adu-Frimpong M, Ji H, Toreniyazov E, Wang Q, Yu J, Xu X. SMEDDS for improved oral bioavailability and anti-hyperuricemic activity of licochalcone A. J Microencapsul 2021; 38:459-471. [DOI: 10.1080/02652048.2021.1963341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Zhongan Zhu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jing Liu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuhang Yang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Michael Adu-Frimpong
- Department of Applied Chemistry and Biochemistry, Faculty of Applied Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, GH, UK
| | - Hao Ji
- Jiangsu Tian Sheng Pharmaceutical Co., Ltd., Zhenjiang, China
| | - Elmurat Toreniyazov
- Tashkent State Agricultural University (Nukus Branch), Nukus, Uzbekistan
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| |
Collapse
|
17
|
Chen JH, Wu CH, Chiang CK. Therapeutic Approaches Targeting Proteostasis in Kidney Disease and Fibrosis. Int J Mol Sci 2021; 22:ijms22168674. [PMID: 34445377 PMCID: PMC8395452 DOI: 10.3390/ijms22168674] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pathological insults usually disturb the folding capacity of cellular proteins and lead to the accumulation of misfolded proteins in the endoplasmic reticulum (ER), which leads to so-called “ER stress”. Increasing evidence indicates that ER stress acts as a trigger factor for the development and progression of many kidney diseases. The unfolded protein responses (UPRs), a set of molecular signals that resume proteostasis under ER stress, are thought to restore the adaptive process in chronic kidney disease (CKD) and renal fibrosis. Furthermore, the idea of targeting UPRs for CKD treatment has been well discussed in the past decade. This review summarizes the up-to-date literature regarding studies on the relationship between the UPRs, systemic fibrosis, and renal diseases. We also address the potential therapeutic possibilities of renal diseases based on the modulation of UPRs and ER proteostasis. Finally, we list some of the current UPR modulators and their therapeutic potentials.
Collapse
Affiliation(s)
- Jia-Huang Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan; (J.-H.C.); (C.-H.W.)
| | - Chia-Hsien Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan; (J.-H.C.); (C.-H.W.)
- Department of Physiology of Visceral Function and Body Fluid, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan; (J.-H.C.); (C.-H.W.)
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei 100225, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei 10672, Taiwan
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 88347)
| |
Collapse
|
18
|
Arulkumaran N, Pollen SJ, Tidswell R, Gaupp C, Peters VBM, Stanzani G, Snow TAC, Duchen MR, Singer M. Selective mitochondrial antioxidant MitoTEMPO reduces renal dysfunction and systemic inflammation in experimental sepsis. Br J Anaesth 2021; 127:577-586. [PMID: 34332740 DOI: 10.1016/j.bja.2021.05.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Excess mitochondrial reactive oxygen species (mROS) in sepsis is associated with organ failure, in part by generating inflammation through the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. We determined the impact of a mitochondrial-targeted antioxidant (MitoTEMPO) on mitochondrial dysfunction in renal proximal tubular epithelial cells, peritoneal immune cell function ex vivo, and organ dysfunction in a rat model of sepsis. METHODS The effects of MitoTEMPO were assessed ex vivo using adenosine triphosphate and lipopolysaccharide-stimulated rat peritoneal immune cells and fresh rat kidney slices exposed to serum from septic rats. We assessed mROS production and phagocytotic capacity (flow cytometry), mitochondrial functionality (multiphoton imaging, respirometry), and NLRP3 inflammasome activation in cell culture. The effect of MitoTEMPO on organ dysfunction was evaluated in a rat model of faecal peritonitis. RESULTS MitoTEMPO decreased septic serum-induced mROS (P<0.001) and maintained normal reduced nicotinamide adenine dinucleotide redox state (P=0.02) and mitochondrial membrane potential (P<0.001) in renal proximal tubular epithelial cells ex vivo. In lipopolysaccharide-stimulated peritoneal immune cells, MitoTEMPO abrogated the increase in mROS (P=0.006) and interleukin-1β (IL-1β) (P=0.03) without affecting non-mitochondrial oxygen consumption or the phagocytotic-induced respiratory burst (P>0.05). In vivo, compared with untreated septic animals, MitoTEMPO reduced systemic IL-1β (P=0.01), reduced renal oxidative stress as determined by urine isoprostane levels (P=0.04), and ameliorated renal dysfunction (reduced serum urea (P<0.001) and creatinine (P=0.05). CONCLUSIONS Reduction of mROS by a mitochondria-targeted antioxidant reduced IL-1β, and protected mitochondrial, cellular, and organ functionality after septic insults.
Collapse
Affiliation(s)
- Nishkantha Arulkumaran
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK.
| | - Sean J Pollen
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Robert Tidswell
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Charlotte Gaupp
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Vera B M Peters
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Giacomo Stanzani
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Timothy A C Snow
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Michael R Duchen
- Department of Cell and Development Biology, University College London, London, UK
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| |
Collapse
|
19
|
de Souza SI, Rocha EC, Ferraz HR, Dias JA, Seguro AC, Volpini RA, Canale D, de Bragança AC, Shimizu MHM, Marques LM, de Magalhães ACM, Coimbra TM, de Jesus Soares T. Acute kidney injury induced by glycerol is worsened by orchiectomy and attenuated by testosterone replacement. Steroids 2021; 165:108755. [PMID: 33171132 DOI: 10.1016/j.steroids.2020.108755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/15/2020] [Accepted: 10/24/2020] [Indexed: 12/24/2022]
Abstract
Although several studies have demonstrated that the male gender represents an independent risk factor for renal disease, evidence shows that androgens exert renal protective actions. The findings are controversial and no studies have evaluated the effects of orchiectomy and testosterone replacement on glycerol-induced renal injury. Male Wistar rats were submitted to orchiectomy or sham surgery and divided into four groups: SC, sham control rats injected with NaCl; SG, sham rats injected with glycerol; OG, orchiectomized rats injected with glycerol; OGT, orchiectomized rats injected with glycerol and testosterone. Testosterone was administered daily for 14 days in the OGT group. After 11 days of testosterone replacement in the OGT group, SC rats were submitted to a saline injection, while SG, OG and OGT rats received glycerol. All rats were euthanized three days after injections. OG rats presented higher serum creatinine and urea, and sodium excretion, compared to SC and SG, while testosterone attenuated these changes. Acute tubular necrosis was also mitigated by testosterone. Renal immunostaining for macrophages, lymphocytes and NF-κB was higher in OG compared to SC and SG. In addition, renal interleukin-1β, Caspase 3 and AT1 gene expression was higher in OG rats compared to SG. Testosterone attenuated these alterations, except the NF-κB immunostaining. The renal NO was lower in OG rats compared to SG. Only the OG rats presented decreases in serum NO and renal HO-1, and increased TNF-α, angiotensinogen and AT1 expression compared to SC. We conclude that orchiectomy worsened glycerol-induced kidney injury, while testosterone attenuated this renal damage.
Collapse
Affiliation(s)
- Samira Itana de Souza
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Edilene Cândido Rocha
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Halanna Rocha Ferraz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Jéssica Afonso Dias
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Antonio Carlos Seguro
- Laboratório de Investigação Médica 12 (LIM12), Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Rildo Aparecido Volpini
- Laboratório de Investigação Médica 12 (LIM12), Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Daniele Canale
- Laboratório de Investigação Médica 12 (LIM12), Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ana Carolina de Bragança
- Laboratório de Investigação Médica 12 (LIM12), Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Maria Heloisa Massola Shimizu
- Laboratório de Investigação Médica 12 (LIM12), Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Lucas Miranda Marques
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | | | - Terezila Machado Coimbra
- Departamento de Fisiologia, Universidade de São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil.
| |
Collapse
|
20
|
Zhang H, Zahid A, Ismail H, Tang Y, Jin T, Tao J. An overview of disease models for NLRP3 inflammasome over-activation. Expert Opin Drug Discov 2020; 16:429-446. [PMID: 33131335 DOI: 10.1080/17460441.2021.1844179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Inflammatory reactions, including those mediated by the NLRP3 inflammasome, maintain the body's homeostasis by removing pathogens, repairing damaged tissues, and adapting to stressed environments. However, uncontrolled activation of the NLRP3 inflammasome tends to cause various diseases using different mechanisms. Recently, many inhibitors of the NLRP3 inflammasome have been reported and many are being developed. In order to assess their efficacy, specificity, and mechanism of action, the screening process of inhibitors requires various types of cell and animal models of NLRP3-associated diseases.Areas covered: In the following review, the authors give an overview of the cell and animal models that have been used during the research and development of various inhibitors of the NLRP3 inflammasome.Expert opinion: There are many NLRP3 inflammasome inhibitors, but most of the inhibitors have poor specificity and often influence other inflammatory pathways. The potential risk for cross-reaction is high; therefore, the development of highly specific inhibitors is essential. The selection of appropriate cell and animal models, and combined use of different models for the evaluation of these inhibitors can help to clarify the target specificity and therapeutic effects, which is beneficial for the development and application of drugs targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ayesha Zahid
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hazrat Ismail
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science. Hefei National Science Center for Physical Sciences at Microscale. University of Science and Technology of China, Hefei, China
| | - Yujie Tang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Shanghai, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
21
|
Yu C, Zhang C, Kuang Z, Zheng Q. The Role of NLRP3 Inflammasome Activities in Bone Diseases and Vascular Calcification. Inflammation 2020; 44:434-449. [PMID: 33215255 PMCID: PMC7985100 DOI: 10.1007/s10753-020-01357-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023]
Abstract
Continuous stimulation of inflammation is harmful to tissues of an organism. Inflammatory mediators not only have an effect on metabolic and inflammatory bone diseases but also have an adverse effect on certain genetic and periodontal diseases associated with bone destruction. Inflammatory factors promote vascular calcification in various diseases. Vascular calcification is a pathological process similar to bone development, and vascular diseases play an important role in the loss of bone homeostasis. The NLRP3 inflammasome is an essential component of the natural immune system. It can recognize pathogen-related molecular patterns or host-derived dangerous signaling molecules, recruit, and activate the pro-inflammatory protease caspase-1. Activated caspase-1 cleaves the precursors of IL-1β and IL-18 to produce corresponding mature cytokines or recognizes and cleaves GSDMD to mediate cell pyroptosis. In this review, we discuss the role of NLRP3 inflammasome in bone diseases and vascular calcification caused by sterile or non-sterile inflammation and explore potential treatments to prevent bone loss.
Collapse
Affiliation(s)
- Chenyang Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Caihua Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Zhihui Kuang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Qiang Zheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China. .,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.
| |
Collapse
|
22
|
The selective c-Met inhibitor capmatinib offsets cisplatin-nephrotoxicity and doxorubicin-cardiotoxicity and improves their anticancer efficacies. Toxicol Appl Pharmacol 2020; 398:115018. [DOI: 10.1016/j.taap.2020.115018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
|
23
|
Amin FM, Abdelaziz RR, Hamed MF, Nader MA, Shehatou GSG. Dimethyl fumarate ameliorates diabetes-associated vascular complications through ROS-TXNIP-NLRP3 inflammasome pathway. Life Sci 2020; 256:117887. [PMID: 32497629 DOI: 10.1016/j.lfs.2020.117887] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 02/08/2023]
Abstract
Vascular complications are a leading cause of morbidity and mortality among diabetic patients. This work aimed to investigate possible influences of dimethyl fumarate (DMF) on streptozotocin (STZ) diabetes-associated vascular complications in rats, exploring its potential to modulate ROS-TXNIP-NLRP3 inflammasome pathway. Two weeks after induction of diabetes (via a single injection of 50 mg/kg STZ, i.p.), diabetic rats were administered either DMF (25 mg/kg/day) or its vehicle for further eight weeks. Age-matched normal and DMF-administered non-diabetic rats served as controls. DMF treatment elicited a mild ameliorative effect on diabetic glycemia. DMF reduced serum TG and AGE levels and enhanced serum HDL-C concentrations in diabetic rats. Moreover, DMF significantly diminished aortic levels of ROS and MDA and restored aortic GSH, SOD and Nrf2 to near-normal levels in STZ rats. Aortic mRNA levels of TXNIP, NLRP3 and NF-κB p65 in diabetic rats were significantly reduced by DMF treatment. Serum and aortic protein levels of TXNIP and aortic contents of IL-1β, iNOS, NLRP3 and TGF-β1 were significantly lower in DMF-diabetic animals than non-treated diabetic rats. Furthermore, protein expression of TNF-α and caspase-3 in diabetic aortas was greatly attenuated by DMF administration. DMF enhanced eNOS mRNA and protein levels and increased bioavailable NO in diabetic aortas. Functionally, DMF attenuated contractile responses of diabetic aortic rings to KCl and phenylephrine and enhanced their relaxant responses to acetylcholine. DMF also mitigated diabetes-induced fibrous tissue proliferation in aortic tunica media. Collectively, these findings demonstrate that DMF offered vasculoprotective influences on diabetic aortas via attenuation of ROS-TXNIP-NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Fatma M Amin
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed F Hamed
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - George S G Shehatou
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Egypt.
| |
Collapse
|
24
|
Diaz-Ricart M, Torramade-Moix S, Pascual G, Palomo M, Moreno-Castaño AB, Martinez-Sanchez J, Vera M, Cases A, Escolar G. Endothelial Damage, Inflammation and Immunity in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12060361. [PMID: 32492843 PMCID: PMC7354562 DOI: 10.3390/toxins12060361] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) patients have an accelerated atherosclerosis, increased risk of thrombotic-ischemic complications, and excessive mortality rates when compared with the general population. There is also evidence of an endothelial damage in which the proinflammatory state, the enhanced oxidative stress, or the accumulation of toxins due to their reduced renal clearance in uremia play a role. Further, there is evidence that uremic endothelial cells are both involved in and victims of the activation of the innate immunity. Uremic endothelial cells produce danger associated molecular patterns (DAMPS), which by binding to specific pattern recognition receptors expressed in multiple cells, including endothelial cells, induce the expression of adhesion molecules, the production of proinflammatory cytokines and an enhanced production of reactive oxygen species in endothelial cells, which constitute a link between immunity and inflammation. The connection between endothelial damage, inflammation and defective immunity in uremia will be reviewed here.
Collapse
Affiliation(s)
- Maribel Diaz-Ricart
- Hematopathology, Pathology Department, Center for Biomedical Diagnosis (CDB), Hospital Clinic, 08036 Barcelona, Spain; (S.T.-M.); (M.P.); (A.B.M.-C.); (J.M.-S.); (G.E.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain
- Barcelona Endothelium Team, 08036 Barcelona, Spain
- Correspondence:
| | - Sergi Torramade-Moix
- Hematopathology, Pathology Department, Center for Biomedical Diagnosis (CDB), Hospital Clinic, 08036 Barcelona, Spain; (S.T.-M.); (M.P.); (A.B.M.-C.); (J.M.-S.); (G.E.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain
| | | | - Marta Palomo
- Hematopathology, Pathology Department, Center for Biomedical Diagnosis (CDB), Hospital Clinic, 08036 Barcelona, Spain; (S.T.-M.); (M.P.); (A.B.M.-C.); (J.M.-S.); (G.E.)
- Barcelona Endothelium Team, 08036 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, 08036 Barcelona, Spain
| | - Ana Belen Moreno-Castaño
- Hematopathology, Pathology Department, Center for Biomedical Diagnosis (CDB), Hospital Clinic, 08036 Barcelona, Spain; (S.T.-M.); (M.P.); (A.B.M.-C.); (J.M.-S.); (G.E.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain
- Barcelona Endothelium Team, 08036 Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hematopathology, Pathology Department, Center for Biomedical Diagnosis (CDB), Hospital Clinic, 08036 Barcelona, Spain; (S.T.-M.); (M.P.); (A.B.M.-C.); (J.M.-S.); (G.E.)
- Barcelona Endothelium Team, 08036 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, 08036 Barcelona, Spain
| | - Manel Vera
- Nephrology Department. Hospital Clinic, 08036 Barcelona, Spain; (M.V.); (A.C.)
| | - Aleix Cases
- Nephrology Department. Hospital Clinic, 08036 Barcelona, Spain; (M.V.); (A.C.)
| | - Gines Escolar
- Hematopathology, Pathology Department, Center for Biomedical Diagnosis (CDB), Hospital Clinic, 08036 Barcelona, Spain; (S.T.-M.); (M.P.); (A.B.M.-C.); (J.M.-S.); (G.E.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain
- Barcelona Endothelium Team, 08036 Barcelona, Spain
| |
Collapse
|
25
|
Synergistic activation of p53 by actinomycin D and nutlin-3a is associated with the upregulation of crucial regulators and effectors of innate immunity. Cell Signal 2020; 69:109552. [PMID: 32032660 PMCID: PMC7126238 DOI: 10.1016/j.cellsig.2020.109552] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 02/02/2023]
Abstract
Actinomycin D and nutlin-3a (A + N) activate p53, partly through induction of phosphorylation on Ser392. The death of A549 cells induced by A + N morphologically resembles inflammation-inducing pyroptosis - cell destruction triggered by activated caspase-1. The treatment with A + N (or camptothecin) strongly upregulated caspase-1 and its two activators: IFI16 and NLRP1, however, caspase-1 activation was not detected. A549 cells may have been primed for pyroptosis, with the absence of a crucial trigger. The investigation of additional innate immunity elements revealed that A + N (or camptothecin) stimulated the expression of NLRX1, STING (stimulator of interferon genes) and two antiviral proteins, IFIT1 and IFIT3. IFI16 and caspase-1 are coded by p53-regulated genes which led us to investigate regulation of NLRP1, NLRX1, STING, IFIT1 and IFIT3 in p53-dependent mode. The upregulation of NLRP1, NLRX1 and STING was attenuated in p53 knockdown cells. The upsurge of the examined genes, and activation of p53, was inhibited by C16, an inhibitor of PKR kinase. PKR was tested due to its ability to phosphorylate p53 on Ser392. Surprisingly, C16 was active even in PKR knockdown cells. The ability of C16 to prevent activation of p53 and expression of innate immunity genes may be the source of its strong anti-inflammatory action. Moreover, cells exposed to A + N can influence neighboring cells in paracrine fashion, for instance, they shed ectodomain of COL17A1 protein and induce, in p53-dependent mode, the expression of gene for interleukin-7. Further, the activation of p53 also spurred the expression of SOCS1, an inhibitor of interferon triggered STAT1-dependent signaling. We conclude that, stimulation of p53 primes cells for the production of interferons (through upregulation of STING), and may activate negative-feedback within this signaling system by enhancing the production of SOCS1. Actinomycin D and nutlin-3a strongly and synergistically activate p53 protein Strongly activated p53 promotes expression of innate immunity genes Strong activation of innate immunity genes can be prevented by C16 compound By inducing SOCS1 protein p53 can prevent overactivation of interferon signaling Strongly activated p53 can send signal to nearby immune cells through interleukin-7
Collapse
|
26
|
Hirsutella sinensis inhibits NLRP3 inflammasome activation to block aristolochic acid-induced renal tubular epithelial cell transdifferentiation. Hum Cell 2019; 33:79-87. [PMID: 31776855 DOI: 10.1007/s13577-019-00306-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 11/20/2019] [Indexed: 10/25/2022]
Abstract
In recent years, kidney damage caused by ingestion of Chinese medicinal herbs containing Aristolochic acid (AA) has attracted extensive attention. However, whether the nephrotoxicity of AA is related to NLRP3 inflammasome has not been reported. Hirsutella sinensis (HS) has a certain therapeutic effect on aristolochic acid nephropathy (AAN) and is related to NLRP3 inflammasome. Therefore, this study explores whether HS plays a role in renal injury induced by AA through NLRP3 inflammasome pathway. AA-stimulated renal tubular epithelial cells showed that AA could promote the expression of NLRP3, ASC, and α-SMA, increase the secretion and expression of caspase-1, IL-1β, and IL-18, and inhibit the expression of E-cadherin in a dose- and time-dependent manner. When NLRP3 was down-regulated, the expression of α-SMA and E-cadherin did not change significantly, but significantly blocked the regulation of α-SMA and E-cadherin expression by AA. When AA and HS were added to renal tubular epithelial cells at the same time, the effects of AA on the expression of NLRP3, ASC, caspase-1, IL-1β, IL-18, and α-SMA gradually decreased to the level of control group with the increase of HS dosage. At the same time, HS can reduce the transdifferentiation of renal tubular epithelial cells by inhibiting the activation of NLRP3 inflammasome. These findings will provide important pharmacological references for the treatment of AAN and the clinical application of HS.
Collapse
|
27
|
The Role of Cardiolipin and Mitochondrial Damage in Kidney Transplant. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3836186. [PMID: 31885786 PMCID: PMC6899302 DOI: 10.1155/2019/3836186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) is highly incident and prevalent in the world. The death of patients with CKD is primarily due to cardiovascular disease. Renal transplantation (RT) emerges as the best management alternative for patients with CKD. However, the incidence of acute renal graft dysfunction is 11.8% of the related living donor and 17.4% of the cadaveric donor. Anticardiolipin antibodies (ACAs) or antiphospholipid antibodies (APAs) are important risk factors for acute renal graft dysfunction. The determination of ACA or APA to candidates for RT could serve as prognostic markers of early graft failure and would indicate which patients could benefit from anticoagulant therapy. Cardiolipin is a fundamental molecule that plays an important role in the adequate conformation of the mitochondrial cristae and the correct assembly of the mitochondrial respiratory supercomplexes and other proteins essential for proper mitochondrial function. Cardiolipin undergoes a nonrandom oxidation process by having pronounced specificity unrelated to the polyunsaturation pattern of its acyl groups. Accumulation of hydroxyl derivatives and cardiolipin hydroperoxides has been observed in the affected tissues, and recent studies showed that oxidation of cardiolipin is carried out by a cardiolipin-specific peroxidase activity of cardiolipin-bound cytochrome c. Cardiolipin could be responsible for the proapoptotic production of death signals. Cardiolipin modulates the production of energy and participates in inflammation, mitophagy, and cellular apoptosis. The determination of cardiolipin or its antibodies is an attractive therapeutic, diagnostic target in RT and kidney diseases.
Collapse
|
28
|
Li D, Qi C, Zhou J, Wen Z, Zhu X, Xia H, Song J. LPS-induced inflammation delays the transportation of ASP + due to down-regulation of OCTN1/2 in alveolar epithelial cells. J Drug Target 2019; 28:437-447. [PMID: 31591905 DOI: 10.1080/1061186x.2019.1678169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Organic cation transporters (OCTNs) can significantly affect drug disposition in alveolar epithelial cells (A549), but this process is not well understood. We investigated the expression and function of OCTN1/2 in A549 cells under different inflammatory status to examine pulmonary drug distribution. This experiment used lipopolysaccharide (LPS)-treated A549 cells to mimic inflammation in alveolar epithelial cells, and the expression of OCTN1/2, interleukin-6 (IL6), IL18, IL1β and tumour necrosis factor-alpha (TNF-α) was investigated by western blot and quantitative real-time PCR (qRT-PCR). The fluorescent compound 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide (ASP+) was chosen as a probe to study the activity of OCTN1/2. OCTN1/2 down-regulation induced by LPS was more pronounced than that in normal control (NC) groups. Experiments further detected the release of inflammatory factors that revealed a negative correlation between OCTN1/2 expression and inflammation secretion in human alveolar epithelial cells exposed to different concentrations of LPS. The Michaelis constant (Km) and apparent permeability coefficient (Papp) of ASP+ were also decreased significantly. Our results thus show that LPS-induced inflammation could inhibit the expression and activity of OCTN1/2 in vitro and reduce the distribution of inhaled medicine in pulmonary diseases.
Collapse
Affiliation(s)
- Dalang Li
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Chuanzong Qi
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Jian Zhou
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Zeqiang Wen
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Xiangyu Zhu
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Hongguang Xia
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Jue Song
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
29
|
Inoue T, Kusano T, Amano H, Nakamoto H, Okada H. Cellular communication network factor 2 (CCN2) promotes the progression of acute kidney injury to chronic kidney disease. Biochem Biophys Res Commun 2019; 517:96-102. [PMID: 31320136 DOI: 10.1016/j.bbrc.2019.07.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 07/06/2019] [Indexed: 11/28/2022]
Abstract
Here we evaluated the efficacy of depleting cellular communication network factor 2 (CCN2) produced by renal tubular epithelial cells in preventing the progression of severe acute kidney injury (AKI) to chronic kidney disease (CKD). We used conditional Ccn2 knockout mice in which expression of Ccn2 was controlled by γ-glutamyl transpeptidase promoter-regulated Cre recombinase. AKI was induced by ischemia-reperfusion injury. An effect of inhibiting Ccn2 expression by tubular epithelial cells on acute damage, assessed according to the levels of kidney injury molecule-1, was not detected 3 days after injury. However, by day 14, interstitial fibrosis and the levels of the extracellular matrix and profibrotic cytokines were reduced in Ccn2 knockout mice compared with wild-type mice. The ectopic expression of the pan-caspase inhibitor p35 reduced the number of apoptotic cells in damaged tubular epithelial cells 3 days after ischemia-reperfusion injury. In contrast, interstitial fibrosis was exacerbated, accompanied by increased levels of transforming growth factor-β and plasminogen-activator inhibitor-1 14 days after insult. Depletion of CCN2 from tubular epithelial cells slowed the progression of interstitial fibrosis, which was promoted by ectopic expression of p35 in the same cells. These results indicate that tubular epithelial cells, which should be eliminated by apoptosis during physiological repair of AKI, produced CCN2 in the damaged kidney and that CCN2 expression in damaged tubular epithelial cells made a critical contribution to the transition from AKI to CKD. Moreover, inhibiting CCN2 expression may represent a therapeutic approach for preventing the progression of AKI to CKD, irrespective of the stage of kidney disease.
Collapse
Affiliation(s)
- Tsutomu Inoue
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.
| | - Takeru Kusano
- Department of General Internal Medicine, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.
| | - Hiroaki Amano
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.
| | - Hidetomo Nakamoto
- Department of General Internal Medicine, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.
| | - Hirokazu Okada
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.
| |
Collapse
|
30
|
Ito S, Ohno Y, Tanaka T, Kobuchi S, Ayajiki K, Manabe E, Masuyama T, Jun-Ichi S, Tsujino T. Neutrophil/lymphocyte ratio elevation in renal dysfunction is caused by distortion of leukocyte hematopoiesis in bone marrow. Ren Fail 2019; 41:284-293. [PMID: 31014150 PMCID: PMC6493297 DOI: 10.1080/0886022x.2019.1597736] [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: 12/18/2022] Open
Abstract
Objective: We investigate the mechanism of neutrophil/lymphocyte ratio (NLR) elevation, a useful prognostic marker in patients with cardiovascular diseases (CVDs). Methods: In this clinical study, we retrospectively searched for factors associated with NLR elevation in cardiovascular outpatients. In animal experiments using mice with adenine-induced nephropathy, we further examined the hematopoietic process in bone marrow and explored the mechanism of NLR elevation. Result: In patients with CVDs or their risk factors, multiple regression analysis revealed that decrease in estimated glemerular filtration rate and increase in white blood cell count were significantly associated with increase in NLR. In mice with adenine-induced nephropathy, NLR and serum indoxyl sulfate (IS) levels were increased. Fluorescence-activated cell sorting revealed the increase in the number of myeloid progenitors and decrease in the number of common lymphoid progenitors, suggesting biased granulocyte side in the hematopoietic process in bone marrow. Treatment with oral charcoal adsorbent AST-120 decreased serum concentration of IS and normalized NLR and bone marrow abnormalities in mice with adenine-induced nephropathy. Conclusion: Renal function was a strong determinant of NLR in cardiovascular outpatients. NLR elevation due to renal impairment is caused by distortion of the hematopoietic process in bone marrow. IS plays a significant role in these processes.
Collapse
Affiliation(s)
- Satoyasu Ito
- a Division of Pharmaceutical Therapeutics, Department of Pharmacy, School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan
| | - Yoshiya Ohno
- b Division of Immunobiology, Department of Pharmacy, School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan
| | - Toshiyuki Tanaka
- b Division of Immunobiology, Department of Pharmacy, School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan
| | - Shuhei Kobuchi
- c Division of Pharmacology, Department of Pharmacy, School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan
| | - Kazuhide Ayajiki
- c Division of Pharmacology, Department of Pharmacy, School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan
| | - Eri Manabe
- d Cardiovascular Division, Department of Internal Medicine , Hyogo College of Medicine , Nishinomiya , Japan
| | - Tohru Masuyama
- d Cardiovascular Division, Department of Internal Medicine , Hyogo College of Medicine , Nishinomiya , Japan
| | - Sakamoto Jun-Ichi
- e Graduate School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan
| | - Takeshi Tsujino
- a Division of Pharmaceutical Therapeutics, Department of Pharmacy, School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan.,d Cardiovascular Division, Department of Internal Medicine , Hyogo College of Medicine , Nishinomiya , Japan.,e Graduate School of Pharmacy , Hyogo University of Health Sciences , Kobe , Japan
| |
Collapse
|
31
|
Alarcon MML, Trentin-Sonoda M, Panico K, Schleier Y, Duque T, Moreno-Loaiza O, de Yurre AR, Ferreira F, Caio-Silva W, Coury PR, Paiva CN, Medei E, Carneiro-Ramos MS. Cardiac arrhythmias after renal I/R depend on IL-1β. J Mol Cell Cardiol 2019; 131:101-111. [PMID: 31029578 DOI: 10.1016/j.yjmcc.2019.04.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 01/10/2023]
Abstract
AIMS Cardiac arrhythmias are one of the most important remote complications after kidney injury. Renal ischemia reperfusion (I/R) is a major cause of acute renal injury predisposing to several remote dysfunctions, including cardiac electrical disturbance. Since IL-1β production dependent on NLRP3 represents a link between tissue malfunctioning and cardiac arrhythmias, here we tested the hypothesis that longer ventricular repolarization and arrhythmias after renal I/R depend on this innate immunity sensor. METHODS AND RESULTS Nlrp3-/- and Casp1-/- mice reacted to renal I/R with no increase in plasma IL-1β, different from WT (wild-type) I/R. A prolonged QJ interval and an increased susceptibility to ventricular arrhythmias were found after I/R compared to Sham controls in wild-type mice at 15 days post-perfusion, but not in Nlrp3-/- or CASP1-/- I/R, indicating that the absence of NLRP3 or CASP1 totally prevented longer QJ interval after renal I/R. In contrast with WT mice, we found no renal atrophy and no renal dysfunction in Nlrp3-/- and Casp1-/- mice after renal I/R. Depletion of macrophages in vivo after I/R and a day before IL-1β peak (at 7 days post-perfusion) totally prevented prolongation of QJ interval, suggesting that macrophages might participate as sensors of tissue injury. Moreover, treatment of I/R-WT mice with IL-1r antagonist (IL-1ra) from 8 to 15 days post perfusion did not interfere with renal function, but reversed QJ prolongation, prevented the increase in susceptibility to ventricular arrhythmias and rescued a close to normal duration and amplitude of calcium transient. CONCLUSION Taken together, these results corroborate the hypothesis that IL-1β is produced after sensing renal injury through NRLP3-CASP1, and IL-1β on its turn triggers longer ventricular repolarization and increase susceptibility to cardiac arrhythmias. Still, they offer a therapeutic approach to treat cardiac arrhythmias that arise after renal I/R.
Collapse
Affiliation(s)
| | - Mayra Trentin-Sonoda
- Center of Natural and Human Sciences (CCNH), Universidade Federal do ABC, Santo André, SP, Brazil
| | - Karine Panico
- Center of Natural and Human Sciences (CCNH), Universidade Federal do ABC, Santo André, SP, Brazil
| | - Ygor Schleier
- Laboratory of Cardioimmunology, Federal University of Rio de Janeiro, Institute of Biophysics Carlos Chagas Filho, Rio de Janeiro, Brazil
| | - Thabata Duque
- Laboratory of Cardioimmunology, Federal University of Rio de Janeiro, Institute of Biophysics Carlos Chagas Filho, Rio de Janeiro, Brazil
| | - Oscar Moreno-Loaiza
- Laboratory of Cardioimmunology, Federal University of Rio de Janeiro, Institute of Biophysics Carlos Chagas Filho, Rio de Janeiro, Brazil
| | - Ainhoa Rodriguez de Yurre
- Laboratory of Cardioimmunology, Federal University of Rio de Janeiro, Institute of Biophysics Carlos Chagas Filho, Rio de Janeiro, Brazil
| | - Fabianno Ferreira
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wellington Caio-Silva
- Center of Natural and Human Sciences (CCNH), Universidade Federal do ABC, Santo André, SP, Brazil
| | - Pedrosa Roberto Coury
- Clementino Fraga Filho Hospital - Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia N Paiva
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emiliano Medei
- Laboratory of Cardioimmunology, Federal University of Rio de Janeiro, Institute of Biophysics Carlos Chagas Filho, Rio de Janeiro, Brazil; National Center for Structural Biology and Bioimaging - CENABIO/UFRJ, Rio de Janeiro, Brazil.
| | | |
Collapse
|
32
|
|
33
|
Arulkumaran N, Sixma ML, Pollen S, Ceravola E, Jentho E, Prendecki M, Bass PS, Tam FWK, Unwin RJ, Singer M. P2X 7 receptor antagonism ameliorates renal dysfunction in a rat model of sepsis. Physiol Rep 2019; 6. [PMID: 29488356 PMCID: PMC5828936 DOI: 10.14814/phy2.13622] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 01/27/2023] Open
Abstract
Sepsis is a major clinical problem associated with significant organ dysfunction and high mortality. The ATP‐sensitive P2X7 receptor activates the NLRP3 inflammasome and is a key component of the innate immune system. We used a fluid‐resuscitated rat model of fecal peritonitis and acute kidney injury (AKI) to investigate the contribution of this purinergic receptor to renal dysfunction in sepsis. Six and 24 h time‐points were chosen to represent early and established sepsis, respectively. A selective P2X7 receptor antagonist (A‐438079) dissolved in dimethyl sulfoxide (DMSO) was infused 2 h following induction of sepsis. Compared with sham‐operated animals, septic animals had significant increases in heart rate (−1(−4 to 8)% vs. 21(12–26)%; P = 0.003), fever (37.4(37.2–37.6)°C vs. 38.6(38.2–39.0)°C; P = 0.0009), and falls in serum albumin (29(27–30)g/L vs. 26(24–28); P = 0.0242). Serum IL‐1β (0(0–10)(pg/mL) vs. 1671(1445–33778)(pg/mL); P < 0.001) and renal IL‐1β (86(50–102)pg/mg protein vs. 200 (147–248)pg/mg protein; P = 0.0031) were significantly elevated in septic compared with sham‐operated animals at 6 h. Serum creatinine was elevated in septic animals compared with sham‐operated animals at 24 h (23(22–25) μmol/L vs. 28 (25–30)μmol/L; P = 0.0321). Renal IL‐1β levels were significantly lower in A‐438079‐treated animals compared with untreated animals at 6 h (70(55–128)pg/mg protein vs. 200(147–248)pg/mg protein; P = 0.021). At 24 h, compared with untreated animals, A‐438079‐treated animals had more rapid resolution of tachycardia (22(13–36)% vs. −1(−6 to 7)%; P = 0.019) and fever (39.0(38.6–39.1)°C vs. 38.2(37.6–38.7)°C; P < 0.024), higher serum albumin (23(21–25)g/L vs. (27(25–28)g/L); P = 0.006), lower arterial lactate (3.2(2.5–4.3)mmol/L vs. 1.4(0.9–1.8)mmol/L; P = 0.037), and lower serum creatinine concentrations (28(25–30)μmol/L vs. 22(17–27)μmol/L; P = 0.019). P2X7A treatment ameliorates the systemic inflammatory response and renal dysfunction in this clinically relevant model of sepsis‐related AKI.
Collapse
Affiliation(s)
- Nishkantha Arulkumaran
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom.,Division of Medicine, Department of Nephrology, University College London, London, United Kingdom.,Imperial College Renal and Transplant Centre, Hammersmith Hospital, London, United Kingdom
| | - Marije L Sixma
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - Sean Pollen
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - Elias Ceravola
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - Elisa Jentho
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - Maria Prendecki
- Imperial College Renal and Transplant Centre, Hammersmith Hospital, London, United Kingdom
| | - Paul S Bass
- Division of Medicine, Department of Nephrology, University College London, London, United Kingdom.,Department of cellular pathology, Royal Free hospital, London, United Kingdom
| | - Frederick W K Tam
- Imperial College Renal and Transplant Centre, Hammersmith Hospital, London, United Kingdom
| | - Robert J Unwin
- Division of Medicine, Department of Nephrology, University College London, London, United Kingdom
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| |
Collapse
|
34
|
SGK1-dependent stimulation of vascular smooth muscle cell osteo-/chondrogenic transdifferentiation by interleukin-18. Pflugers Arch 2019; 471:889-899. [PMID: 30706178 PMCID: PMC6533237 DOI: 10.1007/s00424-019-02256-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/04/2019] [Accepted: 01/13/2019] [Indexed: 01/18/2023]
Abstract
The serum- and glucocorticoid-inducible kinase 1 (SGK1) is a key regulator of osteo-/chondrogenic transdifferentiation and subsequent calcification of vascular smooth muscle cells (VSMCs). The phenotypical transdifferentiation of VSMCs is associated with increased interleukin-18 (IL-18) levels and generalized inflammation. Therefore, the present study investigated the possible involvement of SGK1 in IL-18-induced vascular calcification. Experiments were performed in primary human aortic smooth muscle cells (HAoSMCs) treated with recombinant human IL-18 protein in control or high phosphate conditions and following SGK1 knockdown by siRNA or pharmacological inhibition of SGK1, PI3K, and PDK1. As a result, IL-18 treatment increased SGK1 mRNA and protein expression in HAoSMCs. IL-18 upregulated SGK1 mRNA expression in a dose-dependent manner. This effect was paralleled by upregulation of the mRNA expression of MSX2 and CBFA1, osteogenic transcription factors, and of tissue-nonspecific alkaline phosphatase (ALPL), an osteogenic enzyme, as markers of increased osteo-/chondrogenic transdifferentiation. Phosphate treatment increased SGK1 and osteogenic markers mRNA expression as well as ALPL activity and induced calcification of HAoSMCs, all effects significantly augmented by additional treatment with IL-18. Conversely, silencing of SGK1 or cotreatment with the SGK1 inhibitor EMD638683 blunted the effects of IL-18 on osteo-/chondrogenic transdifferentiation and calcification of HAoSMCs. The procalcific effects of IL-18 were similarly suppressed in the presence of PI3K or PDK1 inhibitors. In conclusion, SGK1 expression is upregulated by IL-18 in VSMCs and SGK1 participates in the intracellular signaling of IL-18-induced osteo-/chondrogenic transdifferentiation of VSMCs. Thus, SGK1 may serve as therapeutic target to limit the progression of medial vascular calcification during vascular inflammation.
Collapse
|
35
|
Inflammation-Related Mechanisms in Chronic Kidney Disease Prediction, Progression, and Outcome. J Immunol Res 2018; 2018:2180373. [PMID: 30271792 PMCID: PMC6146775 DOI: 10.1155/2018/2180373] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022] Open
Abstract
Persistent, low-grade inflammation is now considered a hallmark feature of chronic kidney disease (CKD), being involved in the development of all-cause mortality of these patients. Although substantial improvements have been made in clinical care, CKD remains a major public health burden, affecting 10–15% of the population, and its prevalence is constantly growing. Due to its insidious nature, CKD is rarely diagnosed in early stages, and once developed, its progression is unfortunately irreversible. There are many factors that contribute to the setting of the inflammatory status in CKD, including increased production of proinflammatory cytokines, oxidative stress and acidosis, chronic and recurrent infections, altered metabolism of adipose tissue, and last but not least, gut microbiota dysbiosis, an underestimated source of microinflammation. In this scenario, a huge step forward was made by the increasing progression of omics approaches, specially designed for identification of biomarkers useful for early diagnostic and follow-up. Recent omics advances could provide novel insights in deciphering the disease pathophysiology; thus, identification of circulating biomarker panels using state-of-the-art proteomic technologies could improve CKD early diagnosis, monitoring, and prognostics. This review aims to summarize the recent knowledge regarding the relationship between inflammation and CKD, highlighting the current proteomic approaches, as well as the inflammasomes and gut microbiota dysbiosis involvement in the setting of CKD, culminating with the troubling bidirectional connection between CKD and renal malignancy, raised on the background of an inflammatory condition.
Collapse
|
36
|
APOL1: The Balance Imposed by Infection, Selection, and Kidney Disease. Trends Mol Med 2018; 24:682-695. [PMID: 29886044 DOI: 10.1016/j.molmed.2018.05.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) affects millions of people and constitutes a major health and financial burden worldwide. People of African descent are at an increased risk of developing kidney disease, which is mostly explained by two variants in the Apolipoprotein L1 (APOL1) gene that are found only in people of west African origin. It is hypothesized that these variants were genetically selected due to the protection they afford against African sleeping sickness, caused by the parasite Trypanosoma brucei. Targeting mutant APOL1 could have substantial therapeutic potential for treating kidney disease. In this review, we will describe the intriguing interplay between microbiology, genetics, and kidney disease as revealed in APOL1-associated kidney disease, discuss APOL1-induced cytotoxicity and its therapeutic implications.
Collapse
|
37
|
Conley SM, Abais JM, Boini KM, Li PL. Inflammasome Activation in Chronic Glomerular Diseases. Curr Drug Targets 2018; 18:1019-1029. [PMID: 27538510 DOI: 10.2174/1389450117666160817103435] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 07/01/2016] [Accepted: 07/19/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND The intracellular multiprotein complex termed the inflammasome functions as a platform of pro-inflammatory cytokine production such as IL-1β and IL-18. Under certain conditions, however, the inflammasome produces non-canonical effects such as induction of cell death, pyroptosis and cell metabolism alterations. OBJECTIVE In mammalian cells, several types of inflammasomes were identified, but the most widely studied one is the inflammasome containing NOD-like receptor with pyrin domain 3 (NLRP3), which has recently been reported as a central pathogenic mechanism of chronic degenerative diseases. Many activators or risk factors exert their actions through the activation of the NLRP3 inflammasome to produce a variety of functional changes in different cells including inflammatory, metabolic or survival responses. Several molecular signaling pathways are shown to mediate the activation of the NLRP3 inflammasome, and they are related to the modifications in K+ efflux, increased lysosome leakage and activation of cathepsin B or enhanced reactive oxygen species (ROS) production. In the kidney, inflammation is believed to mediate or promote the progression of glomerular sclerotic pathologies resulting in end-stage renal disease (ESRD). NLRP3 inflammasome activation may turn on glomerular inflammation and other cell damages, contributing to the onset of glomerular injury and ESRD. This inflammasome activation not only occurs in immune cells, but also in residential cells such as endothelial cells and podocytes in the glomeruli. SUMMARY This review briefly summarizes current evidence of NLRP3 inflammasome activation and related molecular mechanisms in renal glomeruli. The possible canonical and non-canonical effects of this inflammasome activation and its potential implication in the development of different glomerular diseases are highlighted.
Collapse
Affiliation(s)
- Sabena M Conley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298. United States
| | - Justine M Abais
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298. United States
| | - Krishna M Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204. United States
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298. United States
| |
Collapse
|
38
|
Alicic RZ, Johnson EJ, Tuttle KR. Inflammatory Mechanisms as New Biomarkers and Therapeutic Targets for Diabetic Kidney Disease. Adv Chronic Kidney Dis 2018; 25:181-191. [PMID: 29580582 DOI: 10.1053/j.ackd.2017.12.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 02/08/2023]
Abstract
Diabetic kidney disease (DKD) is the leading cause of CKD and end-stage kidney disease (ESKD) worldwide. Approximately 30-40% of people with diabetes develop this microvascular complication, placing them at high risk of losing kidney function as well as of cardiovascular events, infections, and death. Current therapies are ineffective for arresting kidney disease progression and mitigating risks of comorbidities and death among patients with DKD. As the global count of people with diabetes will soon exceed 400 million, the need for effective and safe treatment options for complications such as DKD becomes ever more urgent. Recently, the understanding of DKD pathogenesis has evolved to recognize inflammation as a major underlying mechanism of kidney damage. In turn, inflammatory mediators have emerged as potential biomarkers and therapeutic targets for DKD. Phase 2 clinical trials testing inhibitors of monocyte-chemotactic protein-1 chemokine C-C motif-ligand 2 and the Janus kinase/signal transducer and activator of transcription pathway, in particular, have produced promising results.
Collapse
|
39
|
Chin LH, Hsu YJ, Hsu SC, Chen YH, Chang YL, Huang SM, Tsai CS, Lin CY. The regulation of NLRP3 inflammasome expression during the development of cardiac contractile dysfunction in chronic kidney disease. Oncotarget 2017; 8:113303-113317. [PMID: 29371912 PMCID: PMC5768329 DOI: 10.18632/oncotarget.22964] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation plays a crucial role in the long-term complications in patients with chronic kidney disease (CKD). This study aimed to assess the role of NLR pyrin domain-containing protein (NLRP3) inflammasome in cardiac contractile dysfunctions in CKD. The cardiac contractile function was evaluated and the expression of NLRP3 inflammasome and related cytokines in the heart was assessed in a murine sham-operated and 5/6 nephrectomy CKD model in vivo. In vitro, H9c2 cells were treated with uremic toxin indoxyl sulfate (IS), with or without NLRP3 inflammasome inhibition, which was achieved by using small interfering RNA (siRNA)-mediated knockdown of the NLRP3 gene. Moreover, the activation of nuclear factor κB (NF-κB) signaling and apoptosis marker levels were assessed in the IS-treated H9c2 cells. The results demonstrated that CKD can lead to the development of cardiac contractile dysfunction in vivo associated with the upregulation of NLRP3 inflammasome, IL-1β, IL-18, and contribute to the myocardial apoptosis. In vitro experiments showed the upregulation of inflammasome, IL-1β, and IL-18 levels, and cell apoptosis in the IS-treated H9c2 cells through the activation of NF-κB signaling pathway. The transfection of cells with si-NLRP3 was shown to alleviate IL-1β, IL-18, and cell apoptosis. Moreover, decreased cell viability induced by IS was shown to be attenuated by IL-1β or IL-18-neutralizing antibody. In summary, CKD can result in the development of cardiac contractile dysfunction associated with the upregulation of NLRP3 inflammasome/IL-1β/IL-18 axis induced by the uremic toxins.
Collapse
Affiliation(s)
- Li-Han Chin
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Che Hsu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Superintendent's Office, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Chih-Yuan Lin
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
40
|
Zhang K, Zhang Y, Feng W, Chen R, Chen J, Touyz RM, Wang J, Huang H. Interleukin-18 Enhances Vascular Calcification and Osteogenic Differentiation of Vascular Smooth Muscle Cells Through TRPM7 Activation. Arterioscler Thromb Vasc Biol 2017; 37:1933-1943. [DOI: 10.1161/atvbaha.117.309161] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/09/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Kun Zhang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Yinyin Zhang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Weijing Feng
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Renhua Chen
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Jie Chen
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Rhian M. Touyz
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Jingfeng Wang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Hui Huang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| |
Collapse
|
41
|
Wang Y, Yu B, Wang L, Yang M, Xia Z, Wei W, Zhang F, Yuan X. Pioglitazone ameliorates glomerular NLRP3 inflammasome activation in apolipoprotein E knockout mice with diabetes mellitus. PLoS One 2017; 12:e0181248. [PMID: 28708885 PMCID: PMC5510862 DOI: 10.1371/journal.pone.0181248] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/28/2017] [Indexed: 11/18/2022] Open
Abstract
Objective The NLRP3 inflammasome plays an important role in the pathogenesis of inflammation in diabetic nephropathy (DN). Pioglitazone (PIO) has been found to exert an anti-inflammatory effect in patients with diabetes mellitus, but it is still unclear whether PIO exhibits a similar effect in DN. We aimed to explore the effect and underlying mechanism of PIO on DN, as well as investigate if NLRP3 is a pharmacologic target of PIO. Methods We divided 48 apolipoprotein E (apoE) (-/-) mice into 4 groups: apoE (-/-), apoE (-/-) with PIO, diabetic apoE (-/-), and diabetic apoE (-/-) with PIO. Wild type male C57BL/6 mice were used as controls (n = 8 per group). After 8 weeks of PIO treatment, we examined the baseline characteristics and metabolic parameters of each group, and we used enzyme-linked immunosorbent assay (ELISA), western blot, and immunohistochemical staining to evaluate the expression levels of advanced glycation end products (AGEs), receptor for advanced glycation end products (RAGE), NLRP3, nuclear factor—kappa B (NF-κB), caspase-1, interleukin (IL)-18, and IL-1β in each group. Results Compared to the diabetic apoE (-/-) group, PIO treatment decreased blood glucose, cholesterol, serum blood urea nitrogen (BUN), and creatinine levels. It also depressed the glomerular mesangial expansion. PIO down-regulated expression of AGEs, RAGE, and NF-κB, all of which further depressed NLRP3, caspase-1, IL-18, and IL-1β levels. Conclusion Pioglitazone can ameliorate diabetic renal damage, and this effect is related to the inhibition of renal AGE/RAGE axis activation and the down-regulation of NF-κB expression. These effects lead to a decline in NLRP3 levels and downstream secretion of inflammatory cytokines.
Collapse
Affiliation(s)
- Yao Wang
- Department of nephrology, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
| | - Bo Yu
- Department of emergency, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
| | - Li Wang
- Department of cardiology, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
| | - Ming Yang
- Department of nephrology, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhiyin Xia
- Department of nephrology, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei Wei
- Department of nephrology, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
| | - Fengyu Zhang
- Central lab, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaochen Yuan
- Department of cardiology, the affiliated hospital of Yangzhou University (Yangzhou NO.1 people’s hospital), Yangzhou University, Yangzhou, Jiangsu, China
- * E-mail:
| |
Collapse
|
42
|
Sekijima M, Sahara H, Miki K, Villani V, Ariyoshi Y, Iwanaga T, Tomita Y, Yamada K. Hydrogen sulfide prevents renal ischemia-reperfusion injury in CLAWN miniature swine. J Surg Res 2017; 219:165-172. [PMID: 29078877 DOI: 10.1016/j.jss.2017.05.123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/17/2017] [Accepted: 05/25/2017] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) has recently been reported to demonstrate both antiinflammatory and cytoprotective effects; however, its efficacy has not been well documented in large animal models. In this study, we examined whether the administration of H2S offers cytoprotective effects on renal ischemia-reperfusion injury (IRI) in a preclinical miniature swine model. METHODS Major histocompatibility complex-inbred, CLAWN miniature swine (n = 9) underwent a right nephrectomy, followed by induction of a 120-min period of warm ischemia via placement of clamps on the left renal artery and vein. Group 1 (n = 3) underwent renal ischemia without H2S administration. Groups 2 (n = 3) and 3 (n = 3) received Na2S (prodrug of H2S) 10 min before reperfusion of the ischemic kidneys followed by a 30-min of Na2S postreperfusion intravenously (group 2) or selective administration of Na2S via the left renal artery (group 3). IRI was assessed by kidney biopsies, levels of inflammatory cytokines in sera and kidney tissue. RESULTS Animals in group 1 had significantly higher serum creatinine levels compared with animals in groups 2 and 3 (P < 0.01). Histology showed severe tubular damage with TUNEL-positive cells in group 1 on postoperative day 2 compared with mild damage in group 2 and minimal damage in group 3. Furthermore, levels of inflammatory cytokines in both serum (interleukin-6 [IL-6], tumor necrosis factor-α, and high-mobility group box 1) and renal tissue (IL-1 and IL-6) in group 3 were markedly lower than in group 2, suggesting beneficial effects of selective Na2S administration. CONCLUSIONS Na2S administration, especially via an organ selective approach, appears to potentially offer cytoprotective and antiinflammatory effects following renal IRI.
Collapse
Affiliation(s)
- Mitsuhiro Sekijima
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Hisashi Sahara
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Katsuyuki Miki
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan; The 3rd Department of the Surgery, Kidney Center, Tokyo Women's Medical University, Tokyo, Japan
| | - Vincenzo Villani
- Transplantation Biology Research Center Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts
| | - Yuichi Ariyoshi
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Takehiro Iwanaga
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Yusuke Tomita
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Kazuhiko Yamada
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
43
|
DeWolf SE, Shigeoka AA, Scheinok A, Kasimsetty SG, Welch AK, McKay DB. Expression of TLR2, NOD1, and NOD2 and the NLRP3 Inflammasome in Renal Tubular Epithelial Cells of Male versus Female Mice. Nephron Clin Pract 2017; 137:68-76. [PMID: 28614830 DOI: 10.1159/000456016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/03/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Gender-biased outcomes are associated with acute kidney injury (AKI) and human and animal studies have shown that females are preferentially protected from renal ischemia. However, the reason for this is not known. One clue might lie with pattern recognition receptors (PRRs), which are triggers of ischemic injury when ligated by molecules in the ischemic milieu. Several PRR families are expressed by renal tubular epithelial cells (RTEs) and incite cell death signaling and production of pro-inflammatory molecules. Blockade of specific PRRs (e.g., TLR2, NOD1, NOD2, and NLRP3) provides highly significant protection from ischemic RTE injury. As a first step to understand gender-biased outcomes of AKI, we tested whether constitutive gender-based differences exist in expression of these PRRS in RTEs. METHODS To determine whether PRR expression differences exist, primary RTEs isolated from male and female WT kidneys were examined by FACS, qPCR, and Western Blot for expression of TLR2, NOD1, NOD2, and NLRP3 inflammasome components. RESULTS No RTE gender-based differences in TLR2, NOD1, NOD2, NLRP3, or ASC were found. RTEs from female kidneys had approximately half the mRNA, but the same protein concentration of pro-caspase-1 compared to RTEs isolated from male kidneys. CONCLUSIONS Our findings indicate that intrinsic gender differences in RTE expression of TLR2, NOD1, NOD2, NLRP3, and ASC are not responsible for the gender-biased outcomes observed in ischemia/reperfusion injury. The lower caspase-1 mRNA expression in RTEs from females warrants further exploration of additional upstream signals that might differentially regulate caspase-1 in male vs. female RTEs.
Collapse
Affiliation(s)
- Sean E DeWolf
- Division of Nephrology/Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | |
Collapse
|
44
|
Pichler R, Afkarian M, Dieter BP, Tuttle KR. Immunity and inflammation in diabetic kidney disease: translating mechanisms to biomarkers and treatment targets. Am J Physiol Renal Physiol 2017; 312:F716-F731. [PMID: 27558558 PMCID: PMC6109808 DOI: 10.1152/ajprenal.00314.2016] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/16/2016] [Indexed: 01/10/2023] Open
Abstract
Increasing incidences of obesity and diabetes have made diabetic kidney disease (DKD) the leading cause of chronic kidney disease and end-stage renal disease worldwide. Despite current pharmacological treatments, including strategies for optimizing glycemic control and inhibitors of the renin-angiotensin system, DKD still makes up almost one-half of all cases of end-stage renal disease in the United States. Compelling and mounting evidence has clearly demonstrated that immunity and inflammation play a paramount role in the pathogenesis of DKD. This article reviews the involvement of the immune system in DKD and identifies important roles of key immune and inflammatory mediators. One of the most recently identified biomarkers is serum amyloid A, which appears to be relatively specific for DKD. Novel and evolving treatment approaches target protein kinases, transcription factors, chemokines, adhesion molecules, growth factors, advanced glycation end-products, and other inflammatory molecules. This is the beginning of a new era in the understanding and treatment of DKD, and we may have finally reached a tipping point in our fight against the growing burden of DKD.
Collapse
Affiliation(s)
- Raimund Pichler
- Division of Nephrology, University of Washington, Seattle, Washington;
| | - Maryam Afkarian
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington; and
| | - Brad P Dieter
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington; and
- Providence Health Care, Spokane, Washington
| | - Katherine R Tuttle
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington; and
- Providence Health Care, Spokane, Washington
| |
Collapse
|
45
|
Beckerman P, Bi-Karchin J, Park ASD, Qiu C, Dummer PD, Soomro I, Boustany-Kari CM, Pullen SS, Miner JH, Hu CAA, Rohacs T, Inoue K, Ishibe S, Saleem MA, Palmer MB, Cuervo AM, Kopp JB, Susztak K. Transgenic expression of human APOL1 risk variants in podocytes induces kidney disease in mice. Nat Med 2017; 23:429-438. [PMID: 28218918 DOI: 10.1038/nm.4287] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
African Americans have a heightened risk of developing chronic and end-stage kidney disease, an association that is largely attributed to two common genetic variants, termed G1 and G2, in the APOL1 gene. Direct evidence demonstrating that these APOL1 risk alleles are pathogenic is still lacking because the APOL1 gene is present in only some primates and humans; thus it has been challenging to demonstrate experimental proof of causality of these risk alleles for renal disease. Here we generated mice with podocyte-specific inducible expression of the APOL1 reference allele (termed G0) or each of the risk-conferring alleles (G1 or G2). We show that mice with podocyte-specific expression of either APOL1 risk allele, but not of the G0 allele, develop functional (albuminuria and azotemia), structural (foot-process effacement and glomerulosclerosis) and molecular (gene-expression) changes that closely resemble human kidney disease. Disease development was cell-type specific and likely reversible, and the severity correlated with the level of expression of the risk allele. We further found that expression of the risk-variant APOL1 alleles interferes with endosomal trafficking and blocks autophagic flux, which ultimately leads to inflammatory-mediated podocyte death and glomerular scarring. In summary, this is the first demonstration that the expression of APOL1 risk alleles is causal for altered podocyte function and glomerular disease in vivo.
Collapse
Affiliation(s)
- Pazit Beckerman
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jing Bi-Karchin
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ae Seo Deok Park
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chengxiang Qiu
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patrick D Dummer
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Irfana Soomro
- Division of Nephrology, New York University, New York, New York, USA
| | - Carine M Boustany-Kari
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | - Jeffrey H Miner
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chien-An A Hu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine and Health Sciences Center, Albuquerque, New Mexico, USA
| | - Tibor Rohacs
- Department of Pharmacology, Physiology &Neuroscience, Rutgers-New Jersey Medical School, Newark, New Jersey, USA
| | - Kazunori Inoue
- Division of Nephrology, Yale University, School of Medicine, New Haven, Connecticut, USA
| | - Shuta Ishibe
- Division of Nephrology, Yale University, School of Medicine, New Haven, Connecticut, USA
| | - Moin A Saleem
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Matthew B Palmer
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, USA
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
46
|
Chen L, Lan Z. Polydatin attenuates potassium oxonate-induced hyperuricemia and kidney inflammation by inhibiting NF-κB/NLRP3 inflammasome activation via the AMPK/SIRT1 pathway. Food Funct 2017; 8:1785-1792. [DOI: 10.1039/c6fo01561a] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study was designed to investigate the effects of polydatin (PLD) on potassium oxonate-induced hyperuricemic rats.
Collapse
Affiliation(s)
- Lvyi Chen
- School of Pharmaceutical Sciences
- South-Central University for Nationalities
- Wuhan 430074
- PR China
| | - Zhou Lan
- School of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan 430065
- PR China
| |
Collapse
|
47
|
Influence of the Expression of Inflammatory Markers on Kidney after Fetal Programming in an Experimental Model of Renal Failure. J Immunol Res 2016; 2016:9151607. [PMID: 28018922 PMCID: PMC5149704 DOI: 10.1155/2016/9151607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/25/2016] [Accepted: 11/08/2016] [Indexed: 11/30/2022] Open
Abstract
Objective. To evaluate the expression of inflammatory markers in experimental renal failure after fetal programming. Methods. The offspring aged two and five months were divided into four groups: CC (control dams, control offspring); DC (diabetic dams, control offspring); CFA (control dams, folic acid offspring, 250 mg/Kg); and DFA (diabetic dams, folic acid offspring). Gene expression of inflammatory markers MCP-1, IL-1, NOS3, TGF-β, TNF-α, and VEGF was evaluated by RT-PCR. Results. MCP-1 was increased in the CFA and DFA groups at two and five months of age, as well as in DC5 when compared to CC5. There was a higher expression of IL-1 in the CFA2, DFA2, and DC2 groups. There was a decrease in NOS3 and an increase in TNF-α in DFA5 in relation to CFA5. The gene expression of TGF-β increased in cases that had received folic acid at two and five months, and VEGF decreased in the CFA5 and DFA5 groups. DC5 showed increased VEGF expression in comparison with CC5. Conclusions. Gestational diabetes mellitus and folic acid both change the expression of inflammatory markers, thus demonstrating that the exposure to harmful agents in adulthood has a more severe impact in cases which underwent fetal reprogramming.
Collapse
|
48
|
Szeto HH, Liu S, Soong Y, Seshan SV, Cohen-Gould L, Manichev V, Feldman LC, Gustafsson T. Mitochondria Protection after Acute Ischemia Prevents Prolonged Upregulation of IL-1 β and IL-18 and Arrests CKD. J Am Soc Nephrol 2016; 28:1437-1449. [PMID: 27881606 DOI: 10.1681/asn.2016070761] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/25/2016] [Indexed: 11/03/2022] Open
Abstract
The innate immune system has been implicated in both AKI and CKD. Damaged mitochondria release danger molecules, such as reactive oxygen species, DNA, and cardiolipin, which can cause NLRP3 inflammasome activation and upregulation of IL-18 and IL-1β It is not known if mitochondrial damage persists long after ischemia to sustain chronic inflammasome activation. We conducted a 9-month study in Sprague-Dawley rats after 45 minutes of bilateral renal ischemia. We detected glomerular and peritubular capillary rarefaction, macrophage infiltration, and fibrosis at 1 month. Transmission electron microscopy revealed mitochondrial degeneration, mitophagy, and deformed foot processes in podocytes. These changes progressed over the study period, with a persistent increase in renal cortical expression of IL-18, IL-1β, and TGF-β, despite a gradual decline in TNF-α expression and macrophage infiltration. Treatment with a mitoprotective agent (SS-31; elamipretide) for 6 weeks, starting 1 month after ischemia, preserved mitochondrial integrity, ameliorated expression levels of all inflammatory markers, restored glomerular capillaries and podocyte structure, and arrested glomerulosclerosis and interstitial fibrosis. Further, helium ion microscopy vividly demonstrated the restoration of podocyte structure by SS-31. The protection by SS-31 was sustained for ≥6 months after treatment ended, with normalization of IL-18 and IL-1β expression. These results support a role for mitochondrial damage in inflammasome activation and CKD and suggest mitochondrial protection as a novel therapeutic approach that can arrest the progression of CKD. Notably, SS-31 is effective when given long after AKI and provides persistent protection after termination of drug treatment.
Collapse
Affiliation(s)
- Hazel H Szeto
- Department of Pharmacology, .,Research Program in Mitochondrial Therapeutics
| | - Shaoyi Liu
- Department of Pharmacology.,Research Program in Mitochondrial Therapeutics
| | - Yi Soong
- Department of Pharmacology.,Research Program in Mitochondrial Therapeutics
| | | | - Leona Cohen-Gould
- Department of Biochemistry, Weill Cornell Medical College, New York, New York; and
| | - Viacheslav Manichev
- Institute of Advanced Materials, Devices, and Nanotechnology, and.,Institute of Advanced Materials, Devices, and Nanotechnology, and
| | - Leonard C Feldman
- Institute of Advanced Materials, Devices, and Nanotechnology, and.,Department of Physics and Astronomy, Rutgers University, Piscataway, New Jersey
| | - Torgny Gustafsson
- Institute of Advanced Materials, Devices, and Nanotechnology, and.,Department of Physics and Astronomy, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
49
|
de Almeida RMC, Clendenon SG, Richards WG, Boedigheimer M, Damore M, Rossetti S, Harris PC, Herbert BS, Xu WM, Wandinger-Ness A, Ward HH, Glazier JA, Bacallao RL. Transcriptome analysis reveals manifold mechanisms of cyst development in ADPKD. Hum Genomics 2016; 10:37. [PMID: 27871310 PMCID: PMC5117508 DOI: 10.1186/s40246-016-0095-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/04/2016] [Indexed: 12/18/2022] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) causes progressive loss of renal function in adults as a consequence of the accumulation of cysts. ADPKD is the most common genetic cause of end-stage renal disease. Mutations in polycystin-1 occur in 87% of cases of ADPKD and mutations in polycystin-2 are found in 12% of ADPKD patients. The complexity of ADPKD has hampered efforts to identify the mechanisms underlying its pathogenesis. No current FDA (Federal Drug Administration)-approved therapies ameliorate ADPKD progression. Results We used the de Almeida laboratory’s sensitive new transcriptogram method for whole-genome gene expression data analysis to analyze microarray data from cell lines developed from cell isolates of normal kidney and of both non-cystic nephrons and cysts from the kidney of a patient with ADPKD. We compared results obtained using standard Ingenuity Volcano plot analysis, Gene Set Enrichment Analysis (GSEA) and transcriptogram analysis. Transcriptogram analysis confirmed the findings of Ingenuity, GSEA, and published analysis of ADPKD kidney data and also identified multiple new expression changes in KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways related to cell growth, cell death, genetic information processing, nucleotide metabolism, signal transduction, immune response, response to stimulus, cellular processes, ion homeostasis and transport and cofactors, vitamins, amino acids, energy, carbohydrates, drugs, lipids, and glycans. Transcriptogram analysis also provides significance metrics which allow us to prioritize further study of these pathways. Conclusions Transcriptogram analysis identifies novel pathways altered in ADPKD, providing new avenues to identify both ADPKD’s mechanisms of pathogenesis and pharmaceutical targets to ameliorate the progression of the disease. Electronic supplementary material The online version of this article (doi:10.1186/s40246-016-0095-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rita M C de Almeida
- Biocomplexity Institute and Department of Physics, Indiana University, Bloomington, IN, 47405, USA.,Instituto de Física and Instituto Nacional de Ciência e Tecnologia, Universidade Federal do Rio Grande do Sul, 91501-970, Porto Alegre, RS, Brazil
| | - Sherry G Clendenon
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | | | | | - Michael Damore
- AMGEN Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320-1799, USA
| | - Sandro Rossetti
- Division of Nephrology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Peter C Harris
- Division of Nephrology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Britney-Shea Herbert
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Wei Min Xu
- Division of Nephrology, Department of Medicine, Richard Roudebush VAMC and Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Angela Wandinger-Ness
- Department of Pathology MSC08-4640 and Cancer Research and Treatment Center, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Heather H Ward
- Division of Nephrology, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - James A Glazier
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 47405, USA
| | - Robert L Bacallao
- Division of Nephrology, Department of Medicine, Richard Roudebush VAMC and Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
50
|
Hongyan L, Suling W, Weina Z, Yajie Z, Jie R. Antihyperuricemic effect of liquiritigenin in potassium oxonate-induced hyperuricemic rats. Biomed Pharmacother 2016; 84:1930-1936. [PMID: 27863839 DOI: 10.1016/j.biopha.2016.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/25/2016] [Accepted: 11/01/2016] [Indexed: 11/15/2022] Open
Abstract
The aim is to investigate the anti-hyperuricemic and renal protective effects of liquiritigenin in potassium oxonate-induced hyperuricemic rats. Hyperuricemia in rats was induced were induced with potassium oxonate (250mg/kg) intragastrically for 7 days, and liquiritigenin (20, 40mg/kg) and allopurinol (5mg/kg) were daily administrated to the rats orally 1h after the potassium oxonate exposure. Liquiritigenin significantly reversed the elevated productions of uric acid in serum and urine and pro-inflammation cytokines in serum and kidney, which shown that liquiritigenin has renal protective effects. Histological study shows that liquiritigenin inhibited severe necrosis and inflammatory cell infiltration in potassium oxonate-treated rats. Furthermore, liquiritigenin mediated the activities of aquaporins 4 (AQP4), and regulated the activation of NF-κB p65 and the degradation of IκBα. Finally, significant increases of nod-like receptor protein 3 (NLRP3) inflammasome, apoptosis-associated speck-like protein adaptor (ASC) adaptor and cleaved caspased-1 were restored by liquiritigenin. Therefore, liquiritigenin might improve renal inflammation by suppressing renal AQP4/NF-κB/IκBα and NLRP3 inflammasome activation in hyperuricemic rats.
Collapse
Affiliation(s)
- Long Hongyan
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001, China
| | - Wu Suling
- Department of Rheumatology, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001, China.
| | - Zhu Weina
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001, China
| | - Zhang Yajie
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001, China
| | - Ruan Jie
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001, China
| |
Collapse
|