501
|
Sun X, Meng H, Wan W, Xie M, Wen C. Application potential of stem/progenitor cell-derived extracellular vesicles in renal diseases. Stem Cell Res Ther 2019; 10:8. [PMID: 30616603 PMCID: PMC6323814 DOI: 10.1186/s13287-018-1097-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are nanometer-sized and membrane-bound vesicles, including exosomes and microvesicles. EVs can deliver bioactive macromolecules such as proteins, lipids, and nucleic acids, allowing intercellular communication in multicellular organisms. EVs are secreted by all cell types including stem/progenitor cells. Stem/progenitor cell-derived EVs have been identified to exert immunomodulatory effects on target cells through transferring protein molecules as well as regulatory effects on the phenotype of target cells through fusion with the target cells membrane and/or through direct endocytosis by target cells to transfer nucleic acid substances (such as mRNA, miRNA) to the target cells. In both human and animal models, the use of stem/progenitor cells (such as bone marrow mesenchymal stromal cells) has been shown to promote the recovery of kidney diseases such as acute kidney injury and chronic kidney disease. Stem/progenitor cell-derived extracellular vesicles are an important mechanism by which stem/progenitor cells might repair kidney injury. Here, this review will discuss the latest advances concerning the application potential of stem/progenitor cell-derived extracellular vesicles in renal diseases, including the aspects as follows: anti-inflammatory, proliferation-promoting and anti-apoptotic, proangiogenic, antifibrotic and renal cancer progression-promoting. Therefore, stem/progenitor cell-derived extracellular vesicles may be a promising treatment tool for renal diseases.
Collapse
Affiliation(s)
- Xiao Sun
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, No.139,Renmin road, Changsha, Hunan, People's Republic of China
| | - Huanyu Meng
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, No.139,Renmin road, Changsha, Hunan, People's Republic of China
| | - Wuqing Wan
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, No.139,Renmin road, Changsha, Hunan, People's Republic of China
| | - Min Xie
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, No.139,Renmin road, Changsha, Hunan, People's Republic of China
| | - Chuan Wen
- Division of Hematology and Tumor, Children's Medical Center, The Second Xiangya Hospital, Central South University, No.139,Renmin road, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
502
|
Abstract
Acute kidney injury (AKI) is a frequent complication in hospitalised patients and is diagnosed by urinary output and serum creatinine. Serum creatinine is an indirect marker for renal glomerular filtration, but lacks specificity for damage to kidney tissue and the relatively late response to injury precludes early recognition of AKI. Timely diagnosis of kidney injury using biomarkers that provide information about the aetiology of kidney injury is an unmet clinical need. To overcome the suboptimal performance of serum creatinine, injury biomarkers have been proposed that predict AKI in diverse clinical settings. The clinical performance of these markers is considered moderate due to the lack of specificity for kidney tissue or the underlying injury mechanisms, poor test specificity and confounding by interventions or comorbidities. Hence, it is not unequivocally beneficial to implement current kidney injury biomarkers in the clinical laboratory for diagnostic purposes. In this article we review biomarkers that might fulfil AKI-related unmet clinical needs in the academic hospital setting.
Collapse
|
503
|
How Acute Kidney Injury Contributes to Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:117-142. [PMID: 31399964 DOI: 10.1007/978-981-13-8871-2_7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is a widespread clinical syndrome directly associated with patient short-term and long-term morbidity and mortality. During the last decade, the incidence rate of AKI has been increasing, the repeated and severe episodes of AKI have been recognized as a major risk factor chronic kidney diseases (CKD) and end-stage kidney disease (ESRD) leading to global disease burden. Proposed pathological processes and risk factors that add to the transition of AKI to CKD and ESRD include severity and frequency of kidney injury, older age, gender, genetics and chronic health conditions like diabetes, hypertension, and obesity. Therefore, there is a great interest in learning about the mechanism of AKI leading to renal fibrosis, the ultimate renal lesions of CKD. Over the last several years, a significant attention has been given to the field of renal fibrosis with impressive progression in knowing the mechanism of renal fibrosis to detailed cellular characterization and molecular pathways implicated in tubulointerstitial fibrosis. Research and clinical trial are underway for emerging biomarkers detecting early kidney injury, predicting kidney disease progression and developing strategies to efficiently treat AKI and to minimize AKI progression to CKD and ESRD. Specific interventions to prevent renal fibrosis are still experimental. Potential therapeutic advances based on those molecular mechanisms will hopefully offer promising insights into the development of new therapeutic interventions for patients in the near future.
Collapse
|
504
|
Li C, Xie N, Li Y, Liu C, Hou FF, Wang J. N-acetylcysteine ameliorates cisplatin-induced renal senescence and renal interstitial fibrosis through sirtuin1 activation and p53 deacetylation. Free Radic Biol Med 2019; 130:512-527. [PMID: 30447351 DOI: 10.1016/j.freeradbiomed.2018.11.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/30/2018] [Accepted: 11/07/2018] [Indexed: 12/19/2022]
Abstract
The mechanism underlying the development of chronic kidney disease (CKD) after acute kidney injury (AKI) remains unclear. Maladaptive repair has been considered an important mechanism of CKD post AKI. Renal tubular cells under maladaptive repair have characteristics of premature senescence. These premature senescent cells can generate profibrotic factors that promote organ fibrosis. The purpose of this study was to investigate whether cisplatin induces premature renal senescence and the role of premature renal senescence in the progression of CKD post AKI. As oxidative stress is a major cause of senescence, we further evaluated whether antioxidant therapy could protect renal tubular cells from cisplatin-induced premature senescence and retard the progression of CKD post AKI. The molecular mechanism of this protection was also investigated. We found that cisplatin induced premature renal senescence in vitro and in vivo. In a multiple-cisplatin-treatment murine model, renal interstitial fibrosis was accompanied by premature renal senescence. N-acetylcysteine (NAC), an antioxidant, attenuated premature senescence and decreased renal fibrosis, and its effects were dependent on sirtuin1 (SIRT1) activation and p53 deacetylation. These results indicate that cisplatin can induce premature renal senescence, which is associated with the development of CKD post cisplatin-induced AKI. SIRT1 activation and p53 deacetylation might be identified as potential targets for attenuating premature renal senescence and retarding the progression of CKD post AKI.
Collapse
Affiliation(s)
- Caizhen Li
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Na Xie
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Yan Li
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Chongbin Liu
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Fan Fan Hou
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| | - Jun Wang
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
505
|
Effects of Fluid Bolus Therapy on Renal Perfusion, Oxygenation, and Function in Early Experimental Septic Kidney Injury. Crit Care Med 2019; 47:e36-e43. [DOI: 10.1097/ccm.0000000000003507] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
506
|
Oh TK, Song IA, Cho YJ, Lim C, Jeon YT, Bae HJ, Jo YH. Preadmission Statin Therapy Is Associated with a Lower Incidence of Acute Kidney Injury in Critically Ill Patients: A Retrospective Observational Study. J Clin Med 2018; 8:jcm8010025. [PMID: 30585236 PMCID: PMC6351906 DOI: 10.3390/jcm8010025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate the association between preadmission statin use and acute kidney injury (AKI) incidence among critically ill patients who needed admission to the intensive care unit (ICU) for medical care. Medical records of patients admitted to the ICU were reviewed. Patients who continuously took statin for >1 month prior to ICU admission were defined as statin users. We investigated whether preadmission statin use was associated with AKI incidence within 72 h after ICU admission and whether the association differs according to preadmission estimated glomerular filtration rate (eGFR; in mL min−1 1.73 m−2). Among 21,236 patients examined, 5756 (27.1%) were preadmission statin users and 15,480 (72.9%) were non-statin users. Total AKI incidence within 72 h after ICU admission was 31% lower in preadmission statin users than in non-statin users [odds ratio (OR), 0.69; 95% confidence interval (CI), 0.61–0.79; p < 0.001]. This association was insignificant among individuals with eGFR <30 mL min−1 1.73 m−2 (p > 0.05). Our results suggested that preadmission statin therapy is associated with a lower incidence of AKI among critically ill patients; however, this effect might not be applicable for patients with eGFR <30 mL min−1 1.73 m−2.
Collapse
Affiliation(s)
- Tak Kyu Oh
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea.
| | - In-Ae Song
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea.
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea.
| | - Cheong Lim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea.
| | - Young-Tae Jeon
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea.
| | - Hee-Joon Bae
- Department of Neurology, Stroke Center, Seoul National University Bundang Hospital, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea.
| | - You Hwan Jo
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea.
| |
Collapse
|
507
|
Corremans R, Vervaet BA, D'Haese PC, Neven E, Verhulst A. Metformin: A Candidate Drug for Renal Diseases. Int J Mol Sci 2018; 20:E42. [PMID: 30583483 PMCID: PMC6337137 DOI: 10.3390/ijms20010042] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Over the past decades metformin has been the optimal first-line treatment for type 2 diabetes mellitus (T2DM). Only in the last few years, it has become increasingly clear that metformin exerts benign pleiotropic actions beyond its prescribed use and ongoing investigations focus on a putative beneficial impact of metformin on the kidney. Both acute kidney injury (AKI) and chronic kidney disease (CKD), two major renal health issues, often result in the need for renal replacement therapy (dialysis or transplantation) with a high socio-economic impact for the patients. Unfortunately, to date, effective treatment directly targeting the kidney is lacking. Metformin has been shown to exert beneficial effects on the kidney in various clinical trials and experimental studies performed in divergent rodent models representing different types of renal diseases going from AKI to CKD. Despite growing evidence on metformin as a candidate drug for renal diseases, in-depth research is imperative to unravel the molecular signaling pathways responsible for metformin's renoprotective actions. This review will discuss the current state-of-the-art literature on clinical and preclinical data, and put forward potential cellular mechanisms and molecular pathways by which metformin ameliorates AKI/CKD.
Collapse
Affiliation(s)
- Raphaëlle Corremans
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Benjamin A Vervaet
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| |
Collapse
|
508
|
Araújo NC, Suassuna JHR. Time-intensity curve analysis of contrast-enhanced ultrasound is unable to differentiate renal dysfunction in the early post-transplant period - a prospective study. BMC Nephrol 2018; 19:351. [PMID: 30537946 PMCID: PMC6288962 DOI: 10.1186/s12882-018-1158-0] [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] [Received: 09/07/2018] [Accepted: 11/28/2018] [Indexed: 12/14/2022] Open
Abstract
Background Contrast enhanced ultrasonography (CEUS) assessment of kidney allografts mainly focuses on graft rejection. However, studies on delayed graft function (DGF) without acute rejection are still lacking. The aim of this study was to build a time-intensity curve (TIC) using CEUS in non-immunological DGF to understand the utility of CEUS in early transplantation. Methods Twenty-eight patients in the short-term postoperative period (<14 days) were divided according to the need for dialysis (early graft function [EGF] and [DGF]) and 37 subjects with longer than 90 days follow-up were divided into creatinine tertiles. Time to peak [TTP] and rising time [RT were compared between groups. Results EGF and DGF were similar, except for creatinine. In comparison to the late group, medullary TTP and RT were shorter in the early group as well as the delay regarding contrast arrival in the medulla (in relation to cortex) and reaching the medullary peak (in relation to artery and cortex). In the late group, patients with renal dysfunction showed shorter temporal difference to reach medullary peak in relation to artery and cortex. Conclusions Although it was not possible to differentiate EGF and DGF using TIC, differences between early and late groups point to blood shunting in renal dysfunction.
Collapse
Affiliation(s)
- Nordeval Cavalcante Araújo
- University of the State of Rio de Janeiro, Rio de Janeiro, Brasil. .,Nephrology Section, Hospital Universitário Pedro Ernesto, Boulevard 28 de Setembro, 77 Vila Isabel, 20551-030, Rio de Janeiro, RJ, Brasil.
| | | |
Collapse
|
509
|
Tan X, Zhu H, Tao Q, Guo L, Jiang T, Xu L, Yang R, Wei X, Wu J, Li X, Zhang JS. FGF10 Protects Against Renal Ischemia/Reperfusion Injury by Regulating Autophagy and Inflammatory Signaling. Front Genet 2018; 9:556. [PMID: 30532765 PMCID: PMC6265307 DOI: 10.3389/fgene.2018.00556] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Ischemia-reperfusion (I/R) is a common cause of acute kidney injury (AKI), which is associated with high mortality and poor outcomes. Autophagy plays important roles in the homeostasis of renal tubular cells (RTCs) and is implicated in the pathogenesis of AKI, although its role in the process is complex and controversial. Fibroblast growth factor 10 (FGF10), a multifunctional FGF family member, was reported to exert protective effect against cerebral ischemia injury and myocardial damage. Whether FGF10 has similar beneficial effect, and if so whether autophagy is associated with the potential protective activity against AKI has not been investigated. Herein, we report that FGF10 treatment improved renal function and histological integrity in a rat model of renal I/R injury. We observed that FGF10 efficiently reduced I/R-induced elevation in blood urea nitrogen, serum creatinine as well as apoptosis induction of RTCs. Interestingly, autophagy activation following I/R was suppressed by FGF10 treatment based on the immunohistochemistry staining and immunoblot analyses of LC3, Beclin-1 and SQSTM1/p62. Moreover, combined treatment of FGF10 with Rapamycin partially reversed the renoprotective effect of FGF10 suggesting the involvement of mTOR pathway in the process. Interestingly, FGF10 also inhibited the release of HMGB1 from the nucleus to the extracellular domain and regulated the expression of inflammatory cytokines such as TNF-α, IL-1β and IL-6. Together, these results indicate that FGF10 could alleviate kidney I/R injury by suppressing excessive autophagy and inhibiting inflammatory response and may therefore have the potential to be used for the prevention and perhaps treatment of I/R-associated AKI.
Collapse
Affiliation(s)
- Xiaohua Tan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Qingdao University Medical College, Qingdao, China
| | - Hongmei Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qianyu Tao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lisha Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Tianfang Jiang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Le Xu
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ruo Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiayu Wei
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Jin Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Jin-San Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,Institute of Life Sciences, Wenzhou University, Wenzhou, China
| |
Collapse
|
510
|
El Menyiy N, Al-Waili N, El Ghouizi A, Al-Waili W, Lyoussi B. Evaluation of antiproteinuric and hepato-renal protective activities of propolis in paracetamol toxicity in rats. Nutr Res Pract 2018; 12:535-540. [PMID: 30515282 PMCID: PMC6277308 DOI: 10.4162/nrp.2018.12.6.535] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 09/05/2018] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND/OBJECTIVES Propolis has a rich source of bioactive compounds and has renal and hepatic protective properties. The purpose of this study was to investigate the beneficial effect of hydro-ethanolic extract of propolis against paracetamol-induced liver damage and impairment of kidney function, as well as hematological changes in rats. MATERIALS AND METHODS Six groups of rats were used; the first group was served as a control; the second and third groups were treated by propolis extract at a dose of 50 and 100 mg/kg.B.WT. respectively; the fourth group was treated by paracetamol (200 mg/kg.B.WT.); the fifth group was treated by propolis (50 mg/kg.B.WT.) for eight days and then received similar dose of propolis for following seven days with paracetamol at a dose of 200 mg/kg.B.WT. daily for the seven days; and the sixth group was treated with propolis (100 mg/kg.B.WT.) for eight days and then received similar dose of propolis for following seven days with paracetamol at a dose of 200 mg/kg.B.WT. daily for the seven days. All the animals were treated for a period of 15 days. At the end of the experimental period, blood samples were collected for measurement of the liver enzymes, serum albumin, protein and creatinine, blood urea nitrogen, hematological parameters, and urine volume, protein and albumin. RESULTS Paracetamol over dose significantly lowered hemoglobin, serum total protein, albumin, and uric acid, while it significantly increased blood creatinine, blood urea nitrogen, alanine aminotransferase, aspartate aminotransferase and lactate dehydrogenase activities, white blood cells, and platelet count as compared to the control. However, these alterations were significantly attenuated by the use of propolis extract and the effect was dose dependent. Interestingly, propolis prevented paracetamol induced proteinuria, low hemoglobin and body weight loss. CONCLUSIONS Propolis significantly prevented paracetamol induced renal, hepatic and hematological toxicity and might be useful in the management of liver and renal diseases particularly proteinuria.
Collapse
Affiliation(s)
- Nawal El Menyiy
- Laboratory Physiology-Pharmacology & Environmental Health, Faculty of Sciences Dhar El Mehraz, University Sidi Mohamed Ben Abdallah, Fez 30000, Morocco
| | - Noori Al-Waili
- New York Medical Care for Nephrology, 87-40 134 Street, Richmond Hill, New York City, NY 11418, USA
| | - Asmae El Ghouizi
- Laboratory Physiology-Pharmacology & Environmental Health, Faculty of Sciences Dhar El Mehraz, University Sidi Mohamed Ben Abdallah, Fez 30000, Morocco
| | - Wail Al-Waili
- New York Medical Care for Nephrology, 87-40 134 Street, Richmond Hill, New York City, NY 11418, USA
| | - Badiaa Lyoussi
- Laboratory Physiology-Pharmacology & Environmental Health, Faculty of Sciences Dhar El Mehraz, University Sidi Mohamed Ben Abdallah, Fez 30000, Morocco
| |
Collapse
|
511
|
Li Y, Hou D, Chen X, Zhu J, Zhang R, Sun W, Li P, Tian Y, Kong X. Hydralazine protects against renal ischemia-reperfusion injury in rats. Eur J Pharmacol 2018; 843:199-209. [PMID: 30472201 DOI: 10.1016/j.ejphar.2018.11.015] [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] [Received: 06/23/2018] [Revised: 11/10/2018] [Accepted: 11/12/2018] [Indexed: 01/06/2023]
Abstract
In this study, we investigated whether hydralazine could reduce renal ischemia and reperfusion (I/R) injury in rats. Renal I/R was induced by a 70-min occlusion of the bilateral renal arteries and a 24-h reperfusion, which was confirmed by the increased the mortality, the levels of blood urea nitrogen (BUN), blood creatinine (Cr), renal tissue NO and the visible histological damage of the kidneys. Apoptosis was evaluated by terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling (TUNEL) staining. Furthermore, the serum levels of malonaldehyde (MDA), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) were significantly elevated in renal I/R group, while the superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) levels were suppressed. However, intragastric pretreatment with hydralazine at doses of 7.5-30 mg/kg before renal I/R significantly limited the increase in mortality, BUN, Cr, oxidative stress, inflammatory factors, histological damage and apoptosis in the kidneys. In addition, hydralazine also increased p-AKT, Bcl-2 expression and decreased iNOS, Bax, cleaved caspase-3 expression in the kidneys. In conclusion, hydralazine reduced renal I/R injury probably via inhibiting NO production by iNOS/NO pathway, inhibiting oxidative stress, inflammatory response and apoptosis by a mitochondrial-dependent pathway.
Collapse
Affiliation(s)
- Yong Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Daorong Hou
- Key Laboratory of the Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China
| | - Xuguan Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Jingfeng Zhu
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Ruyi Zhang
- Animal Laboratory, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Yunfan Tian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
512
|
Hurtado Del Pozo C, Garreta E, Izpisúa Belmonte JC, Montserrat N. Modeling epigenetic modifications in renal development and disease with organoids and genome editing. Dis Model Mech 2018; 11:dmm035048. [PMID: 30459215 PMCID: PMC6262817 DOI: 10.1242/dmm.035048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Understanding epigenetic mechanisms is crucial to our comprehension of gene regulation in development and disease. In the past decades, different studies have shown the role of epigenetic modifications and modifiers in renal disease, especially during its progression towards chronic and end-stage renal disease. Thus, the identification of genetic variation associated with chronic kidney disease has resulted in better clinical management of patients. Despite the importance of these findings, the translation of genotype-phenotype data into gene-based medicine in chronic kidney disease populations still lacks faithful cellular or animal models that recapitulate the key aspects of the human kidney. The latest advances in the field of stem cells have shown that it is possible to emulate kidney development and function with organoids derived from human pluripotent stem cells. These have successfully recapitulated not only kidney differentiation, but also the specific phenotypical traits related to kidney function. The combination of this methodology with CRISPR/Cas9 genome editing has already helped researchers to model different genetic kidney disorders. Nowadays, CRISPR/Cas9-based approaches also allow epigenetic modifications, and thus represent an unprecedented tool for the screening of genetic variants, epigenetic modifications or even changes in chromatin structure that are altered in renal disease. In this Review, we discuss these technical advances in kidney modeling, and offer an overview of the role of epigenetic regulation in kidney development and disease.
Collapse
Affiliation(s)
- Carmen Hurtado Del Pozo
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| | - Elena Garreta
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| | | | - Nuria Montserrat
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| |
Collapse
|
513
|
Activation of α7 Nicotinic Acetylcholine Receptor Ameliorates Zymosan-Induced Acute Kidney Injury in BALB/c Mice. Sci Rep 2018; 8:16814. [PMID: 30429582 PMCID: PMC6235888 DOI: 10.1038/s41598-018-35254-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/01/2018] [Indexed: 12/03/2022] Open
Abstract
Zymosan, a natural compound, provokes acute peritonitis and multiple organ dysfunction that affects the kidney, beside other organs via exaggerated inflammatory response. The aim of the present study is to test the role of cholinergic anti-inflammatory pathway (CAP) in alleviating acute kidney injury (AKI) induced by zymosan in BALB/c mice, using galantamine, a cholinesterase inhibitor, known to act via α7 nicotinic acetylcholine receptor (α7 nAChR) to stimulate CAP. Galantamine verified its anti-inflammatory effect by elevating acetylcholine (ACh) level, while abating the interleukin-6/ janus kinase 2 (Y1007/1008)/ signal transducer and activator of transcription 3 (Y705) (IL-6/ pY(1007/1008)-JAK2/ pY705-STAT3) inflammatory axis, with a consequent inhibition in suppressor of cytokine signaling 3 (SOCS3). This effect entails also the nuclear factor-kappa B (p65)/ high mobility group box protein-1/ (NF-κB (p65)/ HMGB-1) signaling pathway. Furthermore, the reno-curattive effect of galantamine was associated by a reduction in plasma creatinine (Cr), cystatin (Cys)-C, IL-18, and renal neutrophil gelatinase-associated lipocalin (NGAL), as well as an improved histopathological structure. Blocking the α7 nAChR by methyllycaconitine abolished the beneficial effect of galantamine to document the involvement of this receptor and the CAP in the amelioration of AKI induced by zymosan.
Collapse
|
514
|
SCUBE1-enhanced bone morphogenetic protein signaling protects against renal ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2018; 1865:329-338. [PMID: 30414502 DOI: 10.1016/j.bbadis.2018.10.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022]
Abstract
We previously reported that the membrane-bound SCUBE1 (signal peptide-CUB-epithelial growth factor domain-containing protein 1) forms a complex with bone morphogenetic protein 2 (BMP2) ligand and its receptors, thus acting as a BMP co-receptor to augment BMP signal activity. However, whether SCUBE1 can bind to and facilitate signaling activity of BMP7, a renal protective molecule for ischemia-reperfusion (I/R) insult, and contribute to the proliferation and repair of renal tubular cells after I/R remains largely unknown. In this study, we first showed that I/R-induced SCUBE1 was expressed in proximal tubular cells, which coincided with the expression of renoprotective BMP7. Molecular and biochemical analyses revealed that SCUBE1 directly binds to BMP7 and its receptors, functioning as a BMP co-receptor to promote BMP7 signaling. Furthermore, we used a new Scube1 deletion (Δ2) mouse strain to further elucidate the renal pathophysiological function of SCUBE1 after I/R injury. As compared with wild-type littermates, Δ2 mice showed severe renal histopathologic features (extensive loss of brush border, tubular necrosis, and tubular dilation) and increased inflammation (neutrophil infiltrate and induction of monocyte chemoattractant protein-1, tumor necrosis factor-α and interleukin-6) during the resolution of I/R damage. They also showed reduced BMP signaling (phosphorylated Smad1/5/8) along with decreased proliferation and increased apoptosis of renal tubular cells. Importantly, lentivirus-mediated overexpression of SCUBE1 enhanced BMP signaling and conferred a concomitant survival outcome for Δ2 proximal tubular epithelial cells after hypoxia-reoxygenation treatment. The protective BMP7 signaling may be facilitated by stress-inducible SCUBE1 after renal I/R, which suggests potential targeted approaches for acute kidney injury.
Collapse
|
515
|
Ali SN, Arif H, Khan AA, Mahmood R. Acute renal toxicity of sodium chlorate: Redox imbalance, enhanced DNA damage, metabolic alterations and inhibition of brush border membrane enzymes in rats. ENVIRONMENTAL TOXICOLOGY 2018; 33:1182-1194. [PMID: 30144278 DOI: 10.1002/tox.22624] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/13/2018] [Accepted: 06/17/2018] [Indexed: 06/08/2023]
Abstract
Sodium chlorate (NaClO3 ) is widely used in paper and pulp industries and as a non-selective herbicide. Humans can be exposed to NaClO3 through contaminated drinking water due to its improper and unchecked usage in industries and as herbicide. NaClO3 is also present as a major stable by-product in drinking water that has been disinfected with chlorine dioxide. In this study, we have investigated the effect of a single acute oral dose of NaClO3 on rat kidney. Adult male Wistar rats were divided into one control and four NaClO3 treated groups that were orally given different doses of NaClO3 and euthanized 24 hr after the treatment. Oral administration of NaClO3 resulted in increased hydrogen peroxide levels, lipid, and protein oxidation while thiol and glutathione content and activities of brush border membrane enzymes were decreased in kidney in a NaClO3 dose-dependent manner. Significant alterations in the activities of enzymes involved in carbohydrate metabolism and antioxidant defense were also observed. Administration of NaClO3 induced DNA fragmentation and increased DNA-protein cross-linking. Histological studies showed marked damage in kidney from NaClO3 treated animals. These results strongly suggest that NaClO3 induces nephrotoxicity via redox imbalance that results in DNA and membrane damage, metabolic alterations and brush border membrane enzyme dysfunction.
Collapse
Affiliation(s)
- Shaikh Nisar Ali
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Hussain Arif
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Aijaz Ahmed Khan
- Department of Anatomy, Faculty of Medicine, J. N. Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
516
|
Işıkkent A, Yılmaz S, Özturan İU, Doğan NÖ, Yaka E, Gültekin H, Kum T, Pekdemir M. Utility of neutrophil gelatinase-associated lipocalin in the management of acute kidney injury: A prospective, observational study. HONG KONG J EMERG ME 2018. [DOI: 10.1177/1024907918806641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Utilization of renal biomarkers such as neutrophil gelatinase-associated lipocalin in the management of acute kidney injury may be useful as a diagnostic tool in the emergency department. Objective: The aim of this study is to determine the relationship between serum neutrophil gelatinase-associated lipocalin level and the severity of the acute kidney injury based on the Risk, Injury, Failure, Loss of kidney function, and End-stage kidney disease (RIFLE) classification, and to investigate the role of the serum neutrophil gelatinase-associated lipocalin level in differentiating the etiology and predicting the 30-day mortality rate and need for dialysis. Methods: This prospective, observational study was conducted from March 2015 to 2016. Adult patients with acute kidney injury in the emergency department were enrolled in the study. Demographic and clinical features such as hypovolemic state, nephrotoxic substance exposure, renal functions, and serum neutrophil gelatinase-associated lipocalin level were evaluated. After the etiology of the acute kidney injury was ascertained, the severity of the acute kidney injury was determined according to RIFLE criteria. Primary outcome was defined as the correlation between serum neutrophil gelatinase-associated lipocalin level and the severity of the acute kidney injury according to RIFLE classification. Secondary outcomes were defined as the relationship between the serum neutrophil gelatinase-associated lipocalin level and the etiology of the acute kidney injury; need for dialysis and 30-day mortality were defined as poor outcomes. Results: A total of 87 patients were included in the study. Mean serum neutrophil gelatinase-associated lipocalin levels were 380.14 ± 276.65 ng/mL in RIFLE-R, 425.80 ± 278.99 ng/mL in RIFLE-I, and 403.60 ± 293.15 ng/mL in RIFLE-F groups. There was no statistically significant relationship between the severity of acute kidney injuries and serum neutrophil gelatinase-associated lipocalin level. Initial serum neutrophil gelatinase-associated lipocalin levels in the emergency department did not indicate a statistically significant ability to predict the etiology of acute kidney injury, 30-day mortality rates, or need for dialysis. Conclusion: Initial serum neutrophil gelatinase-associated lipocalin level in the emergency department is not a determinant tool for predicting the severity, etiology, 30-day mortality rates, or need for dialysis in cases of acute kidney injuries.
Collapse
Affiliation(s)
- Ali Işıkkent
- Karabuk University, Training and Reseach Hospital, Department of Emergency Medicine, Karabuk, Turkey
| | - Serkan Yılmaz
- Department of Emergency Medicine, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - İbrahim Ulaş Özturan
- Brown University, Alpert School of Medicine, Brown Advanced Emergency Medicine Academies, Providence, RI USA
| | - Nurettin Özgür Doğan
- Department of Emergency Medicine, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Elif Yaka
- Department of Emergency Medicine, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Haldun Gültekin
- Sakarya University, Training and Research Hospital, Department of Emergency Medicine, Sakarya, Turkey
| | - Tuğba Kum
- Artvin Public Hospital, Department of Biochemistry, Artvin, Turkey
| | - Murat Pekdemir
- Department of Emergency Medicine, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
517
|
Tang C, Ma Z, Zhu J, Liu Z, Liu Y, Liu Y, Cai J, Dong Z. P53 in kidney injury and repair: Mechanism and therapeutic potentials. Pharmacol Ther 2018; 195:5-12. [PMID: 30347214 DOI: 10.1016/j.pharmthera.2018.10.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Acute kidney injury (AKI) is a major kidney disease with poor clinical outcome. Besides its acute consequence of high mortality, AKI may also contribute significantly to the occurrence and progression of chronic kidney diseases (CKD). Accumulating evidence has demonstrated that maladaptive and incomplete kidney repair after AKI leads to the development of renal fibrosis and, ultimately, CKD. p53, a well-known tumor suppressor, plays a critical role in AKI and subsequent kidney repair through the regulation of various cell biologic processes, including apoptosis, cell cycle arrest, and autophagy. Despite the notable progress in deciphering the involvement of p53 in kidney injury and repair, the underlying mechanisms of p53 in these pathological processes remain largely unknown. Further investigation in this area is essential for the application of p53 as therapeutic target to prevent and treat AKI or impede its progression to CKD. In this review, we summarize the recent advances in understanding p53 regulation of AKI and kidney repair, pinpoint the potential of p53 as a therapeutic target, and present future research interests and directions.
Collapse
Affiliation(s)
- Chengyuan Tang
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification in Hunan, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Zhengwei Ma
- Department of Cellular Biology and Anatomy, Charlie Norwood VA Medical Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jiefu Zhu
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification in Hunan, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Zhiwen Liu
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification in Hunan, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Yuxue Liu
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification in Hunan, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification in Hunan, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Juan Cai
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification in Hunan, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification in Hunan, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Charlie Norwood VA Medical Center, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
518
|
Morello J, Derks RJE, Lopes SS, Steenvoorden E, Monteiro EC, Mayboroda OA, Pereira SA. Zebrafish Larvae Are a Suitable Model to Investigate the Metabolic Phenotype of Drug-Induced Renal Tubular Injury. Front Pharmacol 2018; 9:1193. [PMID: 30459607 PMCID: PMC6232664 DOI: 10.3389/fphar.2018.01193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/28/2018] [Indexed: 12/04/2022] Open
Abstract
Prevention and treatment of drug-induced renal injury (DIRI) rely on the availability of sensitive and specific biomarkers of early kidney injury and predictive animal models of human pathophysiology. This study aimed to evaluate the potential of zebrafish larvae as translational model in metabolic profiling of DIRI. Zebrafish larvae were exposed to the lethal concentration for 10% of the larvae (LC10) or ½ LC10 of gentamicin, paracetamol and tenofovir as tenofovir disoproxil fumarate (TDF) and tenofovir (TFV). Metabolites were extracted from whole larvae and analyzed by liquid chromatography-mass spectrometry. Principal component analysis showed that drug exposition to the LC10 of paracetamol, TFV, and TDF was the main source of the variance of the data. To identify the metabolites responsible for the toxic effects of the drugs, partial least squares discriminant analyses were built between the LC10 and ½ LC10 for each drug. Features with variable importance in projection> 1.0 were selected and Venn diagrams were built to differentiate between the common and drug specific metabolites of DIRI. Creatine, tyrosine, glutamine, guanosine, hypoxanthine were identified as common metabolites, adenosine and tryptophan as paracetamol-specific and xanthine and oxidized glutathione as tenofovir-specific. Those metabolic changes can be associated with alterations in energy metabolism, xenobiotic detoxification and protein catabolism, all described in the human pathophysiology of DIRI. Thus, zebrafish proved to be a suitable model to characterize the metabolic changes associated with DIRI. This information can be useful to early diagnose DIRI and to improve our knowledge on the mechanisms of DIRI.
Collapse
Affiliation(s)
- Judit Morello
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Rico J E Derks
- Center for Proteomics and Metabolomics, Leiden University Medical Centre, Leiden, Netherlands
| | - Susana S Lopes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Evelyne Steenvoorden
- Center for Proteomics and Metabolomics, Leiden University Medical Centre, Leiden, Netherlands
| | - Emilia C Monteiro
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Oleg A Mayboroda
- Center for Proteomics and Metabolomics, Leiden University Medical Centre, Leiden, Netherlands.,Department of Chemistry, Tomsk State University, Tomsk, Russia
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
519
|
Wijerathna TM, Mohamed F, Dissanayaka D, Gawarammana I, Palangasinghe C, Shihana F, Endre Z, Shahmy S, Buckley NA. Albuminuria and other renal damage biomarkers detect acute kidney injury soon after acute ingestion of oxalic acid and potassium permanganate. Toxicol Lett 2018; 299:182-190. [PMID: 30300734 DOI: 10.1016/j.toxlet.2018.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/14/2018] [Accepted: 10/02/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND Deliberate self-poisoning with a combination washing powder containing oxalic acid (H2C2O4) and potassium permanganate (KMnO4) is a significant medical problem in the Southern Province of Sri Lanka. Acute kidney injury (AKI) is a frequent consequence. Biomarkers for early diagnosis of nephrotoxicity could guide appropriate supportive therapies. METHODS We investigated the performance of three serum biomarkers and nine urinary biomarkers in 85 patients in an ongoing multicenter prospective cohort study in Sri Lanka exploring AKI following poisoning. RESULTS Sixty two (62/85, 73%) patients developed AKI (acute kidney injury network, AKIN, criteria). Early and rapid increases in serum creatinine (sCr) peaking on day 3 were observed in AKIN stage 2 and 3 patients. In these patients, serum cystatin C (sCysC) rose more gradually but also peaked on day 3. Biomarker concentrations (normalized to urinary creatinine) of urinary albumin (uAlbumin), clusterin (uClusterin), beta-2-microglobulin (uB2M), osteopontin (uOPN), neutrophil gelatinase-associated lipocalin (uNGAL) and kidney injury molecule-1 (uKIM-1) in the AKIN2/3 group increased above the 95th centile concentration of the healthy population. Within 8 h of ingestion, the normalized uAlbumin and sCysC predicted AKIN2/3 with respective area under receiver operating characteristic curve, AUC-ROC values, of 0.94 (95% CI 0.86-1.00) and 0.85 (95% CI 0.76-0.95). CONCLUSIONS Urinary albumin was the best performing AKI biomarker following ingestion of H2C2O4/KMnO4. This may reflect glomerular injury and/or proximal tubular injury. The urinary albumin concentrations observed in this study could generally be detected using albumin specific dipstick methods, easily available even in resource poor settings.
Collapse
Affiliation(s)
- Thilini M Wijerathna
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka; Department of Pathology, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka.
| | - Fahim Mohamed
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka; Department of Pharmacy, Faculty of Allied Health Science, University of Peradeniya, Peradeniya, Sri Lanka; Australian Kidney Biomarker Reference Laboratory, Department of Nephrology, Prince of Wales Hospital and Clinical School, University of New South Wales, Sydney Australia; Clinical Pharmacology and Toxicology Research Group, Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Dhammika Dissanayaka
- Department of Pathology, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Indika Gawarammana
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka; Department of Medicine, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Chathura Palangasinghe
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Fathima Shihana
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka; Clinical Pharmacology and Toxicology Research Group, Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Zoltan Endre
- Australian Kidney Biomarker Reference Laboratory, Department of Nephrology, Prince of Wales Hospital and Clinical School, University of New South Wales, Sydney Australia
| | - Seyed Shahmy
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Nicholas A Buckley
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka; Clinical Pharmacology and Toxicology Research Group, Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
520
|
Storelli F, Samer C, Reny JL, Desmeules J, Daali Y. Complex Drug-Drug-Gene-Disease Interactions Involving Cytochromes P450: Systematic Review of Published Case Reports and Clinical Perspectives. Clin Pharmacokinet 2018; 57:1267-1293. [PMID: 29667038 DOI: 10.1007/s40262-018-0650-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Drug pharmacokinetics (PK) is influenced by multiple intrinsic and extrinsic factors, among which concomitant medications are responsible for drug-drug interactions (DDIs) that may have a clinical relevance, resulting in adverse drug reactions or reduced efficacy. The addition of intrinsic factors affecting cytochromes P450 (CYPs) activity and/or expression, such as genetic polymorphisms and diseases, may potentiate the impact and clinical relevance of DDIs. In addition, greater variability in drug levels and exposures has been observed when such intrinsic factors are present in addition to concomitant medications perpetrating DDIs. This variability results in poor predictability of DDIs and potentially dramatic clinical consequences. The present review illustrates the issue of complex DDIs using systematically searched published case reports of DDIs involving genetic polymorphisms, renal impairment, cirrhosis, and/or inflammation. Current knowledge on the impact of each of these factors on drug exposure and DDIs is summarized and future perspectives for the management of such complex DDIs in clinical practice are discussed, including the use of advanced Computerized Physician Order Entry (CPOE) systems, the development of model-based dose optimization strategies, and the education of healthcare professionals with respect to personalized medicine.
Collapse
Affiliation(s)
- Flavia Storelli
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
- Geneva-Lausanne School of Pharmacy, University of Geneva, Geneva, Switzerland
| | - Caroline Samer
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Jean-Luc Reny
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, Rehabilitation and Geriatrics, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Jules Desmeules
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
- Geneva-Lausanne School of Pharmacy, University of Geneva, Geneva, Switzerland
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Center for Applied Human Toxicology, Geneva, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland.
- Geneva-Lausanne School of Pharmacy, University of Geneva, Geneva, Switzerland.
- Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Swiss Center for Applied Human Toxicology, Geneva, Switzerland.
| |
Collapse
|
521
|
DiRito JR, Hosgood SA, Tietjen GT, Nicholson ML. The future of marginal kidney repair in the context of normothermic machine perfusion. Am J Transplant 2018; 18:2400-2408. [PMID: 29878499 PMCID: PMC6175453 DOI: 10.1111/ajt.14963] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/13/2018] [Accepted: 05/29/2018] [Indexed: 01/25/2023]
Abstract
Normothermic machine perfusion (NMP) is a technique that utilizes extracorporeal membrane oxygenation to recondition and repair kidneys at near body temperature prior to transplantation. The application of this new technology has been fueled by a significant increase in the use of the kidneys that were donated after cardiac death, which are more susceptible to ischemic injury. Preliminary results indicate that NMP itself may be able to repair marginal organs prior to transplantation. In addition, NMP serves as a platform for delivery of therapeutics. The isolated setting of NMP obviates problems of targeting a particular therapy to an intended organ and has the potential to reduce the harmful effects of systemic drug delivery. There are a number of emerging therapies that have shown promise in this platform. Nutrients, therapeutic gases, mesenchymal stromal cells, gene therapies, and nanoparticles, a newly explored modality, have been successfully delivered during NMP. These technologies may be effective at blocking multiple mechanisms of ischemia- reperfusion injury (IRI) and improving renal transplant outcomes. This review addresses the mechanisms of renal IRI, examines the potential for NMP as a platform for pretransplant allograft modulation, and discusses the introduction of various therapies in this setting.
Collapse
Affiliation(s)
- Jenna R. DiRito
- Department of SurgeryUniversity of CambridgeCambridgeUK,Department of SurgeryYale School of MedicineNew HavenCT
| | | | | | | |
Collapse
|
522
|
Ferreira DA, Cruz R, Venâncio C, Faustino-Rocha AI, Silva A, Mesquita JR, Ortiz AL, Vala H. Evaluation of renal injury caused by acute volume replacement with hydroxyethyl starch 130/0.4 or Ringer's lactate solution in pigs. J Vet Sci 2018; 19:608-619. [PMID: 30041290 PMCID: PMC6167341 DOI: 10.4142/jvs.2018.19.5.608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/21/2018] [Accepted: 07/13/2018] [Indexed: 01/22/2023] Open
Abstract
This work aimed to evaluate the effects on renal tissue integrity after hydroxyethyl starch (HES) 130/0.4 and Ringer's lactate (RL) administration in pigs under general anesthesia after acute bleeding. A total of 30 mL/kg of blood were passively removed from the femoral artery in two groups of Large White pigs, under total intravenous anesthesia with propofol and remifentanil. After bleeding, Group 1 (n = 11) received RL solution (25 mL/kg) and Group 2 (n = 11) received HES 130/0.4 solution (20 mL/kg). Additionally, Group 3 (n = 6) was not submitted to bleeding or volume replacement. Pigs were euthanized and kidneys were processed for histopathological and immunohistochemical analyses. Minimal to moderate glomerular, tubular, and interstitial changes, as well as papillary necrosis, were observed in all experimental groups. Pre-apoptosis and apoptosis indicators were higher in pigs that received HES 130/0.4, indicating a higher renal insult. Both HES 130/0.4 and RL administration may cause renal injury, although renal injury may be more significant in pigs receiving HES 13/0.4. Results also suggest that total intravenous anesthesia with propofol and remifentanil may cause renal injury, and this effect can be dose related.
Collapse
Affiliation(s)
- David A Ferreira
- Department of Veterinary Medicine, ICAAM Research Center, University of Évora, 7006-554 Évora, Portugal
| | - Rita Cruz
- Educational, Technologies and Health Study Center (CI&DETS), Polytechnic Institute of Viseu, 3504-510 Viseu, Portugal
| | - Carlos Venâncio
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| | - Ana I Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| | - Aura Silva
- REQUIMTE - Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - João R Mesquita
- Educational, Technologies and Health Study Center (CI&DETS), Polytechnic Institute of Viseu, 3504-510 Viseu, Portugal
| | - Ana L Ortiz
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Helena Vala
- Educational, Technologies and Health Study Center (CI&DETS), Polytechnic Institute of Viseu, 3504-510 Viseu, Portugal
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| |
Collapse
|
523
|
Yeung ACY, Morozov A, Robertson FP, Fuller BJ, Davidson BR. Neutrophil Gelatinase-Associated Lipocalin (NGAL) in predicting acute kidney injury following orthotopic liver transplantation: A systematic review. Int J Surg 2018; 59:48-54. [PMID: 30273683 DOI: 10.1016/j.ijsu.2018.09.003] [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] [Received: 05/19/2018] [Revised: 07/30/2018] [Accepted: 09/07/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) is common after orthotopic liver transplantation (OLT) usually occurring early post-transplant. Multiple causes include graft preservation injury, blood loss, hypotension but also severity of recipient liver disease. Early intervention in AKI has both short and long term patient benefits. Unfortunately there are no current clinical biomarkers of early AKI. AIM To assess the value of NGAL in predicting AKI following OLT. METHODS Ovid MEDLINE and EMBASE were searched between the years of 2000 and 2017 for studies using keywords: Neutrophil Gelatinase Associated Lipocalin or NGAL variants combined with synonyms for liver transplantation. RESULTS 96 studies were identified. 11 studies including 563 patients were considered suitable for analysis. Both urinary (uNGAL) and plasma NGAL (pNGAL) measurement were found to predict AKI after liver transplantation. Optimal reported area under the receiver-operator characteristics curve (AUROC) values of 0.5-0.83 and 0.54-0.86 respectively. CONCLUSIONS NGAL is a good predictor of early AKI post OLT although there is considerable variation in the published results. Further studies with prospectively defined cut-off values, standardized definitions of AKI and rigorous data reporting should be conducted to establish its clinical usefulness and limitations.
Collapse
Affiliation(s)
- Arthur C Y Yeung
- Department of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor Royal Free London NHS Foundation Trust, Pond Street, NW3 2QG, UK
| | - Andrew Morozov
- Department of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor Royal Free London NHS Foundation Trust, Pond Street, NW3 2QG, UK
| | - Francis P Robertson
- Department of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor Royal Free London NHS Foundation Trust, Pond Street, NW3 2QG, UK.
| | - Barry J Fuller
- Department of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor Royal Free London NHS Foundation Trust, Pond Street, NW3 2QG, UK
| | - Brian R Davidson
- Department of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor Royal Free London NHS Foundation Trust, Pond Street, NW3 2QG, UK
| |
Collapse
|
524
|
Polichnowski AJ. Microvascular rarefaction and hypertension in the impaired recovery and progression of kidney disease following AKI in preexisting CKD states. Am J Physiol Renal Physiol 2018; 315:F1513-F1518. [PMID: 30256130 DOI: 10.1152/ajprenal.00419.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is a major complication in hospitalized patients and is associated with elevated mortality rates. Numerous recent studies indicate that AKI also significantly increases the risk of chronic kidney disease (CKD), end-stage renal disease (ESRD), hypertension, cardiovascular disease, and mortality in those patients who survive AKI. Moreover, the risk of ESRD and mortality after AKI is substantially higher in patients with preexisting CKD. However, the underlying mechanisms by which AKI and CKD interact to promote ESRD remain poorly understood. The recently developed models that superimpose AKI on rodents with preexisting CKD have provided new insights into the pathogenic mechanisms mediating the deleterious interactions between AKI and CKD. These studies show that preexisting CKD impairs recovery from AKI and promotes the development of mechanisms of CKD progression. Specifically, preexisting CKD exacerbates microvascular rarefaction, failed tubular redifferentiation, disruption of cell cycle regulation, hypertension, and proteinuria after AKI. The purpose of this review is to discuss the potential mechanisms by which microvascular rarefaction and hypertension contribute to impaired recovery from AKI and the subsequent progression of renal disease in preexisting CKD states.
Collapse
Affiliation(s)
- Aaron J Polichnowski
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee.,Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
525
|
Song Y, Tao Q, Yu L, Li L, Bai T, Song X, Hu H, Li Y, Tan X. Activation of autophagy contributes to the renoprotective effect of postconditioning on acute kidney injury and renal fibrosis. Biochem Biophys Res Commun 2018; 504:641-646. [PMID: 30205956 DOI: 10.1016/j.bbrc.2018.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 09/01/2018] [Indexed: 01/08/2023]
Abstract
Ischemia/Reperfusion injury contributes to acute kidney injury (AKI) and subsequent chronic kidney disease (CKD) including renal fibrosis. Autophagy is a cytoplasmic components degradation pathway that has complex function in the development of various diseases such as fibrosis in kidney. Our previous work demonstrated that postconditioning (POC) showed excellent therapeutic effect on renal fibrosis via inhibiting the overproduction of reactive oxygen species (ROS) after reperfusion. But the connection of autophagy and POC in the renoprotective effect remains unclear. Here, we defined the relevance of autophagy and POC in the protective effect on AKI and subsequent renal fibrosis. We found that at two days after I/R injury, POC largely reduced renal tubular epithelial cell apoptosis and improved renal function; autophagy was significantly activated in kidneys of the POC rats. At two months after reperfusion, the I/R injury rats displayed severe renal fibrosis and epithelial-mesenchymal transition (EMT), whereas these were remarkably attenuated in the POC treated rats. Overall, our results demonstrated that POC could reduce renal damage and attenuate the degree of EMT after I/R injury via enhanced activation of autophagy.
Collapse
Affiliation(s)
- Yaolin Song
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, 130021, China
| | - Qianyu Tao
- Medical College of Qingdao University, Qingdao, Shandong, 266071, China; College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lixia Yu
- Xixi Hospital of Hangzhou, Hangzhou, Zhejiang, 310000, China
| | - Ling Li
- Medical College of Qingdao University, Qingdao, Shandong, 266071, China
| | - Tingting Bai
- Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361000, China
| | - Xiaoxiao Song
- Department of E.N.T, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Haiqi Hu
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, 130021, China.
| | - Xiaohua Tan
- Medical College of Qingdao University, Qingdao, Shandong, 266071, China.
| |
Collapse
|
526
|
Gozdzik W, Zielinski S, Zielinska M, Ratajczak K, Skrzypczak P, Rodziewicz S, Kübler A, Löfström K, Dziegiel P, Olbromski M, Adamik B, Ryniak S, Harbut P, Albert J, Frostell C. Beneficial effects of inhaled nitric oxide with intravenous steroid in an ischemia-reperfusion model involving aortic clamping. Int J Immunopathol Pharmacol 2018; 32:394632017751486. [PMID: 29376749 PMCID: PMC5851102 DOI: 10.1177/0394632017751486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
This study evaluated the effects of inhaled nitric oxide (iNO) therapy combined
with intravenous (IV) corticosteroids on hemodynamics, selected cytokines, and
kidney messenger RNA toll-like receptor 4 (mRNA TLR4) expression in
ischemia–reperfusion injury animal model. The primary endpoint was the
evaluation of circulatory, respiratory, and renal function over time. We also
investigated the profile of selected cytokines and high-mobility group box 1
(HMGB1) protein, as well as renal mRNA TLR4 activation determined by
quantitative real-time polymerase chain reaction analysis. Pigs (n = 19) under
sevoflurane AnaConDa anesthesia/sedation were randomized and subjected to
abdominal laparotomy and alternatively suprarenal aortic cross-clamping (SRACC)
for 90 min or sham surgery: Group 1 (n = 8) iNO (80 ppm) + IV corticosteroids
(25 mg ×3) started 30 min before SRACC and continued 2 h after SRACC release,
followed with decreased iNO (30 ppm) until the end of observation, Group 2
(n = 8) 90 min SRACC, Group 3 (n = 3)—sham surgery. Renal biopsies were sampled
1 hr before SRACC and at 3 and 20 h after SRACC release. Aortic clamping
increased TLR4 mRNA expression in ischemic kidneys, but significant changes were
recorded only in the control group (P = 0.016).
Treatment with iNO and hydrocortisone reduced TLR4 mRNA expression to
pre-ischemic conditions, and the difference observed in mRNA expression was
significant between control and treatment group after 3 h (P = 0.042). Moreover, animals subjected to treatment with iNO and
hydrocortisone displayed an attenuated systemic inflammatory response and
lowered pulmonary vascular resistance plus increased oxygen delivery. The
results indicated that iNO therapy combined with IV corticosteroids improved
central and systemic hemodynamics, oxygen delivery, and diminished the systemic
inflammatory response and renal mRNA TLR4 expression.
Collapse
Affiliation(s)
- Waldemar Gozdzik
- 1 Department of Anaesthesiology and Intensive Therapy, Wrocław Medical University, Wrocław, Poland
| | - Stanisław Zielinski
- 1 Department of Anaesthesiology and Intensive Therapy, Wrocław Medical University, Wrocław, Poland
| | - Marzena Zielinska
- 1 Department of Anaesthesiology and Intensive Therapy, Wrocław Medical University, Wrocław, Poland
| | - Kornel Ratajczak
- 2 Department and Clinic of Surgery, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Piotr Skrzypczak
- 2 Department and Clinic of Surgery, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Sylwia Rodziewicz
- 2 Department and Clinic of Surgery, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Andrzej Kübler
- 1 Department of Anaesthesiology and Intensive Therapy, Wrocław Medical University, Wrocław, Poland
| | - Kalle Löfström
- 3 Department of Anesthesia and Intensive Care, Danderyd Hospital, Stockholm, Sweden
| | - Piotr Dziegiel
- 4 Department of Histology and Embryology, Wrocław Medical University, Wrocław, Poland
| | - Mateusz Olbromski
- 4 Department of Histology and Embryology, Wrocław Medical University, Wrocław, Poland
| | - Barbara Adamik
- 1 Department of Anaesthesiology and Intensive Therapy, Wrocław Medical University, Wrocław, Poland
| | - Stanislaw Ryniak
- 3 Department of Anesthesia and Intensive Care, Danderyd Hospital, Stockholm, Sweden
| | - Piotr Harbut
- 3 Department of Anesthesia and Intensive Care, Danderyd Hospital, Stockholm, Sweden
| | - Johanna Albert
- 3 Department of Anesthesia and Intensive Care, Danderyd Hospital, Stockholm, Sweden
| | - Claes Frostell
- 5 Department of Clinical Sciences at Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| |
Collapse
|
527
|
Tibúrcio FR, Rodrigues KEDS, Belisário AR, Simões-e-Silva AC. Glomerular hyperfiltration and β-2 microglobulin as biomarkers of incipient renal dysfunction in cancer survivors. Future Sci OA 2018; 4:FSO333. [PMID: 30271618 PMCID: PMC6153459 DOI: 10.4155/fsoa-2018-0045] [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] [Received: 04/18/2018] [Accepted: 06/26/2018] [Indexed: 11/17/2022] Open
Abstract
Herein, we aimed to evaluate the occurrence of impaired renal function after cancer treatment with potentially nephrotoxic chemotherapy in children. A cross-sectional study was performed in 41 cancer survivors after chemotherapy with potentially nephrotoxic drugs. 26 (63.4%) children were detected with glomerular hyperfiltration, and urinary levels of β-2 microglobulin (B2MG) were higher than reference range in all patients. Levels of B2MG were positively correlated with plasma creatinine and negatively correlated with glomerular filtration rate. Plasma creatinine, systolic blood pressure and cholesterol were independently associated with B2MG values. The final multivariate model for glomerular hyperfiltration risk included plasma levels of urea and of magnesium. Urinary levels of B2MG and glomerular hyperfiltration may emerge as potential biomarkers of early renal dysfunction in childhood cancer survivors.
Collapse
Affiliation(s)
- Fernanda R Tibúrcio
- Department of Pediatrics, Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Pediatric Nephrology Unity, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Karla E de S Rodrigues
- Department of Pediatrics, Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Pediatric Nephrology Unity, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - André R Belisário
- Department of Pediatrics, Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Pediatric Nephrology Unity, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Cristina Simões-e-Silva
- Department of Pediatrics, Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Pediatric Nephrology Unity, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
528
|
Kildey K, Law BMP, Muczynski KA, Wilkinson R, Helen H, Kassianos AJ. Identification and Quantitation of Leukocyte Populations in Human Kidney Tissue by Multi-parameter Flow Cytometry. Bio Protoc 2018; 8:e2980. [PMID: 34395780 DOI: 10.21769/bioprotoc.2980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/02/2018] [Accepted: 08/06/2018] [Indexed: 11/02/2022] Open
Abstract
Inflammatory immune cells play direct pathological roles in cases of acute kidney injury (AKI) and chronic kidney disease (CKD). However, the identification and characterization of distinct populations of leukocytes in human kidney biopsies have been confounded by the limitations of immunohistochemical (IHC)-based techniques used to detect them. This methodology is not amenable to the combinations of multiple markers necessary to unequivocally define discrete immune cell populations. We have developed a multi-parameter, flow cytometric-based approach that addresses the need for panels of cell-specific markers in the identification of immune cell populations, allowing both the accurate detection and quantitation of leukocyte subpopulations from a single, clinical kidney biopsy specimen. In this approach, fresh human kidney tissue is dissociated into a single cell suspension followed by antibody-labeling and flow cytometric-based acquisition and analysis. This novel technique provides a major step forward in identifying and enumerating immune cell subpopulations in human kidney disease and is a powerful platform to complement traditional histopathological examinations of clinical kidney biopsies.
Collapse
Affiliation(s)
- Katrina Kildey
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Becker M P Law
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Ray Wilkinson
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,Medical School, University of Queensland, Brisbane, Queensland, Australia
| | - Healy Helen
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Medical School, University of Queensland, Brisbane, Queensland, Australia
| | - Andrew J Kassianos
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,Medical School, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
529
|
Abstract
OBJECTIVE Nephritis has been rarely associated with methimazole, primarily in the development of nephrotic syndrome. We describe a case of acute kidney injury without evidence of nephrotic syndrome following methimazole initiation. METHODS We present the relevant history, laboratory data, and nuclear medicine data and review relevant documentation from the literature. RESULTS A 72-year-old male recently diagnosed with new-onset atrial fibrillation was found to have suppressed thyroid-stimulating hormone (TSH) levels; elevated free T3, T4, and thyroid-stimulating immunoglobulin (TSI) levels; and a nonnodular thyroid gland with normal iodine uptake. He was diagnosed with Graves' disease and treated with propylthiouracil (PTU) for 5 years. When his poor compliance with PTU was impeding his antithyroid treatment, he was converted to methimazole. Within 1 month following methimazole initiation, his serum creatinine (SCr) had risen to 1.6× baseline in the absence of other contributing nephrotoxins. SCr returned to baseline within 2 weeks of methimazole discontinuation, and the patient was subsequently managed on PTU. CONCLUSION Acute kidney injury with or without the presence of nephrotic syndrome may occur during treatment with methimazole. Renal function should be closely monitored after the initiation of methimazole to prevent progressive renal dysfunction.
Collapse
Affiliation(s)
- Abigail Shell
- Department of Pharmacy, Memphis VA Medical Center, Memphis, TN, USA
| | | |
Collapse
|
530
|
Honarpisheh M, Foresto-Neto O, Steiger S, Kraft F, Koehler P, von Rauchhaupt E, Potempa J, Adamowicz K, Koziel J, Lech M. Aristolochic acid I determine the phenotype and activation of macrophages in acute and chronic kidney disease. Sci Rep 2018; 8:12169. [PMID: 30111809 PMCID: PMC6093867 DOI: 10.1038/s41598-018-30628-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 08/03/2018] [Indexed: 01/12/2023] Open
Abstract
Acute and chronic kidney injuries are multifactorial traits that involve various risk factors. Experimental animal models are crucial to unravel important aspects of injury and its pathophysiological mechanisms. Translating knowledge obtained from experimental approaches into clinically useful information is difficult; therefore, significant attention needs to be paid to experimental procedures that mimic human disease. Herein, we compared aristolochic acid I (AAI) acute and chronic kidney injury model with unilateral ischemic-reperfusion injury (uIRI), cisplatin (CP)- or folic acid (FA)-induced renal damage. The administration of AAI showed significant changes in serum creatinine and BUN upon CKD. The number of neutrophils and macrophages were highly increased as well as AAI-induced CKD characterized by loss of tubular epithelial cells and fibrosis. The in vitro and in vivo data indicated that macrophages play an important role in the pathogenesis of AA-induced nephropathy (AAN) associated with an excessive macrophage accumulation and an alternative activated macrophage phenotype. Taken together, we conclude that AA-induced injury represents a suitable and relatively easy model to induce acute and chronic kidney injury. Moreover, our data indicate that this model is appropriate and superior to study detailed questions associated with renal macrophage phenotypes.
Collapse
Affiliation(s)
- Mohsen Honarpisheh
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Orestes Foresto-Neto
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Stefanie Steiger
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Franziska Kraft
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Paulina Koehler
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Ekaterina von Rauchhaupt
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Jan Potempa
- Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Karina Adamowicz
- Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Joanna Koziel
- Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Lech
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany. .,Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
531
|
Barrera-Chimal J, Rocha L, Amador-Martínez I, Pérez-Villalva R, González R, Cortés-González C, Uribe N, Ramírez V, Berman N, Gamba G, Bobadilla NA. Delayed spironolactone administration prevents the transition from acute kidney injury to chronic kidney disease through improving renal inflammation. Nephrol Dial Transplant 2018; 34:794-801. [DOI: 10.1093/ndt/gfy246] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Indexed: 01/03/2023] Open
Affiliation(s)
- Jonatan Barrera-Chimal
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Unidad de Medicina Traslacional, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Leslie Rocha
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Nephrology Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Isabel Amador-Martínez
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Unidad de Medicina Traslacional, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Rosalba Pérez-Villalva
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Nephrology Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rafael González
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Nephrology Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Cesar Cortés-González
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Mexico City, Mexico and
| | - Norma Uribe
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Victoria Ramírez
- Department of Nephrology Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nathan Berman
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Nephrology Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Gerardo Gamba
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Nephrology Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Norma A Bobadilla
- Molecular Physiology Unit, Department of Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Nephrology Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
532
|
Wen X, Cui L, Morrisroe S, Maberry D, Emlet D, Watkins S, Hukriede NA, Kellum JA. A zebrafish model of infection-associated acute kidney injury. Am J Physiol Renal Physiol 2018; 315:F291-F299. [PMID: 29537312 PMCID: PMC6139521 DOI: 10.1152/ajprenal.00328.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 02/21/2018] [Accepted: 03/11/2018] [Indexed: 12/25/2022] Open
Abstract
Sepsis-associated acute kidney injury (S-AKI) independently predicts mortality among critically ill patients. The role of innate immunity in this process is unclear, and there is an unmet need for S-AKI models to delineate the pathophysiological response. Mammals and zebrafish ( Danio rerio) share a conserved nephron structure and homologous innate immune systems, making the latter suitable for S-AKI research. We introduced Edwardsiella tarda to the zebrafish. Systemic E. tarda bacteremia resulted in sustained bacterial infection and dose-dependent mortality. A systemic immune reaction was characterized by increased mRNA expressions of il1b, tnfa, tgfb1a, and cxcl8-l1 ( P < 0.0001, P < 0.001, P < 0.001, and P < 0.01, respectively). Increase of host stress response genes ccnd1 and tp53 was observed at 24 h postinjection ( P < 0.0001 and P < 0.05, respectively). Moderate E. tarda infection induced zebrafish mortality of over 50% in larvae and 20% in adults, accompanied by pericardial edema in larvae and renal dysfunction in both larval and adult zebrafish. Expression of AKI markers insulin-like growth factor-binding protein-7 (IGFBP7), tissue inhibitor of metalloproteinases 2 (TIMP-2), and kidney injury molecule-1 (KIM-1) was found to be significantly increased in the septic animals at the transcription level ( P < 0.01, P < 0.05, and P < 0.05) and in nephric tubules compared with noninfected animals. In conclusion, we established a zebrafish model of S-AKI induced by E. tarda injection, with both larval and adult zebrafish showing nephron injury in the setting of infection.
Collapse
Affiliation(s)
- Xiaoyan Wen
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Liyan Cui
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Seth Morrisroe
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Donald Maberry
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - David Emlet
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Simon Watkins
- Center for Biologic Imaging, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Neil A Hukriede
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
- Department of Developmental Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
533
|
Yan M, Shu S, Chunyuan G, Tang C, Dong Z. Endoplasmic reticulum stress in ischemic and nephrotoxic acute kidney injury. Ann Med 2018; 50:381-390. [PMID: 29895209 PMCID: PMC6333465 DOI: 10.1080/07853890.2018.1489142] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/02/2018] [Accepted: 03/19/2018] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is a medical condition characterized by kidney damage with a rapid decline of renal function, which is associated with high mortality and morbidity. Recent research has further established an intimate relationship between AKI and chronic kidney disease. Perturbations of kidney cells in AKI result in the accumulation of unfolded and misfolded proteins in the endoplasmic reticulum (ER), leading to unfolded protein response (UPR) or ER stress. In this review, we analyze the role and regulation of ER stress in AKI triggered by renal ischemia-reperfusion and cisplatin nephrotoxicity. The balance between the two major components of UPR, the adaptive pathway and the apoptotic pathway, plays a critical role in determining the cell fate in ER stress. The adaptive pathway is evoked to attenuate translation, induce chaperones, maintain protein homeostasis and promote cell survival. Prolonged ER stress activates the apoptotic pathway, resulting in the elimination of dysfunctional cells. Therefore, regulating ER stress in kidney cells may provide a therapeutic target in AKI. KEY MESSAGES Perturbations of kidney cells in acute kidney injury result in the accumulation of unfolded and misfolded proteins in ER, leading to unfolded protein response (UPR) or ER stress. The balance between the adaptive pathway and the apoptotic pathway of UPR plays a critical role in determining the cell fate in ER stress. Modulation of ER stress in kidney cells may provide a therapeutic strategy for acute kidney injury.
Collapse
Affiliation(s)
- Mingjuan Yan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Nephrology, The First people’s Hospital of Changde City, Changde, Hunan, China
| | - Shaoqun Shu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guo Chunyuan
- Department of Nephrology, The First people’s Hospital of Changde City, Changde, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, U.S.A
| |
Collapse
|
534
|
Moonen L, D'Haese PC, Vervaet BA. Epithelial Cell Cycle Behaviour in the Injured Kidney. Int J Mol Sci 2018; 19:E2038. [PMID: 30011818 PMCID: PMC6073451 DOI: 10.3390/ijms19072038] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI), commonly caused by ischemia-reperfusion injury, has far-reaching health consequences. Despite the significant regenerative capacity of proximal tubular epithelium cells (PTCs), repair frequently fails, leading to the development of chronic kidney disease (CKD). In the last decade, it has been repeatedly demonstrated that dysregulation of the cell cycle can cause injured kidneys to progress to CKD. More precisely, severe AKI causes PTCs to arrest in the G1/S or G2/M phase of the cell cycle, leading to maladaptive repair and a fibrotic outcome. The mechanisms causing these arrests are far from known. The arrest might, at least partially, be attributed to DNA damage since activation of the DNA-damage response pathway leads to cell cycle arrest. Alternatively, cytokine signalling via nuclear factor kappa beta (NF-κβ) and p38-mitogen-activated protein kinase (p38-MAPK) pathways, and reactive oxygen species (ROS) can play a role independent of DNA damage. In addition, only a handful of cell cycle regulators (e.g., p53, p21) have been thoroughly studied during renal repair. Still, why and how PTCs decide to arrest their cell cycle and how this arrest can efficiently be overcome remain open and challenging questions. In this review we will discuss the evidence for cell cycle involvement during AKI and development of CKD together with putative therapeutic approaches.
Collapse
Affiliation(s)
- Lies Moonen
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Benjamin A Vervaet
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| |
Collapse
|
535
|
Rowart P, Wu J, Caplan MJ, Jouret F. Implications of AMPK in the Formation of Epithelial Tight Junctions. Int J Mol Sci 2018; 19:E2040. [PMID: 30011834 PMCID: PMC6073107 DOI: 10.3390/ijms19072040] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 01/13/2023] Open
Abstract
Tight junctions (TJ) play an essential role in the epithelial barrier. By definition, TJ are located at the demarcation between the apical and baso-lateral domains of the plasma membrane in epithelial cells. TJ fulfill two major roles: (i) TJ prevent the mixing of membrane components; and (ii) TJ regulate the selective paracellular permeability. Disruption of TJ is regarded as one of the earliest hallmarks of epithelial injury, leading to the loss of cell polarity and tissue disorganization. Many factors have been identified as modulators of TJ assembly/disassembly. More specifically, in addition to its role as an energy sensor, adenosine monophosphate-activated protein kinase (AMPK) participates in TJ regulation. AMPK is a ubiquitous serine/threonine kinase composed of a catalytic α-subunit complexed with regulatory β-and γ-subunits. AMPK activation promotes the early stages of epithelial TJ assembly. AMPK phosphorylates the adherens junction protein afadin and regulates its interaction with the TJ-associated protein zonula occludens (ZO)-1, thereby facilitating ZO-1 distribution to the plasma membrane. In the present review, we detail the signaling pathways up-and down-stream of AMPK activation at the time of Ca2+-induced TJ assembly.
Collapse
Affiliation(s)
- Pascal Rowart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Avenue de L'Hôpital 11, 4000 Liège, Belgium.
| | - Jingshing Wu
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - François Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Avenue de L'Hôpital 11, 4000 Liège, Belgium.
- Division of Nephrology, Centre Hospitalier Universitaire de Liège (CHU of Liège), University of Liège (CHU ULiège), 13-B4000 Liège, Belgium.
| |
Collapse
|
536
|
Chihanga T, Ruby HN, Ma Q, Bashir S, Devarajan P, Kennedy MA. NMR-based urine metabolic profiling and immunohistochemistry analysis of nephron changes in a mouse model of hypoxia-induced acute kidney injury. Am J Physiol Renal Physiol 2018; 315:F1159-F1173. [PMID: 29993280 DOI: 10.1152/ajprenal.00500.2017] [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: 02/08/2023] Open
Abstract
Acute kidney injury can be caused by multiple factors, including sepsis, respiratory failure, heart failure, trauma, or nephrotoxic medications, among others. Here, a mouse model was used to investigate potential urinary metabolic biomarkers of hypoxia-induced AKI. Urine metabolic profiles of 48 Swiss Webster mice were assessed using nuclear magnetic resonance spectroscopy (NMR) for 7 days following 72 h exposure to a hypoxic 6.5% oxygen environment. Histological analyses indicated a lack of gross nephron structural changes in the aftermath of hypoxia. Immunohistochemical (IHC) analyses, however, indicated elevated expression of protein injury biomarkers in distal and proximal tubules but not glomeruli. Kidney injury molecule-1 levels peaked in distal tubules at 72 h and were still increasing in proximal tubules at 7 days posthypoxia, whereas cystatin C levels were elevated at 24 h but decreased thereafter, and were elevated and still increasing in proximal tubules at 7 days posthypoxia. Neutrophil gelatinase-associated lipocalin levels were modestly elevated from 24 h to 7 days posthypoxia. NMR-based metabolic profiling revealed that urine metabolites involved in energy metabolism and associated biosynthetic pathways were initially decreased at 24 h posthypoxia, consistent with metabolic suppression as a mechanism for cell survival, but were significantly elevated at 48 and 72 h posthypoxia, indicating a burst in organism metabolism associated with reactivation of cellular energetics during recovery after cessation of hypoxia and return to a normoxic environment. The IHC results indicated that kidney injury persists long after plasma and urine biomarkers of hypoxia return to normal values.
Collapse
Affiliation(s)
- Tafadzwa Chihanga
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| | - Hannah N Ruby
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| | - Qing Ma
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, Ohio
| | - Sabina Bashir
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| | - Prasad Devarajan
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, Ohio
| | - Michael A Kennedy
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| |
Collapse
|
537
|
Chakraborty A, Viswanathan P. Methylation-Demethylation Dynamics: Implications of Changes in Acute Kidney Injury. Anal Cell Pathol (Amst) 2018. [DOI: https://doi.org/10.1155/2018/8764384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Over the years, the epigenetic landscape has grown increasingly complex. Until recently, methylation of DNA and histones was considered one of the most important epigenetic modifications. However, with the discovery of enzymes involved in the demethylation process, several exciting prospects have emerged that focus on the dynamic regulation of methylation and its crucial role in development and disease. An interplay of the methylation-demethylation machinery controls the process of gene expression. Since acute kidney injury (AKI), a major risk factor for chronic kidney disease and death, is characterised by aberrant expression of genes, understanding the dynamics of methylation and demethylation will provide new insights into the intricacies of the disease. Research on epigenetics in AKI has only made its mark in the recent years but has provided compelling evidence that implicates the involvement of methylation and demethylation changes in its pathophysiology. In this review, we explore the role of methylation and demethylation machinery in cellular epigenetic control and further discuss the contribution of methylomic changes and histone modifications to the pathophysiology of AKI.
Collapse
Affiliation(s)
- Anubhav Chakraborty
- Renal Research Lab, Centre for Bio-Medical Research, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Bio-Medical Research, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
538
|
Lin TJ, Huang YL, Chang JS, Liu KT, Yen MC, Chen FW, Shih YL, Jao JC, Huang PC, Yeh IJ. Optimal dosage and early intervention of L-ascorbic acid inhibiting K 2Cr 2O 7-induced renal tubular cell damage. J Trace Elem Med Biol 2018; 48:1-7. [PMID: 29773167 DOI: 10.1016/j.jtemb.2018.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 02/02/2018] [Accepted: 02/23/2018] [Indexed: 01/12/2023]
Abstract
Chromium poisoning can cause renal failure and death. Chromium intoxication may be managed using L-ascorbic acid (vitamin C) therapy. However, the evidence supporting the effectiveness of this treatment is insufficient, and the mechanism of action has not been clarified in renal cells. In this study, our results showed that the optimal regimen of L-ascorbic acid therapy in human epithelial renal proximal tubule cells, HK-2 cells, was 30 μg/mL. Supplementation of L-ascorbic acid with 30 μg/mL and within 8 h of chromium intoxication (K2Cr2O7, Cr6+) was effective to inhibit renal tubular cell damage by blocking generation of free radicals, cell apoptosis, and autophagy. Intracellular chromium concentrations were estimated using electrothermal atomic absorption spectrometry. Treatment of L-ascorbic acid within 8 h of chromium intoxication significantly decreased the entry of chromium into the cells. Moreover, concomitant administration of L-ascorbic acid with repeatedly dosing at 8-hourly intervals had a better protective effect at lower concentration of L-ascorbic acid when compared to single dosing of L-ascorbic acid at an early time point of chromium intoxication. These findings might help physicians develop effective therapy strategies in renal failure.
Collapse
Affiliation(s)
- Tzeng-Jih Lin
- Emergency Department, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Family Medicine Department, Taoyuan Branch, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yeou-Lih Huang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jung-San Chang
- Department of Renal Care, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuan-Ting Liu
- Emergency Department, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Meng-Chi Yen
- Emergency Department, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fen-Wei Chen
- Emergency Department, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yueh-Lun Shih
- Emergency Department, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jo-Chi Jao
- Department of Medical Imaging and Radiological Sciences, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Chi Huang
- Department of Pathology, Taipei City Hospital, Taipei, Taiwan
| | - I-Jeng Yeh
- Emergency Department, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
539
|
Campion CG, Zaoui K, Verissimo T, Cossette S, Matsuda H, Solban N, Hamet P, Tremblay J. COMMD5/HCaRG Hooks Endosomes on Cytoskeleton and Coordinates EGFR Trafficking. Cell Rep 2018; 24:670-684.e7. [DOI: 10.1016/j.celrep.2018.06.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 05/16/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022] Open
|
540
|
Baban B, Hoda N, Malik A, Khodadadi H, Simmerman E, Vaibhav K, Mozaffari MS. Impact of cannabidiol treatment on regulatory T-17 cells and neutrophil polarization in acute kidney injury. Am J Physiol Renal Physiol 2018; 315:F1149-F1158. [PMID: 29897289 DOI: 10.1152/ajprenal.00112.2018] [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: 12/18/2022] Open
Abstract
Hallmark features of acute kidney injury (AKI) include mobilization of immune and inflammatory mechanisms culminating in tissue injury. Emerging information indicates heterogeneity of neutrophils with pro- and anti-inflammatory functions (N1 and N2, respectively). Also, regulatory T-17 (Treg17) cells curtail T helper 17 (Th-17)-mediated proinflammatory responses. However, the status of Treg17 cells and neutrophil phenotypes in AKI are not established. Furthermore, cannabidiol exerts immunoregulatory effects, but its impact on Treg17 cells and neutrophil subtypes is not established. Thus, we examined the status of Treg17 cells and neutrophil subtypes in AKI and determined whether cannabidiol favors regulatory neutrophils and T cells accompanied with renoprotection. Accordingly, mice were subjected to bilateral renal ischemia-reperfusion injury (IRI), without or with cannabidiol treatment; thereafter, kidneys were processed for flow cytometry analyses. Renal IRI increased N1 and Th-17 but reduced N2 and Treg17 cells accompanied with disruption of mitochondrial membrane potential (ψm) and increased apoptosis/necrosis and kidney injury molecule-1 (KIM-1) immunostaining compared with their sham controls. Importantly, cannabidiol treatment preserved ψm and reduced cell death and KIM-1 accompanied by restoration of N1 and N2 imbalance and preservation of Treg17 cells while decreasing Th-17 cells. The ability of cannabidiol to favor development of Treg17 cells was further established using functional mixed lymphocytic reaction. Subsequent studies showed higher renal blood flow and reduced serum creatinine in cannabidiol-treated IRI animals. Collectively, our novel observations establish that renal IRI causes neutrophil polarization in favor of N1 and also reduces Treg17 cells in favor of Th-17, effects that are reversed by cannabidiol treatment accompanied with significant renoprotection.
Collapse
Affiliation(s)
- Babak Baban
- Department of Oral Biology, Dental College of Georgia, Augusta University , Augusta, Georgia.,Department of Neurology, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Nasrul Hoda
- Department of Neurology, Medical College of Georgia, Augusta University , Augusta, Georgia.,Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University , Augusta, Georgia
| | - Aneeq Malik
- Department of Oral Biology, Dental College of Georgia, Augusta University , Augusta, Georgia
| | - Hesam Khodadadi
- Department of Oral Biology, Dental College of Georgia, Augusta University , Augusta, Georgia
| | - Erika Simmerman
- Department of Surgery, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Mahmood S Mozaffari
- Department of Oral Biology, Dental College of Georgia, Augusta University , Augusta, Georgia
| |
Collapse
|
541
|
Renal resistive index as an early predictor and discriminator of acute kidney injury in critically ill patients; A prospective observational cohort study. PLoS One 2018; 13:e0197967. [PMID: 29889830 PMCID: PMC5995360 DOI: 10.1371/journal.pone.0197967] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/05/2018] [Indexed: 01/04/2023] Open
Abstract
Background Acute kidney injury (AKI) complicates shock. Diagnosis is based on rising creatinine, a late phenomenon. Intrarenal vasoconstriction occurs earlier. Measuring flow resistance in the renal circulation, Renal Resistive Index (RRI), could become part of vital organ function assessment using Doppler ultrasound. Our aim was to determine whether RRI on ICU admission is an early predictor and discriminator of AKI developed within the first week. Methods In this prospective cohort of mixed ICU patients with and without shock, RRI was measured <24-h of admission. Besides routine variables, sublingual microcirculation and bioelectrical impedance were measured. AKI was defined by the Kidney Disease Improving Global Outcomes criteria. Uni- and multivariate regression and Receiver Operating Characteristics curve analyses were performed. Results Ninety-nine patients were included, median age 67 years (IQR 59–75), APACHE III score 67 (IQR 53–89). Forty-nine patients (49%) developed AKI within the first week. AKI patients had a higher RRI on admission than those without: 0.71 (0.69–0.73) vs. 0.65 (0.63–0.68), p = 0.001. The difference was significant for AKI stage 2: RRI = 0.72 (0.65–0.80) and 3: RRI = 0.74 (0.67–0.81), but not for AKI stage 1: RRI = 0.67 (0.61–0.74). On univariate analysis, RRI significantly predicted AKI 2–3: OR 1.012 (1.006–1.019); Area Under the Curve (AUC) of RRI for AKI 2–3 was 0.72 (0.61–0.83), optimal cut-off 0.74, sensitivity 53% and specificity 87%. On multivariate analysis, RRI remained significant, independent of APACHE III and fluid balance; adjusted OR: 1.008 (1.000–1.016). Conclusions High RRI on ICU admission was a significant predictor for development of AKI stage 2–3 during the first week. High RRI can be used as an early warning signal RRI, because of its high specificity. A combined score including RRI, APACHE III and fluid balance improved AKI prediction, suggesting that vasoconstriction or poor vascular compliance, severity of disease and positive fluid balance independently contribute to AKI development. Trial registration ClinicalTrials.gov NCT02558166.
Collapse
|
542
|
Gaião SM, Paiva JAODC. Biomarkers of renal recovery after acute kidney injury. Rev Bras Ter Intensiva 2018; 29:373-381. [PMID: 29044306 PMCID: PMC5632981 DOI: 10.5935/0103-507x.20170051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/28/2017] [Indexed: 01/21/2023] Open
Abstract
Novel biomarkers can be suitable for early acute kidney injury diagnosis and the
prediction of the need for dialysis. It remains unclear whether such biomarkers
may also play a role in the prediction of recovery after established acute
kidney injury or in aiding the decision of when to stop renal support therapy.
PubMed, Web of Science and Google Scholar were searched for studies that
reported on the epidemiology of renal recovery after acute kidney injury, the
risk factors of recovery versus non-recovery after acute kidney injury, and
potential biomarkers of acute kidney injury recovery. The reference lists of
these articles and relevant review articles were also reviewed. Final references
were selected for inclusion in the review based on their relevance. New
biomarkers exhibited a potential role in the early diagnosis of acute kidney
injury recovery. Urine HGF, IGFBP-7, TIMP-2 and NGAL may improve our ability to
predict the odds and timing of recovery and eventually renal support withdrawal.
Acute kidney injury recovery requires more study, and its definition needs to be
standardized to allow for better and more powerful research on biomarkers
because some of them show potential for the prediction of acute kidney injury
recovery.
Collapse
Affiliation(s)
- Sérgio Mina Gaião
- Department of Emergency and Intensive Care, Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto - Porto, Portugal.,Infection and Sepsis Group - Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto - Porto, Portugal
| | - José Artur Osório de Carvalho Paiva
- Department of Emergency and Intensive Care, Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto - Porto, Portugal.,Infection and Sepsis Group - Centro Hospitalar São João, Faculdade de Medicina, Universidade do Porto - Porto, Portugal
| |
Collapse
|
543
|
Mazdak H, Ghavami M, Dolatkhah S, Daneshpajouhnejad P, Fesharakizadeh M, Fesharakizadeh S, Atapour A, Mahzouni P, Hashemi M, Salajegheh R, Taheri D. Pathological assessment of allograft nephrectomy: An Iranian experience. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2018; 23:55. [PMID: 30057639 PMCID: PMC6040153 DOI: 10.4103/jrms.jrms_440_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/20/2017] [Accepted: 04/16/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND The aim of this study was to determine the pathologic causes of renal allograft failure in transplant nephrectomy specimens. MATERIALS AND METHODS In this cross-sectional study performed in the referral transplant center of Isfahan, Iran, medical files of all patients who underwent nephrectomy in 2008-2013 were studied. Age at transplantation, sex, donor's characteristics, causes of primary renal failure, duration of allograft function, and pathologic reasons of nephrectomy were extracted. Slides of nephrectomy biopsies were evaluated. Data were analyzed using SPSS. RESULTS Medical files of 39 individuals (male: 56.4%; mean age: 35.1 ± 16.0 years) were evaluated. The main disease of patients was hypertension (17.9%), and most cases (64.1%) were nephrectomized < 6 months posttransplantation. Renal vein thrombosis (RVT) (51.3%) and T-cell-mediated rejection (TCMR) (41.0%) were the most prevalent causes of transplanted nephrectomy. Cause of primary renal failure was correlated to nephrectomy result (P = 0.04). TCMR was the only pathologic finding in all of patients nephrectomized >2 years posttransplantation. There were 14 cases in which biopsy results showed a relationship between primary disease of patients and pathologic assessment of allograft (P = 0.04). A significant relationship between transplantation-nephrectomy interval and both the nephrectomy result and histopathologic result existed (P < 0.0001). A relationship between primary allograft biopsy appearance and further assessment of nephrectomized specimen (P < 0.001) existed as well. CONCLUSION The most pathologic diagnoses of nephrectomy in a period of less than and more than 6 months posttransplantation were RVT and TCMR, respectively. Early obtained allograft protocol biopsy is suggested, which leads to better diagnosis of allograft failure.
Collapse
Affiliation(s)
- Hamid Mazdak
- Department of Urology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojgan Ghavami
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahaboddin Dolatkhah
- Department of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parnaz Daneshpajouhnejad
- Isfahan Medical Students’ Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Pathology, Isfahan Kidney Disease Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Fesharakizadeh
- Department of Surgery, Najaf Abad Branch of Islamic Azad University, Isfahan, Iran
| | | | - Abdolamir Atapour
- Department of Nephrology, Isfahan University of Medical Sciences, Isfahan, Iran
- Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Mahzouni
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozaffar Hashemi
- Department of Thoracic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roxana Salajegheh
- Department of Pathology, Faculty of Medicine, Yazd University of Medical Sciences, Yazd, Iran
| | - Diana Taheri
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
- Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
544
|
Nontraumatic Exertional Rhabdomyolysis Leading to Acute Kidney Injury in a Sickle Trait Positive Individual on Renal Biopsy. Case Rep Nephrol 2018; 2018:5841216. [PMID: 29850311 PMCID: PMC5925017 DOI: 10.1155/2018/5841216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 02/07/2018] [Accepted: 03/04/2018] [Indexed: 01/06/2023] Open
Abstract
A 26-year-old African American male with a history of congenital cerebral palsy, sickle cell trait, and intellectual disability presented with abdominal pain that started four hours prior to the hospital visit. The patient denied fever, chills, diarrhea, or any localized trauma. The patient was at a party at his community center last evening and danced for 2 hours, physically exerting himself more than usual. Labs revealed blood urea nitrogen (BUN) level of 41 mg/dL and creatinine (Cr) of 2.8 mg/dL which later increased to 4.2 mg/dL while still in the emergency room. Urinalysis revealed hematuria with RBC > 50 on high power field. Imaging of the abdomen revealed no acute findings for abdominal pain. With fractional excretion of sodium (FeNa) > 3%, findings suggested nonoliguric acute tubular necrosis. Over the next couple of days, symptoms of dyspepsia resolved; however, BUN/Cr continued to rise to a maximum of 122/14 mg/dL. With these findings, along with stable electrolytes, urine output matching the intake, and prior use of proton pump inhibitors, medical decision was altered for the possibility of acute interstitial nephritis. Steroids were subsequently started and biopsy was taken. Biopsy revealed heavy deposits of myoglobin. Creatinine phosphokinase (CPK) levels drawn ten days later after the admission were found to be elevated at 334 U/dl, presuming the levels would have been much higher during admission. This favored a diagnosis of acute kidney injury (AKI) secondary to exertional rhabdomyolysis. We here describe a case of nontraumatic exertional rhabdomyolysis in a sickle cell trait (SCT) individual that was missed due to findings of microscopic hematuria masking underlying myoglobinuria and fractional excretion of sodium > 3%. As opposed to other causes of ATN, rhabdomyolysis often causes FeNa < 1%. The elevated fractional excretion of sodium in this patient was possibly due to the underlying inability of SCT positive individuals to reabsorb sodium/water and concentrate their urine. Additionally, because of their inability to concentrate urine, SCT positive individuals are prone to intravascular depletion leading to renal failure as seen in this patient. Disease was managed with continuing hydration and tapering steroids. Kidney function improved and the patient was discharged with a creatinine of 3 mg/dL. A month later, renal indices were completely normal with persistence of microscopic hematuria from SCT.
Collapse
|
545
|
Aghakhani Chegeni S, Rahimzadeh M, Montazerghaem H, Khayatian M, Dasturian F, Naderi N. Preliminary Report on the Association Between STAT3 Polymorphisms and Susceptibility to Acute Kidney Injury After Cardiopulmonary Bypass. Biochem Genet 2018; 56:627-638. [PMID: 29846833 DOI: 10.1007/s10528-018-9865-6] [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: 01/08/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023]
Abstract
Cardiopulmonary bypass-associated acute kidney injury (CPB-AKI) is a well-recognized complication which is clearly linked to increased morbidity and mortality. Due to important role of inflammation in CPB-AKI pathogenesis, we explored the association between polymorphisms in STAT3, an inflammation-associated transcription factor, and the risk of CPB-AKI. In this study, STAT3 rs1053004 and rs744166 polymorphisms were analyzed in 129 patients undergoing coronary artery bypass grafting in Jorjani heart center, Bandar Abbas, Iran. The genotypes were determined using sequence-specific primers (PCR-SSP). Sixty-three patients met the criteria for AKI after cardiac surgery (AKI group). The remaining 66 patients did not develop AKI (non-AKI group). Rs1053004 GG genotype was significantly associated with a decreased risk (OR 0.4, 95% CI 0.17-0.9, P = 0.03) of CPB-AKI. Subgroup analyses revealed that GG genotype has also a protective effect in older patients (Age ≥ 60) (OR 0.19, 95% CI 0.04-0.8, P = 0.01). However, rs744166 did not show any difference between AKI and non-AKI groups. The result of our study for the first time provides evidence that rs1053004 polymorphism is significantly associated with a decreased risk of CPB-AKI in Iranian population, especially in older subjects.
Collapse
Affiliation(s)
- Sara Aghakhani Chegeni
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.,Food Health Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahsa Rahimzadeh
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hossein Montazerghaem
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahmood Khayatian
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farzaneh Dasturian
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Nadereh Naderi
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran. .,Department of Immunology, Faculty of Medicine, Hormozgan University of Medical Sciences, EmamHossein Boulevard, P.O. Box: 7919693116, Bandar Abbas, Iran.
| |
Collapse
|
546
|
Zhang W, Sha Y, Wei K, Wu C, Ding D, Yang Y, Zhu C, Zhang Y, Ding G, Zhang A, Jia Z, Huang S. Rotenone ameliorates chronic renal injury caused by acute ischemia/reperfusion. Oncotarget 2018; 9:24199-24208. [PMID: 29849933 PMCID: PMC5966262 DOI: 10.18632/oncotarget.24733] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/25/2018] [Indexed: 01/14/2023] Open
Abstract
Acute kidney injury (AKI) has been widely recognized as an important risk factor leading to the occurrence and progression of chronic kidney disease (CKD). Thus, development of the strategies in retarding the transition of AKI to CKD is becoming a hot research field. Recently, accumulating evidence suggested a pathogenic role of mitochondrial dysfunction in both AKI and CKD. Therefore, in the present study, we evaluated the effect of mitochondrial complex 1 inhibition by rotenone on the chronic renal damage induced by acute ischemia-reperfusion. The mice were treated with 45 min unilateral renal ischemia and reperfusion (I/R) to induce an acute renal injury. After three days of I/R injury, rotenone at a dose of 200 ppm in food was administered to the mice. Strikingly, after three weeks treatment with rotenone, we found that the unilateral I/R-induced tubular damage, tubulointerstitial fibrosis were all attenuated by rotenone as determined by the tubular injury score, Masson staining, and the levels of collagen-I, collagen-III, fibronectin, PAI-1, and TGF-β. Meanwhile, the enhanced inflammatory markers of TNF-α, IL-1β, IL-6, and IL-18 and apoptotic markers of Bax and caspase-3 were all significantly blunted by inhibiting mitochondrial complex-1. Moreover, rotenone treatment also partially protected the mitochondria as shown by the restoration of mitochondrial SOD (SOD2), ATPB, and mitochondrial DNA copy number. These findings suggested that inhibition of mitochondrial complex-1 activity by rotenone could retard the progression of AKI to CKD probably via protecting the mitochondrial function to some extent.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Yugen Sha
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Ke Wei
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Chunfeng Wu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Dan Ding
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Yunwen Yang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Chunhua Zhu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Guixia Ding
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing 210029, China.,Nanjing Key Laboratory of Pediatrics, Nanjing 210008, China
| |
Collapse
|
547
|
Greite R, Thorenz A, Chen R, Jang MS, Rong S, Brownstein MJ, Tewes S, Wang L, Baniassad B, Kirsch T, Bräsen JH, Lichtinghagen R, Meier M, Haller H, Hueper K, Gueler F. Renal ischemia-reperfusion injury causes hypertension and renal perfusion impairment in the CD1 mice which promotes progressive renal fibrosis. Am J Physiol Renal Physiol 2018; 314:F881-F892. [DOI: 10.1152/ajprenal.00519.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a severe complication of major surgery and a risk factor for increased morbidity and mortality. Here, we investigated mechanisms that might contribute to IRI-induced progression to chronic kidney disease (CKD). Acute kidney injury (AKI) was induced by unilateral IRI for 35 min in CD1 and C57BL/6 (B6) mice. Unilateral IRI was used to overcome early mortality. Renal morphology, NGAL upregulation, and neutrophil infiltration as well as peritubular capillary density were studied by immunohistochemistry. The composition of leukocyte infiltrates in the kidney after IRI was investigated by flow cytometry. Systemic blood pressure was measured with a tail cuff, and renal perfusion was quantified by functional magnetic resonance imaging (fMRI). Mesangial matrix expansion was assessed by silver staining. Following IRI, CD1 and B6 mice developed similar morphological signs of AKI and increases in NGAL expression, but neutrophil infiltration was greater in CD1 than B6 mice. IRI induced an increase in systemic blood pressure of 20 mmHg in CD1, but not in B6 mice; and CD1 mice also had a greater loss of renal perfusion and kidney volume than B6 mice ( P < 0.05). CD1 mice developed substantial interstitial fibrosis and decreased peritubular capillary (PTC) density by day 14 while B6 mice showed only mild renal scarring and almost normal PTC. Our results show that after IRI, CD1 mice develop more inflammation, hypertension, and later mesangial matrix expansion than B6 mice do. Subsequently, CD1 animals suffer from CKD due to impaired renal perfusion and pronounced permanent loss of peritubular capillaries.
Collapse
Affiliation(s)
- Robert Greite
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Anja Thorenz
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Rongjun Chen
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Mi-Sun Jang
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Song Rong
- Nephrology, Hannover Medical School, Hannover, Germany
- The Transplantation Center of the Affiliated Hospital, Zunyi Medical College, Zunyi, China
| | | | - Susanne Tewes
- Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany
| | - Li Wang
- Nephrology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | - Martin Meier
- Imaging Center, Institute of Laboratory Animal Sciences, Hannover Medical School, Hannover, Germany
| | | | - Katja Hueper
- Diagnostic and Interventional Radiology, Hannover Medical School, Hannover, Germany
| | - Faikah Gueler
- Nephrology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
548
|
Benoit SW, Devarajan P. Acute kidney injury: emerging pharmacotherapies in current clinical trials. Pediatr Nephrol 2018; 33:779-787. [PMID: 28601936 PMCID: PMC5723563 DOI: 10.1007/s00467-017-3695-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022]
Abstract
Acute kidney injury (AKI) is a significant source of morbidity and mortality in pediatric patients, affecting more than one quarter of critically ill children. Despite significant need, there are no targeted therapies to reliably prevent or treat AKI. Recent advances in our understanding of renal injury and repair signaling pathways have enabled the development of several targeted pharmaceuticals. Here we review emerging pharmacotherapies for AKI that are currently in clinical trials. Categorized by their general mechanism of action, the therapies discussed include anti-inflammatory agents (recAP, AB103, ABT-719), antioxidants (iron chelators, heme arginate), vasodilators (levosimendan), apoptosis inhibitors (QPI-1002), and repair agents (THR-184, BB-3, mesenchymal stem cells).
Collapse
Affiliation(s)
| | - Prasad Devarajan
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7022, Cincinnati, OH, 45229-3039, USA.
| |
Collapse
|
549
|
Regular voluntary running has favorable histological effects on doxorubicin-induced kidney toxicity in Wistar rats. Cell Tissue Res 2018; 374:177-187. [PMID: 29713815 DOI: 10.1007/s00441-018-2840-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 04/10/2018] [Indexed: 12/25/2022]
Abstract
Knowing the therapeutic effects of regular physical exercise on kidney toxicity induced by a single dose of doxorubicin (DOX) in animal models, the aim of this study is to verify the effectiveness of regular voluntary running on kidney histology after a prolonged DOX administration, mimicking a chemotherapy protocol. Thirty-four male Wistar rats were randomly divided into two clusters: DOX (n = 17) and SSS (sterile saline solution, n = 17), receiving a weekly intraperitoneal injection of DOX (2 mg/kg) or vehicle for 7 weeks, respectively. Two weeks after the last injection, five animals from each cluster (SSSG, n = 5; DOXG, n = 5) were euthanized, while the remaining ones were divided into sedentary (DOXsed, n = 6; SSSsed, n = 6) and active subgroups (DOXact, n = 6; SSSact, n = 6). Active animals were placed individually in cages with a running wheel for regular voluntary activity. After 2 months, the animals were euthanized and kidneys were histologically examined. Compared to SSSG, kidneys from DOXG revealed higher levels of damage, more collagen content and thickening of Bowman's capsule (p < .05). The levels of damage and thickness of Bowman's capsule increased in DOXsed as compared to DOXG (p < .05). Compared to DOXsed, the DOXact presented an overall improvement in kidney structure (p < .05), with a decrease in collagen content and of the thickness of Bowman's capsule. The results allow concluding that regular voluntary running attenuate the long-term harmful effects on kidney structure induced by a prolonged DOX treatment. These results, supporting the potential benefit of physical activity in patients under DOX treatment, need to be tested in humans.
Collapse
|
550
|
Andrade L, Rodrigues CE, Gomes SA, Noronha IL. Acute Kidney Injury as a Condition of Renal Senescence. Cell Transplant 2018; 27:739-753. [PMID: 29701108 PMCID: PMC6047270 DOI: 10.1177/0963689717743512] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Acute kidney injury (AKI), characterized by a sharp drop in glomerular filtration, continues to be a significant health burden because it is associated with high initial mortality, morbidity, and substantial health-care costs. There is a strong connection between AKI and mechanisms of senescence activation. After ischemic or nephrotoxic insults, a wide range of pathophysiological events occur. Renal tubular cell injury is characterized by cell membrane damage, cytoskeleton disruption, and DNA degradation, leading to tubular cell death by necrosis and apoptosis. The senescence mechanism involves interstitial fibrosis, tubular atrophy, and capillary rarefaction, all of which impede the morphological and functional recovery of the kidneys, suggesting a strong link between AKI and the progression of chronic kidney disease. During abnormal kidney repair, tubular epithelial cells can assume a senescence-like phenotype. Cellular senescence can occur as a result of cell cycle arrest due to increased expression of cyclin kinase inhibitors (mainly p21), downregulation of Klotho expression, and telomere shortening. In AKI, cellular senescence is aggravated by other factors including oxidative stress and autophagy. Given this scenario, the main question is whether AKI can be repaired and how to avoid the senescence process. Stem cells might constitute a new therapeutic approach. Mesenchymal stem cells (MSCs) can ameliorate kidney injury through angiogenesis, immunomodulation, and fibrosis pathway blockade, as well as through antiapoptotic and promitotic processes. Young umbilical cord–derived MSCs are better at increasing Klotho levels, and thus protecting tissues from senescence, than are adipose-derived MSCs. Umbilical cord–derived MSCs improve glomerular filtration and tubular function to a greater degree than do those obtained from adult tissue. Although senescence-related proteins and microRNA are upregulated in AKI, they can be downregulated by treatment with umbilical cord–derived MSCs. In summary, stem cells derived from young tissues, such as umbilical cord–derived MSCs, could slow the post-AKI senescence process.
Collapse
Affiliation(s)
- Lucia Andrade
- 1 Laboratory of Basic Science LIM-12, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Camila E Rodrigues
- 1 Laboratory of Basic Science LIM-12, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Samirah A Gomes
- 2 Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Irene L Noronha
- 2 Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| |
Collapse
|