251
|
Qu Y, An F, Luo Y, Lu Y, Liu T, Zhao W, Lin B. A nephron model for study of drug-induced acute kidney injury and assessment of drug-induced nephrotoxicity. Biomaterials 2017; 155:41-53. [PMID: 29169037 DOI: 10.1016/j.biomaterials.2017.11.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/05/2017] [Accepted: 11/12/2017] [Indexed: 01/19/2023]
Abstract
In this study, we developed a multilayer microfluidic device to simulate nephron, which was formed by "glomerulus", "Bowman's capsule", "proximal tubular lumen" and "peritubular capillary". In this microdevice, artificial renal blood flow was circulating and glomerular filtrate flow was single passing through, mimicking the behavior of a nephron. In this dynamic artificial nephron, we observed typical renal physiology, including the glomerular size-selective barrier, glomerular basement membrane charge-selective barrier, glucose reabsorption and para-aminohippuric acid secretion. To demonstrate the capability of our microdevice, we used it to investigate the pathophysiology of drug-induced acute kidney injury (AKI) and give assessment of drug-induced nephrotoxicity, with cisplatin and doxorubicin as model drugs. In the experiment, we loaded the doxorubicin or cisplatin in the "renal blood flow", recorded the injury of primary glomerular endothelial cells, podocytes, tubular epithelial cells and peritubular endothelial cells by fluorescence imaging, and identified the time-dependence, dose-dependence and the death order of four types of renal cells. Then by measuring multiple biomarkers, including E-cadherin, VEGF, VCAM-1, Nephrin, and ZO-1, we studied the mechanism of cell injuries caused by doxorubicin or cisplatin. Also, we investigated the effect of BSA in the "renal blood flow" on doxorubicin-or-cisplatin-induced nephrotoxicity, and found that BSA enhanced the tight junctions between cells and eased cisplatin-induced nephrotoxicity. In addition, we compared the nephron model and traditional tubule models for assessment of drug-induced nephrotoxicity. And it can be inferred that our biomimetic microdevice simulated the complex, dynamic microenvironment of nephron, yielded abundant information about drug-induced-AKI at the preclinical stage, boosted the drug safety evaluation, and provided a reliable reference for clinical therapy.
Collapse
Affiliation(s)
- Yueyang Qu
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering & School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Fan An
- Institute of Cancer Stem Cell, Dalian Medical University, China
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering & School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China.
| | - Yao Lu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tingjiao Liu
- College of Stomatology, Dalian Medical University, Dalian, China
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering & School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Bingcheng Lin
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering & School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| |
Collapse
|
252
|
Liu LL, Li D, He YL, Zhou YZ, Gong SH, Wu LY, Zhao YQ, Huang X, Zhao T, Xu L, Wu KW, Li MG, Zhu LL, Fan M. miR-210 protects renal cell against hypoxia-induced apoptosis by targeting HIF-1 alpha. Mol Med 2017; 23:258-271. [PMID: 29387863 DOI: 10.2119/molmed.2017.00013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/21/2017] [Indexed: 01/01/2023] Open
Abstract
The kidney is vulnerable to hypoxia-induced injury. One of the mechanisms underlying this phenomenon is cell apoptosis triggered by hypoxia-inducible factor-1-alpha (HIF-1α) activation. MicroRNA-210 (miR-210) is known to be induced by HIF-1α and can regulate various pathological processes, but its role in hypoxic kidney injury remains unclear. Here, in both kinds of rat systemic hypoxia and local kidney hypoxia models, we found miR-210 levels were upregulated significantly in injured kidney, especially in renal tubular cells. A similar increase was observed in hypoxia-treated human renal tubular HK-2 cells. We also verified that miR-210 can directly suppress HIF-1α expression by targeting the 3' untranslated region (UTR) of HIF-1α mRNA in HK-2 cells in severe hypoxia. Accordingly, miR-210 overexpression caused significant inhibition of the HIF-1α pathway and attenuated apoptosis caused by hypoxia, while miR-210 knockdown exerted the opposite effect. Taken together, our findings verify that miR-210 is involved in the molecular response in hypoxic kidney lesions in vivo and attenuates hypoxia-induced renal tubular cell apoptosis by targeting HIF-1α directly and suppressing HIF-1α pathway activation in vitro.
Collapse
Affiliation(s)
- Li-Li Liu
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China.,Navy Aviation and Diving Medical Center, Navy General Hospital of PLA, Beijing, China
| | - Dahu Li
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Yun-Ling He
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Yan-Zhao Zhou
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Sheng-Hui Gong
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Li-Ying Wu
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Yong-Qi Zhao
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Xin Huang
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Tong Zhao
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Lun Xu
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Kui-Wu Wu
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China
| | - Ming-Gao Li
- Navy Aviation and Diving Medical Center, Navy General Hospital of PLA, Beijing, China
| | - Ling-Ling Zhu
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ming Fan
- Department of Cognitive Science, Institute of Basic Medical Sciences, Beijing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
253
|
Burmeister DM, Gómez BI, Dubick MA. Molecular mechanisms of trauma-induced acute kidney injury: Inflammatory and metabolic insights from animal models. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2661-2671. [DOI: 10.1016/j.bbadis.2017.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/14/2017] [Accepted: 04/10/2017] [Indexed: 12/19/2022]
|
254
|
Chen L, Smith J, Mikl J, Fryer R, Pack F, Williams BJ, Phillips JA, Papov VV. A Multiplatform Approach for the Discovery of Novel Drug-Induced Kidney Injury Biomarkers. Chem Res Toxicol 2017; 30:1823-1834. [PMID: 28885000 DOI: 10.1021/acs.chemrestox.7b00159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug-induced kidney injury (DIKI) is a common toxicity observed in pharmaceutical development. We demonstrated the use of label-free liquid chromatography-mass spectrometry (LC-MS) and multiplex liquid chromatography-single reaction monitoring (LC-SRM) as practical extensions of standard immunoassay based safety biomarker assessments for identification of new toxicity marker candidates and for improved mechanistic understanding. Two different anticancer drugs, doxorubicin (DOX) and cisplatin (cis-diamminedichloridoplatinum, CDDP), were chosen as the toxicants due to their different modes of nephrotoxicity. Analyses of urine samples from toxicant treated and untreated rats were compared to identify biochemical analytes that changed in response to toxicant exposure. A discovery (label-free LC-MS) and targeted proteomics (multiplex LC-SRM) approach was used in combination with well established immunoassay experiments for the identification of a panel of urinary protein markers related to drug induced nephrotoxicity in rats. The initial generation of an expanded set of markers was accomplished using the label-free LC-MS discovery screen and ELISA based analysis of six nephrotoxicity biomarker proteins. Diagnostic performance of the expanded analyte set was statistically compared to conventional nephrotoxicity biomarkers. False discovery rate (FDR) analysis revealed 18 and 28 proteins from the CDDP and DOX groups, respectively, exhibiting significant differences between the vehicle and treated groups. Multiplex SRM assays were constructed to more precisely quantify candidate markers selected from the discovery screen and immunoassay experiments. To evaluate the sensitivity and specificity for each of the candidate biomarkers, histopathology severity scores were used as a benchmark for renal injury followed by receiver-operating characteristic (ROC) curve analysis on selected biomarkers. Further examination of the best performing analytes revealed relevant biological significance after consideration of anatomical localization and functional roles. In summary, the inclusion of mass spectrometry together with conventional ELISA based assays resulted in the identification of an expanded set of biomarkers with a realistic potential for providing additional beneficial information in mechanistic investigations of drug induced kidney injury and with similar responsiveness to conventionally applied indicators of renal injury.
Collapse
Affiliation(s)
- Liuxi Chen
- Boehringer Ingelheim Pharmaceuticals , Ridgefield, Connecticut 06877, United States
| | - James Smith
- Boehringer Ingelheim Pharmaceuticals , Ridgefield, Connecticut 06877, United States
| | - Jaromir Mikl
- Boehringer Ingelheim Pharmaceuticals , Ridgefield, Connecticut 06877, United States
| | - Ryan Fryer
- Boehringer Ingelheim Pharmaceuticals , Ridgefield, Connecticut 06877, United States
| | - Frank Pack
- Boehringer Ingelheim Pharmaceuticals , Ridgefield, Connecticut 06877, United States
| | - Brad J Williams
- Waters Corporation, Milford, Massachusetts 01757, United States
| | - Jonathan A Phillips
- Boehringer Ingelheim Pharmaceuticals , Ridgefield, Connecticut 06877, United States
| | - Vladimir V Papov
- Boehringer Ingelheim Pharmaceuticals , Ridgefield, Connecticut 06877, United States
| |
Collapse
|
255
|
Liu J, Kumar S, Dolzhenko E, Alvarado GF, Guo J, Lu C, Chen Y, Li M, Dessing MC, Parvez RK, Cippà PE, Krautzberger AM, Saribekyan G, Smith AD, McMahon AP. Molecular characterization of the transition from acute to chronic kidney injury following ischemia/reperfusion. JCI Insight 2017; 2:94716. [PMID: 28931758 PMCID: PMC5612583 DOI: 10.1172/jci.insight.94716] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/10/2017] [Indexed: 12/16/2022] Open
Abstract
Though an acute kidney injury (AKI) episode is associated with an increased risk of chronic kidney disease (CKD), the mechanisms determining the transition from acute to irreversible chronic injury are not well understood. To extend our understanding of renal repair, and its limits, we performed a detailed molecular characterization of a murine ischemia/reperfusion injury (IRI) model for 12 months after injury. Together, the data comprising RNA-sequencing (RNA-seq) analysis at multiple time points, histological studies, and molecular and cellular characterization of targeted gene activity provide a comprehensive profile of injury, repair, and long-term maladaptive responses following IRI. Tubular atrophy, interstitial fibrosis, inflammation, and development of multiple renal cysts were major long-term outcomes of IRI. Progressive proximal tubular injury tracks with de novo activation of multiple Krt genes, including Krt20, a biomarker of renal tubule injury. RNA-seq analysis highlights a cascade of temporal-specific gene expression patterns related to tubular injury/repair, fibrosis, and innate and adaptive immunity. Intersection of these data with human kidney transplant expression profiles identified overlapping gene expression signatures correlating with different stages of the murine IRI response. The comprehensive characterization of incomplete recovery after ischemic AKI provides a valuable resource for determining the underlying pathophysiology of human CKD.
Collapse
Affiliation(s)
- Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Sanjeev Kumar
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Egor Dolzhenko
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Gregory F Alvarado
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Can Lu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Meng Li
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Mark C Dessing
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Pietro E Cippà
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - A Michaela Krautzberger
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Gohar Saribekyan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Andrew D Smith
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
256
|
Al-Saleh I, Al-Rouqi R, Elkhatib R, Abduljabbar M, Al-Rajudi T. Risk assessment of environmental exposure to heavy metals in mothers and their respective infants. Int J Hyg Environ Health 2017; 220:1252-1278. [PMID: 28869188 DOI: 10.1016/j.ijheh.2017.07.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/26/2017] [Accepted: 07/31/2017] [Indexed: 12/17/2022]
Abstract
Exposure to heavy metals can cause renal injury, which has been well documented in occupational exposure. Studies of low exposure in the general population, however, are still scarce, particularly for vulnerable populations such as mothers and young children. This study evaluated exposure to heavy metals, and biomarkers of renal function and oxidative stress in 944 lactating mothers and their infants and investigated the role of the interaction between heavy metals and oxidative stress in altering renal function. Mother and infant urine samples were analyzed to measure mercury (Hg), cadmium (Cd), and lead (Pb) concentrations for determining body-burden exposure; N-acetyl-β-d-glucosaminidase (NAG), α1-microglobulin (α1-MG), albumin (ALB), and creatinine (Cr) concentrations for determining early renal injury; and 8-hydroxy-2-deoxyguanosine (8-OHdG) and malondialdehyde (MDA) concentrations for determining oxidative stress. The median concentrclearlyations in mothers presented as μg/g Cr (infants as μg/l) for Hg, Cd, and Pb were 0.695 (0.716), 0.322 (0.343), and 3.97 (5.306) respectively. The mothers and their infants had clearly been exposed to heavy metals and had levels higher than the reference values reported for the general populations of USA, Germany, and Canada. Multiple regression analyses clearly demonstrated associations between urinary heavy metals in quartiles and several renal and oxidative biomarkers in mothers and to a lesser extent their infants. ß coefficients for urinary excretions of MDA, 8-OHdG, ALB, α1-MG, NAG, and Cr in mothers were high in the highest quartile of Hg (1.183-51.29μg/g Cr or 1.732-106.95μg/l), Cd (0.565-765.776μg/g Cr or 0.785-1347.0μg/l), and Pb (6.606-83.937μg/g Cr or 9.459-80.826μg/l), except Pb was not associated with ALB. Infants in the highest Pb quartile (9.293-263.098μg/l) had the highest ß coefficients of urinary excretion of MDA, 8-OHdG, ALB, NAG, and Cr. Significant increasing trend in biomarkers across the quartiles of the three metals was seen in both mothers and infants (ptrend <0.001). A receiver operating characteristic analysis supported the predictive abilities of the four renal biomarkers in discriminating between low versus high metal quartiles. The interaction between heavy metals and oxidative stress contributed to the high excretions of renal biomarkers, but the mechanism remains unclear. These findings add to the limited evidence that low exposure to heavy metals in the general population is associated with alterations in renal function that could eventually progress to renal damage if exposure continues and that children are more susceptible due to the immaturity of their body organs.
Collapse
Affiliation(s)
- Iman Al-Saleh
- Environmental Health Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O. Box: 3354, Riyadh 11211, Saudi Arabia.
| | - Reem Al-Rouqi
- Environmental Health Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O. Box: 3354, Riyadh 11211, Saudi Arabia
| | - Rola Elkhatib
- Environmental Health Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O. Box: 3354, Riyadh 11211, Saudi Arabia
| | - Mai Abduljabbar
- Environmental Health Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O. Box: 3354, Riyadh 11211, Saudi Arabia
| | - Tahreer Al-Rajudi
- Environmental Health Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O. Box: 3354, Riyadh 11211, Saudi Arabia
| |
Collapse
|
257
|
Kim SS, Song SH, Kim JH, Jeon YK, Kim BH, Kang MC, Chun SW, Hong SH, Chung M, Kim YK, Kim J, Kim YB. Urine clusterin/apolipoprotein J is linked to tubular damage and renal outcomes in patients with type 2 diabetes mellitus. Clin Endocrinol (Oxf) 2017; 87:156-164. [PMID: 28434209 PMCID: PMC5511063 DOI: 10.1111/cen.13360] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/03/2017] [Accepted: 04/13/2017] [Indexed: 01/18/2023]
Abstract
OBJECTIVE The objective of this study was to evaluate the association of urine clusterin/apolipoprotein J (Apo J) with the development and/or progression of diabetic kidney disease (DKD) in type 2 diabetes. MATERIALS AND METHODS A total of 159 type 2 diabetic patients and 20 nondiabetic subjects with estimated glomerular filtration rate (eGFR) ≥60 mL/min/1.73 m2 were enrolled. The baseline values of urine clusterin and tubular damage markers were measured. The primary outcome was the annual decline rate in eGFR, and secondary outcomes were the development of chronic kidney disease (CKD) stage 3 or greater and the persistence/progression of albuminuria. The median follow-up duration of enrolled patients was 3.0 (1.0-5.9) years. RESULTS Baseline clusterin levels in urine were significantly increased in type 2 diabetic subjects compared with those of nondiabetic subjects. The levels of urine clusterin had a significant correlation with urine tubular damage markers. A positive correlation between the annual rate of decline in eGFR and urine clusterin after adjusting for clinical confounding factors was detected. Multivariate analysis further indicated that urine clusterin correlated with the development of CKD stage 3 or greater and persistence/progression of albuminuria. In type 2 diabetic subjects with albuminuria, urine clusterin remained associated with the annual decline rate in eGFR and the progression of CKD stage. CONCLUSIONS Urine clusterin reflects tubular damage in the early stage of DKD. The increase in urine clusterin along with albuminuria could be an independent predictive marker for the progression of DKD in type 2 diabetes.
Collapse
Affiliation(s)
- Sang Soo Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel, Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Jong Ho Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Yun Kyung Jeon
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Bo Hyun Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Min-Cheol Kang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel, Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts
| | - Sung Wan Chun
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel, Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts
| | - Soo Hyun Hong
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel, Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts
| | - Michelle Chung
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel, Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts
| | - Yong Ki Kim
- Kim Yong Ki Internal Medicine Clinic, Busan, Korea
| | - Joo Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel, Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
258
|
Singh M, Tong Y, Webster K, Cesewski E, Haring AP, Laheri S, Carswell B, O'Brien TJ, Aardema CH, Senger RS, Robertson JL, Johnson BN. 3D printed conformal microfluidics for isolation and profiling of biomarkers from whole organs. LAB ON A CHIP 2017. [PMID: 28632265 DOI: 10.1039/c7lc00468k] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The ability to interface microfluidic devices with native complex biological architectures, such as whole organs, has the potential to shift the paradigm for the study and analysis of biological tissue. Here, we show 3D printing can be used to fabricate bio-inspired conformal microfluidic devices that directly interface with the surface of whole organs. Structured-light scanning techniques enabled the 3D topographical matching of microfluidic device geometry to porcine kidney anatomy. Our studies show molecular species are spontaneously transferred from the organ cortex to the conformal microfluidic device in the presence of fluid flow through the organ-conforming microchannel. Large animal studies using porcine kidneys (n = 32 organs) revealed the profile of molecular species in the organ-conforming microfluidic stream was dependent on the organ preservation conditions. Enzyme-linked immunosorbent assay (ELISA) studies revealed conformal microfluidic devices isolate clinically relevant metabolic and pathophysiological biomarkers from whole organs, including heat shock protein 70 (HSP-70) and kidney injury molecule-1 (KIM-1), which were detected in the microfluidic device as high as 409 and 12 pg mL-1, respectively. Overall, these results show conformal microfluidic devices enable a novel minimally invasive 'microfluidic biopsy' technique for isolation and profiling of biomarkers from whole organs within a clinically relevant interval. This achievement could shift the paradigm for whole organ preservation and assessment, thereby helping to relieve the organ shortage crisis through increased availability and quality of donor organs. Ultimately, this work provides a major advance in microfluidics through the design and manufacturing of organ-conforming microfluidic devices and a novel technique for microfluidic-based analysis of whole organs.
Collapse
Affiliation(s)
- Manjot Singh
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, VA 24061 USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
259
|
Jacobo-Estrada T, Santoyo-Sánchez M, Thévenod F, Barbier O. Cadmium Handling, Toxicity and Molecular Targets Involved during Pregnancy: Lessons from Experimental Models. Int J Mol Sci 2017; 18:ijms18071590. [PMID: 28737682 PMCID: PMC5536077 DOI: 10.3390/ijms18071590] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/16/2022] Open
Abstract
Even decades after the discovery of Cadmium (Cd) toxicity, research on this heavy metal is still a hot topic in scientific literature: as we wrote this review, more than 1440 scientific articles had been published and listed by the PubMed.gov website during 2017. Cadmium is one of the most common and harmful heavy metals present in our environment. Since pregnancy is a very particular physiological condition that could impact and modify essential pathways involved in the handling of Cd, the prenatal life is a critical stage for exposure to this non-essential element. To give the reader an overview of the possible mechanisms involved in the multiple organ toxic effects in fetuses after the exposure to Cd during pregnancy, we decided to compile some of the most relevant experimental studies performed in experimental models and to summarize the advances in this field such as the Cd distribution and the factors that could alter it (diet, binding-proteins and membrane transporters), the Cd-induced toxicity in dams (preeclampsia, fertility, kidney injury, alteration in essential element homeostasis and bone mineralization), in placenta and in fetus (teratogenicity, central nervous system, liver and kidney).
Collapse
Affiliation(s)
- Tania Jacobo-Estrada
- Departamento de Sociedad y Política Ambiental, CIIEMAD, Instituto Politécnico Nacional, 30 de Junio de 1520 s/n, La Laguna Ticomán, Ciudad de México 07340, Mexico.
| | - Mitzi Santoyo-Sánchez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, San Pedro Zacatenco, Ciudad de México 07360, Mexico.
| | - Frank Thévenod
- Department of Physiology, Pathophysiology & Toxicology and ZBAF (Centre for Biomedical Education and Research), Faculty of Health-School of Medicine, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), D 58453 Witten, Germany.
| | - Olivier Barbier
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, San Pedro Zacatenco, Ciudad de México 07360, Mexico.
| |
Collapse
|
260
|
Far-Forward Diagnostics in Toxic Industrial Chemical and Material Exposure Scenarios and Biomarker Identification. J Occup Environ Med 2017; 59:e204-e208. [PMID: 28692011 DOI: 10.1097/jom.0000000000001083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
: This study describes key technical solutions for detecting environmental toxicants and diagnosing adverse health effects in military operational settings as outlined at a symposium cosponsored by the Department of Defense and the Johns Hopkins University-Applied Physics Laboratory (October 27 to 28, 2015). Such technologies are urgently needed in order to provide critical decision-aid tools and prognostic assessment of potential clinical sequelae. This review summarizes the state-of-the-science on (1) prioritization of adverse health effects, (2) existing technologies and diagnostic tools available for use in theater, (3) challenges to advancing diagnostic tools far-forward, and (4) the potential utility of anchoring diagnostic tools to adverse outcome pathways. Emerging technologies are increasingly available for physiological, environmental, and individual exposure monitoring. Challenges to overcome in austere environments include cold chain requirements and determination of adequate sampling intervals.
Collapse
|
261
|
Relizani K, Griffith G, Echevarría L, Zarrouki F, Facchinetti P, Vaillend C, Leumann C, Garcia L, Goyenvalle A. Efficacy and Safety Profile of Tricyclo-DNA Antisense Oligonucleotides in Duchenne Muscular Dystrophy Mouse Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:144-157. [PMID: 28918017 PMCID: PMC5498286 DOI: 10.1016/j.omtn.2017.06.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/16/2017] [Accepted: 06/17/2017] [Indexed: 12/30/2022]
Abstract
Antisense oligonucleotides (AONs) hold promise for therapeutic splice-switching correction in many genetic diseases. However, despite advances in AON chemistry and design, systemic use of AONs is limited due to poor tissue uptake and sufficient therapeutic efficacy is still difficult to achieve. A novel class of AONs made of tricyclo-DNA (tcDNA) is considered very promising for the treatment of Duchenne muscular dystrophy (DMD), a neuromuscular disease typically caused by frameshifting deletions or nonsense mutations in the gene-encoding dystrophin and characterized by progressive muscle weakness, cardiomyopathy, and respiratory failure in addition to cognitive impairment. Herein, we report the efficacy and toxicology profile of a 13-mer tcDNA in mdx mice. We show that systemic delivery of 13-mer tcDNA allows restoration of dystrophin in skeletal muscles and to a lower extent in the brain, leading to muscle function improvement and correction of behavioral features linked to the emotional/cognitive deficiency. More importantly, tcDNA treatment was generally limited to minimal glomerular changes and few cell necroses in proximal tubules, with only slight variation in serum and urinary kidney toxicity biomarker levels. These results demonstrate an encouraging safety profile for tcDNA, albeit typical of phosphorothiate AONs, and confirm its therapeutic potential for the systemic treatment of DMD patients.
Collapse
Affiliation(s)
- Karima Relizani
- Université de Versailles Saint-Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France; SQY Therapeutics, UFR des Sciences de la Santé, Université de Versailles Saint-Quentin en Yvelines, 78180 Montigny-le-Bretonneux, France
| | - Graziella Griffith
- Université de Versailles Saint-Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France; SQY Therapeutics, UFR des Sciences de la Santé, Université de Versailles Saint-Quentin en Yvelines, 78180 Montigny-le-Bretonneux, France
| | - Lucía Echevarría
- Université de Versailles Saint-Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France
| | - Faouzi Zarrouki
- Université de Versailles Saint-Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France; Neuro-PSI, UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Patricia Facchinetti
- Université de Versailles Saint-Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France
| | - Cyrille Vaillend
- Neuro-PSI, UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Christian Leumann
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Luis Garcia
- Université de Versailles Saint-Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France.
| | - Aurélie Goyenvalle
- Université de Versailles Saint-Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France.
| |
Collapse
|
262
|
Abstract
More than 4 decades after the creation of the Brooke and Parkland formulas, burn practitioners still argue about which formula is the best. So it is no surprise that there is no consensus about how to resuscitate a thermally injured patient with a significant comorbidity such as heart failure or cirrhosis or how to resuscitate a patient after an electrical or inhalation injury or a patient whose resuscitation is complicated by renal failure. All of these scenarios share a common theme in that the standard rule book does not apply. All will require highly individualized resuscitations.
Collapse
Affiliation(s)
- David T Harrington
- Rhode Island Burn Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 444, Providence, RI 02903, USA.
| |
Collapse
|
263
|
Abstract
Acute kidney injury (AKI) is characterized by an acute decline in renal function and is associated to increased mortality rate, hospitalization time, and total health-related costs. The severity of this ‘fearsome’ clinical complication might depend on, or even be worsened by, the late detection of AKI, when the diagnosis is based on the elevation of serum creatinine (SCr). For these reasons, in recent years a great number of new tools, biomarkers and predictive models have been proposed to clinicians in order to improve diagnosis and prevent the development of AKI. The purpose of this narrative paper is to review the current state of the art in prediction and early detection of AKI and outline future challenges.
Collapse
Affiliation(s)
- Simona Pozzoli
- Chair of Nephrology - IRCCS San Raffaele Scientific Institute, Genomics of Renal Diseases and Hypertension Unit, Università Vita Salute San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Marco Simonini
- Chair of Nephrology - IRCCS San Raffaele Scientific Institute, Genomics of Renal Diseases and Hypertension Unit, Università Vita Salute San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| | - Paolo Manunta
- Chair of Nephrology - IRCCS San Raffaele Scientific Institute, Genomics of Renal Diseases and Hypertension Unit, Università Vita Salute San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
264
|
A comprehensive analysis and annotation of human normal urinary proteome. Sci Rep 2017; 7:3024. [PMID: 28596590 PMCID: PMC5465101 DOI: 10.1038/s41598-017-03226-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/25/2017] [Indexed: 12/14/2022] Open
Abstract
Biomarkers are measurable changes associated with the disease. Urine can reflect the changes of the body while blood is under control of the homeostatic mechanisms; thus, urine is considered an important source for early and sensitive disease biomarker discovery. A comprehensive profile of the urinary proteome will provide a basic understanding of urinary proteins. In this paper, we present an in-depth analysis of the urinary proteome based on different separation strategies, including direct one dimensional liquid chromatography–tandem mass spectrometry (LC/MS/MS), two dimensional LC/MS/MS, and gel-eluted liquid fraction entrapment electrophoresis/liquid-phase isoelectric focusing followed by two dimensional LC/MS/MS. A total of 6085 proteins were identified in healthy urine, of which 2001 were not reported in previous studies and the concentrations of 2571 proteins were estimated (spanning a magnitude of 106) with an intensity-based absolute quantification algorithm. The urinary proteins were annotated by their tissue distribution. Detailed information can be accessed at the “Human Urine Proteome Database” (www.urimarker.com/urine).
Collapse
|
265
|
Datta R, Wong A, Camarata T, Tamanna F, Ilahi I, Vasilyev A. Precise Cellular Ablation Approach for Modeling Acute Kidney Injury in Developing Zebrafish. J Vis Exp 2017. [PMID: 28605371 DOI: 10.3791/55606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Acute Kidney Injury (AKI) is a common medical condition with a high mortality rate. With the repair abilities of the kidney, it is possible to restore adequate kidney function after supportive treatment. However, a better understanding of how nephron cell death and repair occur on the cellular level is required to minimize cell death and to enhance the regenerative process. The zebrafish pronephros is a good model system to accomplish this goal because it contains anatomical segments that are similar to the mammalian nephron. Previously, the most common model used to study kidney injury in fish was the pharmacological gentamicin model. However, this model does not allow for precise spatiotemporal control of injury, and hence it is difficult to study cellular and molecular processes involved in kidney repair. To overcome this limitation, this work presents a method through which, in contrast to the gentamicin approach, a specific Green Fuorescent Protein (GFP)-expressing nephron segment can be photoablated using a violet laser light (405 nm). This novel model of AKI provides many advantages that other methods of epithelial injury lack. Its main advantages are the ability to "dial" the level of injury and the precise spatiotemporal control in the robust in vivo animal model. This new method has the potential to significantly advance the level of understanding of kidney injury and repair mechanisms.
Collapse
Affiliation(s)
| | - Ada Wong
- Department of Biomedical Sciences, NYITCOM
| | | | | | | | | |
Collapse
|
266
|
Mesenchymal stem cells correct haemodynamic dysfunction associated with liver injury after extended resection in a pig model. Sci Rep 2017; 7:2617. [PMID: 28572613 PMCID: PMC5454025 DOI: 10.1038/s41598-017-02670-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/13/2017] [Indexed: 12/15/2022] Open
Abstract
In patients, acute kidney injury (AKI) is often due to haemodynamic impairment associated with hepatic decompensation following extended liver surgery. Mesenchymal stem cells (MSCs) supported tissue protection in a variety of acute and chronic diseases, and might hence ameliorate AKI induced by extended liver resection. Here, 70% liver resection was performed in male pigs. MSCs were infused through a central venous catheter and haemodynamic parameters as well as markers of acute kidney damage were monitored under intensive care conditions for 24 h post-surgery. Cytokine profiles were established to anticipate the MSCs’ potential mode of action. After extended liver resection, hyperdynamic circulation, associated with hyponatraemia, hyperkalaemia, an increase in serum aldosterone and low urine production developed. These signs of hepatorenal dysfunction and haemodynamic impairment were corrected by MSC treatment. MSCs elevated PDGF levels in the serum, possibly contributing to circulatory homeostasis. Another 14 cytokines were increased in the kidney, most of which are known to support tissue regeneration. In conclusion, MSCs supported kidney and liver function after extended liver resection. They probably acted through paracrine mechanisms improving haemodynamics and tissue homeostasis. They might thus provide a promising strategy to prevent acute kidney injury in the context of post-surgery acute liver failure.
Collapse
|
267
|
Guha M, Nguyen L, Poitout-Belissent F, Bedard A, Raman K. Salt Selection Matters: Differential Renal Toxicity With MDV1634.Maleate Versus MDV1634.2HCl. Int J Toxicol 2017; 36:207-219. [PMID: 28466691 DOI: 10.1177/1091581817704379] [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] [Indexed: 11/17/2022]
Abstract
Salt forms of pharmaceutical compounds can have unique pharmacokinetic and toxicity properties. MDV1634 was evaluated for neurology indication and also demonstrated blood pressure (BP)-lowering effects in nonclinical studies. During the chemistry manufacturing campaign, 2 salt forms, dihydrochloride (2HCl) and maleate (MAL), which improved chemical stability and water solubility of the free base were identified. MDV1634.MAL showed better chemical attributes and was evaluated in toxicology studies for further development. A 28-day oral toxicity study in dogs with MDV1634.MAL demonstrated partially reversible renal toxicity. Although MAL salt is generally regarded as safe, renal toxicity is sometimes observed in rats and dogs. To evaluate contribution of each salt form to renal toxicity and BP lowering, an additional 28-day study was conducted with MDV1634.2HCL and MDV1634.MAL, which included toxicokinetics, continuous BP measurement in a subset of dogs, and sensitive urinary biomarker evaluation for temporal monitorability and reversibility of potential renal findings. In the repeat study, both salt forms showed similar exposures during the dosing period, but renal tubular toxicity was observed only with MDV1634.MAL and not with MDV1634.2HCl. The renal findings with MDV1634.MAL included early urinary biomarker changes (increase in albumin, clusterin, β2 microglobulin, and neutrophil gelatinase-associated lipocalin); elevations in serum blood urea nitrogen and creatinine; and microscopic findings of partially reversible tubular basophilia, single cell necrosis, pigmentation, and mineralization. The renal findings in contrast to the BP findings were MAL-specific and considered not related to MDV1634, thereby under scoring the importance of salt forms in pharmaceutical development.
Collapse
Affiliation(s)
| | | | | | - Agathe Bedard
- 2 Charles River Laboratories Montreal, Senneville, Québec, Canada
| | | |
Collapse
|
268
|
Quesada A, Segarra AB, Montoro-Molina S, de Gracia MDC, Osuna A, O’Valle F, Gómez-Guzmán M, Vargas F, Wangensteen R. Glutamyl aminopeptidase in microvesicular and exosomal fractions of urine is related with renal dysfunction in cisplatin-treated rats. PLoS One 2017; 12:e0175462. [PMID: 28399178 PMCID: PMC5388492 DOI: 10.1371/journal.pone.0175462] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 03/27/2017] [Indexed: 12/23/2022] Open
Abstract
PURPOSE The aim of this work was to investigate if the content of glutamyl aminopeptidase (GluAp) in microvesicular and exosomal fractions of urine is related with renal dysfunction in cisplatin-treated rats. METHODS Urine samples were collected 24 hours after injection of cisplatin (7 mg/kg, n = 10) or saline serum (n = 10), and they were subjected to differential centrifugation at 1.000, 17.000 and 200.000 g to obtain microvesicular and exosomal fractions. GluAp was measured with a commercial ELISA kit in both fractions. Serum creatinine (SCr) and body weight were measured 15 days after treatment. We analyzed if early excretion of GluAp in microsomal and exosomal fractions was correlated with final SCr and body weight increase. In a second experiment, enzymatic activities of GluAp and alanyl aminopeptidase (AlaAp) in urine, microvesicular and exosomal fractions were measured three days after injection. We analyzed the correlation of both markers with SCr determined at this point. Finally, we studied the expression of GluAp and extracellular vesicles markers Alix and tumor susceptibility gene (TSG101) in both fractions by immunoblotting. RESULTS GluAp excretion was increased in all fractions of urine after cisplatin treatment, even if data were normalized per mg of creatinine, per body weight or per total protein content of each fraction. We found significant predictive correlations with SCr concentration, and inverse correlations with body weight increase determined 15 days later. Three days after injection, aminopeptidasic activities were markedly increased in all fractions of urine in cisplatin-treated rats. The highest correlation coefficient with SCr was found for GluAp in microvesicular fraction. Increase of GluAp in microvesicular and exosomal fractions from cisplatin-treated rats was confirmed by immunoblotting. Alix and TSG101 showed different patterns of expression in each fraction. CONCLUSIONS Determination of GluAp content or its enzymatic activity in microvesicular and exosomal fractions of urine is an early and predictive biomarker of renal dysfunction in cisplatin-induced nephrotoxicity. Measurement of GluAp in these fractions can serve to detect proximal tubular damage independently of glomerular filtration status.
Collapse
Affiliation(s)
- Andrés Quesada
- Unidad de Nefrología, Hospital Virgen de las Nieves, FIBAO, Granada, Spain
| | - Ana Belén Segarra
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | | | | | - Antonio Osuna
- Unidad de Nefrología, Hospital Virgen de las Nieves, FIBAO, Granada, Spain
| | - Francisco O’Valle
- Departamento de Anatomía Patológica, IBIMER, Universidad de Granada, Granada, Spain
| | - Manuel Gómez-Guzmán
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Félix Vargas
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Rosemary Wangensteen
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| |
Collapse
|
269
|
Lee EK, Shin YJ, Park EY, Kim ND, Moon A, Kwack SJ, Son JY, Kacew S, Lee BM, Bae ON, Kim HS. Selenium-binding protein 1: a sensitive urinary biomarker to detect heavy metal-induced nephrotoxicity. Arch Toxicol 2017; 91:1635-1648. [PMID: 27578022 DOI: 10.1007/s00204-016-1832-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/24/2016] [Indexed: 11/24/2022]
Abstract
Identifying novel biomarkers to detect nephrotoxicity is clinically important. Here, we attempted to identify new biomarkers for mercury-induced nephrotoxicity and compared their sensitivity to that of traditional biomarkers in animal models. Comparative proteomics analysis was performed in kidney tissues of Sprague-Dawley rats after oral treatment with HgCl2 (0.1, 1, or 5 mg/kg/day) for 21 days. Kidney cortex tissues were analyzed by two-dimensional gel electrophoresis/matrix-assisted laser desorption/ionization, and differentially expressed proteins were identified. The corresponding spots were quantitated by RT-PCR. Selenium-binding protein 1 (SBP1) was found to be the most markedly upregulated protein in the kidney cortex of rats after HgCl2 administration. However, blood urea nitrogen, serum creatinine, and glucose levels increased significantly only in the 1 or 5 mg/kg HgCl2-treated groups. A number of urinary excretion proteins, including kidney injury molecule-1, clusterin, monocyte chemoattractant protein-1, and β-microglobulin, increased dose-dependently. Histopathological examination revealed severe proximal tubular damage in high-dose (5 mg/kg) HgCl2-exposed groups. In addition, urinary excretion of SBP1 significantly increased in a dose-dependent manner. To confirm the critical role of SBP1 as a biomarker for nephrotoxicity, normal kidney proximal tubular cells were treated with HgCl2, CdCl2, or cisplatin for 24 h. SBP1 levels significantly increased in conditioned media exposed to nephrotoxicants, but decreased in cell lysates. Our investigations suggest that SBP1 may play a critical role in the pathological processes underlying chemical-induced nephrotoxicity. Thus, urinary excretion of SBP1 might be a sensitive and specific biomarker to detect early stages of kidney injury.
Collapse
Affiliation(s)
- Eui Kyung Lee
- College of Pharmacy, Pusan National University, Busan, 609-735, Republic of Korea
| | - Young-Jun Shin
- College of Pharmacy, Hanyang University, Ansan, 426-791, Republic of Korea
| | - Eun Young Park
- College of Pharmacy, Pusan National University, Busan, 609-735, Republic of Korea
| | - Nam Deuk Kim
- College of Pharmacy, Pusan National University, Busan, 609-735, Republic of Korea
| | - Aree Moon
- College of Pharmacy, Duksung Women's University, Seoul, 132-714, Republic of Korea
| | - Seung Jun Kwack
- Department of Biochemistry and Health Science, Changwon National University, Gyeongnam, 641-773, Republic of Korea
| | - Ji Yeon Son
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Sam Kacew
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON, Canada
| | - Byung Mu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Ok-Nam Bae
- College of Pharmacy, Hanyang University, Ansan, 426-791, Republic of Korea.
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Republic of Korea.
| |
Collapse
|
270
|
Krogstrup NV, Oltean M, Nieuwenhuijs-Moeke GJ, Dor FJMF, Møldrup U, Krag SP, Bibby BM, Birn H, Jespersen B. Remote Ischemic Conditioning on Recipients of Deceased Renal Transplants Does Not Improve Early Graft Function: A Multicenter Randomized, Controlled Clinical Trial. Am J Transplant 2017; 17:1042-1049. [PMID: 27696662 DOI: 10.1111/ajt.14075] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 01/25/2023]
Abstract
Delayed graft function is a frequent complication following deceased donor renal transplantation, and is closely related to ischemia-reperfusion injury. Experimental and clinical studies have shown protection by remote ischemic conditioning (RIC). We hypothesized that recipient RIC before kidney graft reperfusion reduces the time to graft recovery. This multicenter, blinded, randomized, controlled clinical trial included 225 adult recipients of renal transplants from deceased donors at four transplantation centers in Denmark, Sweden, and the Netherlands. Participants were randomized 1:1 to RIC or sham-RIC. RIC consisted of 4 × 5-min thigh occlusion by an inflatable tourniquet each followed by 5-min deflation, performed during surgery prior to graft reperfusion. The tourniquet remained deflated for sham-RIC. The primary endpoint was the estimated time to a 50% decrease in baseline plasma creatinine (tCr50) calculated from plasma creatinine measurements 30 days posttransplant or 30 days after the last, posttransplant dialysis. No significant differences were observed between RIC and sham-RIC-treated patients in the primary outcome median tCr50 (122 h [95% confidence interval [CI] 98-151] vs. 112 h [95% CI 91-139], p = 0.58), or the number of patients receiving dialysis in the first posttransplant week (33% vs. 35%, p = 0.71). Recipient RIC does not reduce the time to graft recovery in kidney transplantation from deceased donors. ClinicalTrials.gov: NCT01395719.
Collapse
Affiliation(s)
- N V Krogstrup
- Department of Renal Medicine, Aarhus University, Aarhus, Denmark.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - M Oltean
- The Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - G J Nieuwenhuijs-Moeke
- Department of Anaesthesiology, University Medical Center Groningen, Groningen, the Netherlands
| | - F J M F Dor
- Division of HPB & Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - U Møldrup
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - S P Krag
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - B M Bibby
- Department of Biostatistics, Aarhus University, Aarhus, Denmark
| | - H Birn
- Department of Renal Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - B Jespersen
- Department of Renal Medicine, Aarhus University, Aarhus, Denmark.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
271
|
King SM, Higgins JW, Nino CR, Smith TR, Paffenroth EH, Fairbairn CE, Docuyanan A, Shah VD, Chen AE, Presnell SC, Nguyen DG. 3D Proximal Tubule Tissues Recapitulate Key Aspects of Renal Physiology to Enable Nephrotoxicity Testing. Front Physiol 2017; 8:123. [PMID: 28337147 PMCID: PMC5340751 DOI: 10.3389/fphys.2017.00123] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 02/15/2017] [Indexed: 01/09/2023] Open
Abstract
Due to its exposure to high concentrations of xenobiotics, the kidney proximal tubule is a primary site of nephrotoxicity and resulting attrition in the drug development pipeline. Current pre-clinical methods using 2D cell cultures and animal models are unable to fully recapitulate clinical drug responses due to limited in vitro functional lifespan, or species-specific differences. Using Organovo's proprietary 3D bioprinting platform, we have developed a fully cellular human in vitro model of the proximal tubule interstitial interface comprising renal fibroblasts, endothelial cells, and primary human renal proximal tubule epithelial cells to enable more accurate prediction of tissue-level clinical outcomes. Histological characterization demonstrated formation of extensive microvascular networks supported by endogenous extracellular matrix deposition. The epithelial cells of the 3D proximal tubule tissues demonstrated tight junction formation and expression of renal uptake and efflux transporters; the polarized localization and function of P-gp and SGLT2 were confirmed. Treatment of 3D proximal tubule tissues with the nephrotoxin cisplatin induced loss of tissue viability and epithelial cells in a dose-dependent fashion, and cimetidine rescued these effects, confirming the role of the OCT2 transporter in cisplatin-induced nephrotoxicity. The tissues also demonstrated a fibrotic response to TGFβ as assessed by an increase in gene expression associated with human fibrosis and histological verification of excess extracellular matrix deposition. Together, these results suggest that the bioprinted 3D proximal tubule model can serve as a test bed for the mechanistic assessment of human nephrotoxicity and the development of pathogenic states involving epithelial-interstitial interactions, making them an important adjunct to animal studies.
Collapse
|
272
|
Deng Y, Chi R, Chen S, Ye H, Yuan J, Wang L, Zhai Y, Gao L, Zhang D, Hu L, Lv B, Long Y, Sun C, Yang X, Zou X, Chen C. Evaluation of clinically available renal biomarkers in critically ill adults: a prospective multicenter observational study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:46. [PMID: 28264714 PMCID: PMC5339963 DOI: 10.1186/s13054-017-1626-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/09/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Although serum cystatin C (sCysC), urinary N-acetyl-β-D-glucosaminidase (uNAG), and urinary albumin/creatinine ratio (uACR) are clinically available, their optimal combination for acute kidney injury (AKI) detection and prognosis prediction remains unclear. We aimed to assess the discriminative abilities of these biomarkers and their possible combinations for AKI detection and intensive care unit (ICU) mortality prediction in critically ill adults. METHODS A multicenter, prospective observational study was conducted in mixed medical-surgical ICUs at three tertiary care hospitals. One thousand eighty-four adult critically ill patients admitted to the ICUs were studied. We assessed the use of individual biomarkers (sCysC, uNAG, and uACR) measured at ICU admission and their combinations with regard to AKI detection and prognosis prediction. RESULTS AUC-ROCs for sCysC, uNAG, and uACR were calculated for total AKI (0.738, 0.650, and 0.683, respectively), severe AKI (0.839, 0.706, and 0.771, respectively), and ICU mortality (0.727, 0.793, and 0.777, respectively). The panel of sCysC plus uNAG detected total and severe AKI with significantly higher accuracy than either individual biomarkers or the other two panels (uNAG plus uACR or sCysC plus uACR). For detecting total AKI, severe AKI, and ICU mortality at ICU admission, this panel yielded AUC-ROCs of 0.756, 0.863, and 0.811, respectively; positive predictive values of 0.71, 0.31, and 0.17, respectively; and negative predictive values of 0.81, 0.97, and 0.98, respectively. Moreover, this panel significantly contributed to the accuracy of the clinical models for AKI detection and ICU mortality prediction, as measured by the AUC-ROC, continuous net reclassification index, and incremental discrimination improvement index. The comparable performance of this panel was further confirmed with bootstrap internal validation. CONCLUSIONS The combination of a functional marker (sCysC) and a tubular damage marker (uNAG) revealed significantly superior discriminative performance for AKI detection and yielded additional prognostic information on ICU mortality.
Collapse
Affiliation(s)
- Yujun Deng
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Ruibin Chi
- Department of Critical Care Medicine, Xiaolan Hospital of Southern Medical University, 65 Jucheng Road, Zhongshan, 528415, Guangdong, People's Republic of China
| | - Shenglong Chen
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Heng Ye
- Department of Critical Care Medicine, Guangzhou Nansha Central Hospital, 105 Fengzhe Road, Nansha, 511400, Guangdong, People's Republic of China
| | - Jie Yuan
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Lin Wang
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Yiling Zhai
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Lu Gao
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Danqing Zhang
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Linhui Hu
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Bo Lv
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Yi Long
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Cheng Sun
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Xiaobing Yang
- National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xia Zou
- School of Public Health, Sun Yat-sen University, 74 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Chunbo Chen
- Department of Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| |
Collapse
|
273
|
Tian L, Shao X, Xie Y, Wang Q, Che X, Zhang M, Xu W, Xu Y, Mou S, Ni Z. Kidney Injury Molecule-1 is Elevated in Nephropathy and Mediates Macrophage Activation via the Mapk Signalling Pathway. Cell Physiol Biochem 2017; 41:769-783. [PMID: 28214834 DOI: 10.1159/000458737] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/21/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIMS Kidney injury molecule-1 (KIM-1) is highly expressed in renal tubular cells after injury and is usually regarded as an early biomarker of acute kidney injury(AKI). The aim of this study was to determine the role of KIM-1 in the development of renal tubular injury Methods: Clinical samples, three different animal models and in vitro experiments were utilized to determine the possible mechanism underlying the involvement of KIM-1 in kidney injury. RESULTS Both plasma and urinary KIM-1 expression levels were significantly higher in AKI and chronic kidney disease (CKD) patients than in healthy volunteers, and urinary KIM-1 expression was significantly higher in CKD patients than in AKI patients. According to the results of our research involving three different mouse models, KIM-1 expression was significantly increased during the early stage of kidney injury and was persistently elevated in renal fibrosis. Our immunofluorescence staining results indicated that KIM-1-positive tubules were surrounded by macrophage infiltrates in regions of kidney injury. Moreover, our transwell, western blotting and real-time PCR data showed that macrophage migration and phenotype transitions were mediated by KIM-1 through the mitogen-activated protein kinase (MAPK) pathway. MAPK pathway inhibition could significantly reverse the effects of KIM-1 with respect to these macrophage phenotype changes and migration. CONCLUSIONS KIM-1 expression was markedly elevated in both acute and chronic kidney injury and may play a pivotal role in macrophage activation via the MAPK pathway in kidney disease.
Collapse
|
274
|
Shukla A, Rai MK, Prasad N, Agarwal V. Short-Term Non-Steroid Anti-Inflammatory Drug Use in Spondyloarthritis Patients Induces Subclinical Acute Kidney Injury: Biomarkers Study. Nephron Clin Pract 2017; 135:277-286. [PMID: 28171854 DOI: 10.1159/000455167] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/14/2016] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Non-steroid anti-inflammatory drug (NSAID) usage is associated with kidney injury. Rise in serum creatinine (sCr) often represents irreversible process. Thus to assess the early effects of regular NSAID use, we studied sensitive serum and urine biomarkers of kidney injury. METHODS In a protocol-based intervention study, 103 subjects were enrolled in 3 mutually exclusive groups. Group 1 included 37 healthy controls having minimal baseline NSAID exposure as per a definition, and group 2 had 41 spondyloarthritis (SpA) patients on regular NSAID therapy for >3 months. Group 3 included 25 SpA patients having minimal NSAID exposure at baseline. Blood and urine samples were collected from all the 3 groups at baseline. Furthermore, group 3 was started on 6-week regular NSAID therapy, and blood and urine samples were re-collected at 1, 6, and 12 weeks. Baseline normal kidney function as per the definition was ensured in all the subjects. Creatinine, neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), cystatin-C, and microalbumin were measured in urine and serum samples to assess kidney injury. RESULTS Kidney injury biomarkers were 2-3-fold higher in SpA patients using regular NSAID therapy compared to healthy controls and SpA patients having minimal NSAID exposure (uKIM-1 and uNGAL p < 0.0001, sKIM-1 and sNGAL p = 0.001). There was no difference in sCr and estimated glomerular filtration rate using Cockcroft-Gault equation between the groups. In SpA patients started on 6 weeks of regular NSAID (group 3), biomarker levels started rising at week 1 and showed a significant rise at week 6. The levels in the patients that stopped NSAID use at 6 weeks showed reversibility at 12 weeks. CONCLUSIONS Regular NSAID use in SpA patients induces subclinical kidney injury represented by rise in biomarkers. These levels start rising as early as 7 days of regular NSAID use and are reversible on stopping the drug.
Collapse
Affiliation(s)
- Anuj Shukla
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | | | | | | |
Collapse
|
275
|
Malas TB, Formica C, Leonhard WN, Rao P, Granchi Z, Roos M, Peters DJM, 't Hoen PAC. Meta-analysis of polycystic kidney disease expression profiles defines strong involvement of injury repair processes. Am J Physiol Renal Physiol 2017; 312:F806-F817. [PMID: 28148532 DOI: 10.1152/ajprenal.00653.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/30/2017] [Accepted: 01/30/2017] [Indexed: 12/25/2022] Open
Abstract
Polycystic kidney disease (PKD) is a major cause of end-stage renal disease. The disease mechanisms are not well understood and the pathogenesis toward renal failure remains elusive. In this study, we present the first RNASeq analysis of a Pkd1-mutant mouse model in a combined meta-analysis with other published PKD expression profiles. We introduce the PKD Signature, a set of 1,515 genes that are commonly dysregulated in PKD studies. We show that the signature genes include many known and novel PKD-related genes and functions. Moreover, genes with a role in injury repair, as evidenced by expression data and/or automated literature analysis, were significantly enriched in the PKD Signature, with 35% of the PKD Signature genes being directly implicated in injury repair. NF-κB signaling, epithelial-mesenchymal transition, inflammatory response, hypoxia, and metabolism were among the most prominent injury or repair-related biological processes with a role in the PKD etiology. Novel PKD genes with a role in PKD and in injury were confirmed in another Pkd1-mutant mouse model as well as in animals treated with a nephrotoxic agent. We propose that compounds that can modulate the injury-repair response could be valuable drug candidates for PKD treatment.
Collapse
Affiliation(s)
- Tareq B Malas
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Chiara Formica
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Wouter N Leonhard
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; and
| | | | | | - Marco Roos
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; and
| |
Collapse
|
276
|
Riefkohl A, Ramírez-Rubio O, Laws RL, McClean MD, Weiner DE, Kaufman JS, Galloway RL, Shadomy SV, Guerra M, Amador JJ, Sánchez JM, López-Pilarte D, Parikh CR, Leibler JH, Brooks DR. Leptospira seropositivity as a risk factor for Mesoamerican Nephropathy. INTERNATIONAL JOURNAL OF OCCUPATIONAL AND ENVIRONMENTAL HEALTH 2017; 23:1-10. [PMID: 28209095 PMCID: PMC6060841 DOI: 10.1080/10773525.2016.1275462] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 12/08/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Leptospirosis is postulated as a possible cause of Mesoamerican Nephropathy (MeN) in Central American workers. OBJECTIVES Investigate job-specific Leptospira seroprevalence and its association with kidney disease biomarkers. METHODS In 282 sugarcane workers, 47 sugarcane applicants and 160 workers in other industries, we measured anti-leptospiral antibodies, serum creatinine, and urinary injury biomarkers, including neutrophil gelatinase-associated lipocalin (NGAL), interleukin-18 (IL-18), and N-acetyl-D-glucosaminidase (NAG). RESULTS Leptospira seroprevalence differed among job categories and was highest among sugarcane cutters (59%). Seropositive sugarcane workers had higher NGAL concentrations (relative mean: 1.28; 95% CI: 0.94-1.75) compared to those who were seronegative, with similar findings among field and non-field workers. CONCLUSIONS Leptospira seroprevalence varied by job category. There was some indication that seropositivity was associated with elevated biomarker levels, but results were inconsistent. Additional studies may help establish whether Leptospira infection plays any role in MeN among Central American workers.
Collapse
Affiliation(s)
- Alejandro Riefkohl
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Oriana Ramírez-Rubio
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Preventive Medicine and Public Health Department, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rebecca L. Laws
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Michael D. McClean
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Daniel E. Weiner
- Division of Nephrology, Department of Medicine, Tufts Medical Center and Tufts University School of Medicine, Boston, MA, USA
| | - James S. Kaufman
- Research Service, VA New York Harbor Healthcare System and New York University School of Medicine, New York, NY, USA
| | - Renee L. Galloway
- Bacterial Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Sean V. Shadomy
- Bacterial Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Marta Guerra
- Bacterial Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Juan José Amador
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - José Marcel Sánchez
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Damaris López-Pilarte
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Chirag R. Parikh
- Section of Nephrology, Department of Medicine, Yale University, New Haven, CT, USA
- Program of Applied Translational Research, Department of Medicine, Yale University, New Haven, CT, USA
| | - Jessica H. Leibler
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Daniel R. Brooks
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
277
|
Mosa O, Skitek M, Jerin A. Validity of Klotho, CYR61 and YKL-40 as ideal predictive biomarkers for acute kidney injury: review study. SAO PAULO MED J 2017; 135:57-65. [PMID: 27759760 PMCID: PMC9969721 DOI: 10.1590/1516-3180.2016.0099220516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/22/2016] [Indexed: 02/08/2023] Open
Abstract
CONTEXT AND OBJECTIVE: Acute kidney injury (AKI) is still a headache for clinicians and scientists as a possible reason for increased death among intensive care unit (ICU) patients after invasive cardiac surgery. Furthermore, the diagnostic process for AKI using conventional biomarkers is not sufficient to ensure early warning of this condition because of the morbid influence of non-renal factors that definitively delay the time for the prognosis. These imposed limitations have led to significant amounts of research targeted towards identifying novel biomarkers for AKI with a sustained degree of sensitivity and specificity. Here, we reviewed previous studies conducted on the Klotho, CYR61 and YKL-40 biomarkers in relation to AKI. DESIGN AND SETTING: Review of the literature conducted in the Institute of Clinical Chemistry & Biochemistry, Ljubljana University Medical Center, Slovenia. METHODS: The literature was searched in PubMed and the Cochrane Library. From the database of this specialty, we selected 17 references that matched our context for detailed analysis and further investigation. RESULTS: The studies reviewed showed notable differences in their results relating to the diagnostic impact of Klotho, CYR61 and YKL-40 on early prediction of AKI. CONCLUSIONS: The results regarding the Klotho, CYR61 and YKL-40 biomarkers showed markedly equivocal performance in the previous studies and did not fulfill the expectations that these factors would form valid possible biomarkers for AKI.
Collapse
Affiliation(s)
- Osama Mosa
- PhD. Lecturer of Clinical Biochemistry, Department of Public Health, Health Science College at Al-Leith, Umm Al-Qura University, Saudi Arabia.
| | - Milan Skitek
- PhD. Professor and Head of Institute of Clinical Chemistry and Biochemistry, Ljubljana University Medical Center, Ljubljana, Slovenia.
| | - Ales Jerin
- PhD. Associate Professor and Head of Department of Hormones and Tumor Markers, Institute of Clinical Chemistry and Biochemistry, Ljubljana University Medical Center, Ljubljana, Slovenia.
| |
Collapse
|
278
|
Christians U, Klawitter J, Klepacki J, Klawitter J. The Role of Proteomics in the Study of Kidney Diseases and in the Development of Diagnostic Tools. BIOMARKERS OF KIDNEY DISEASE 2017:119-223. [DOI: 10.1016/b978-0-12-803014-1.00004-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
279
|
Makris K, Spanou L. Acute Kidney Injury: Diagnostic Approaches and Controversies. Clin Biochem Rev 2016; 37:153-175. [PMID: 28167845 PMCID: PMC5242479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Acute kidney injury (AKI) is a significant independent risk factor for morbidity and mortality. In the last ten years a large number of publications have highlighted the limitations of traditional approaches and the inadequacies of conventional biomarkers to diagnose and monitor renal insufficiency in the acute setting. A great effort was directed not only to the discovery and validation of new biomarkers aimed to detect AKI more accurately but also to standardise the definition of AKI. Despite the advances in both areas, biomarkers have not yet entered into routine clinical practice and the definition of this syndrome has many areas of uncertainty. This review will discuss the controversies in diagnosis and the potential of novel biomarkers to improve the definition of the syndrome.
Collapse
Affiliation(s)
- Konstantinos Makris
- Clinical Biochemistry Department, KAT General Hospital, Kifissia, Athens, 14561, Greece
| | - Loukia Spanou
- Clinical Biochemistry Department, KAT General Hospital, Kifissia, Athens, 14561, Greece
| |
Collapse
|
280
|
Konno T, Nakano R, Mamiya R, Tsuchiya H, Kitanaka T, Namba S, Kitanaka N, Okabayashi K, Narita T, Sugiya H. Expression and Function of Interleukin-1β-Induced Neutrophil Gelatinase-Associated Lipocalin in Renal Tubular Cells. PLoS One 2016; 11:e0166707. [PMID: 27851800 PMCID: PMC5112913 DOI: 10.1371/journal.pone.0166707] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/02/2016] [Indexed: 01/06/2023] Open
Abstract
Acute kidney injury (AKI) is characterized by a sudden loss of renal function. Early recognition of AKI, especially in critically ill patients, is essential for adequate therapy. Currently, neutrophil gelatinase-associated lipocalin (NGAL) is considered to be an effective biomarker of AKI; however, the regulation of its expression and function in renal tubular cells remains unclear. In this study, we investigated the regulation of the expression and function of NGAL in IL-1β-treated Madin–Darby canine kidney (MDCK) cells as a model of renal tubular cells. IL-1β induced a disturbance in the localization of E-cadherin and zonaoccludin-1 (ZO-1). The transepithelial electrical resistance (TER) also decreased 5 days after IL-1β treatment. IL-1β induced NGAL mRNA expression and protein secretion in a time- and dose-dependent manner, which occurred faster than the decrease in TER. In the presence of ERK1/2 and p38 inhibitors, IL-1β-induced NGAL mRNA expression and protein secretion were significantly attenuated. In the presence of recombinant NGAL, IL-1β-induced disturbance in the localization of E-cadherin and ZO-1 was attenuated, and the decrease in TER was partially maintained. These results suggest that NGAL can be used as a biomarker for AKI and that it functions as a protector from AKI.
Collapse
Affiliation(s)
- Tadayoshi Konno
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Rei Nakano
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Ryo Mamiya
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Hisashi Tsuchiya
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Taku Kitanaka
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Shinichi Namba
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Nanako Kitanaka
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Ken Okabayashi
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Takanori Narita
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
| | - Hiroshi Sugiya
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, 1866 Kameino, Fujisawa, Kanagawa, 252–0880, Japan
- * E-mail:
| |
Collapse
|
281
|
Autophagy in kidney disease and aging: lessons from rodent models. Kidney Int 2016; 90:950-964. [DOI: 10.1016/j.kint.2016.04.014] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 04/17/2016] [Accepted: 04/20/2016] [Indexed: 12/14/2022]
|
282
|
Single-Multiplex Detection of Organ Injury Biomarkers using SPRi based Nano-Immunosensor. Sci Rep 2016; 6:36348. [PMID: 27796342 PMCID: PMC5087088 DOI: 10.1038/srep36348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022] Open
Abstract
The clinical assessment of multiple organ dysfunctions at early stages is recognized to be an important factor in prompting definitive treatment decisions that prevent irreversible organ damage. In this article, we propose a real-time, label-free, and multiplex nanoenhanced SPRi platform to quantitatively assess two biomarkers, kidney injury molecule (KIM-1) and high mobility group box-1 (HMGB-1) simultaneously in buffer. Our work involves three major contributions in the design of the immunosensor: (1) we applied site-specific immobilization of antibodies to the solid surface that avoids loss of biological activity caused by covalent attachment; (2) we constructed a well-blocked sensor surface that exhibits minimal non-specific adsorption for singleplex measurements of each biomarker in buffer; and (3) we adopted a sandwich assay that implements functionalized quantum dots (NanoEnhancers) as signal amplifiers to achieve a sensitivity level of 5 pg/mL for KIM-1 and HMGB-1 in buffer. We foresee great potential and success in extending this multiplex and ultra-sensitive platform to assess a variety of other emerging clinical biomarkers at low concentrations and in complex matrices.
Collapse
|
283
|
Harwani SC, Ratcliff J, Sutterwala FS, Ballas ZK, Meyerholz DK, Chapleau MW, Abboud FM. Nicotine Mediates CD161a+ Renal Macrophage Infiltration and Premature Hypertension in the Spontaneously Hypertensive Rat. Circ Res 2016; 119:1101-1115. [PMID: 27660287 PMCID: PMC5085865 DOI: 10.1161/circresaha.116.309402] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/22/2016] [Indexed: 02/06/2023]
Abstract
RATIONALE Renal inflammation contributes to the pathophysiology of hypertension. CD161a+ immune cells are dominant in the (SHR) spontaneously hypertensive rat and expand in response to nicotinic cholinergic activation. OBJECTIVE We aimed to phenotype CD161a+ immune cells in prehypertensive SHR after cholinergic activation with nicotine and determine if these cells are involved in renal inflammation and the development of hypertension. METHODS AND RESULTS Studies used young SHR and WKY (Wistar-Kyoto) rats. Splenocytes and bone marrow cells were exposed to nicotine ex vivo, and nicotine was infused in vivo. Blood pressures, kidney, serum, and urine were obtained. Flow cytometry, Luminex/ELISA, immunohistochemistry, confocal microscopy, and Western blot were used. Nicotinic cholinergic activation induced proliferation of CD161a+/CD68+ macrophages in SHR-derived splenocytes, their renal infiltration, and premature hypertension in SHR. These changes were associated with increased renal expression of MCP-1 (monocyte chemoattractant protein-1) and VLA-4 (very-late antigen-4). LLT1 (lectin-like transcript 1), the ligand for CD161a, was overexpressed in SHR kidney, whereas vascular cellular and intracellular adhesion molecules were similar to those in WKY. Inflammatory cytokines were elevated in SHR kidney and urine after nicotine infusion. Nicotine-mediated renal macrophage infiltration/inflammation was enhanced in denervated kidneys, not explained by angiotensin II levels or expression of angiotensin type-1/2 receptors. Moreover, expression of the anti-inflammatory α7-nAChR (α7-nicotinic acetylcholine receptor) was similar in young SHR and WKY rats. CONCLUSIONS A novel, inherited nicotinic cholinergic inflammatory effect exists in young SHR, measured by expansion of CD161a+/CD68+ macrophages. This leads to renal inflammation and premature hypertension, which may be partially explained by increased renal expression of LLT-1, MCP-1, and VLA-4.
Collapse
MESH Headings
- Age of Onset
- Angiotensin II/metabolism
- Animals
- Antigens, CD/analysis
- Antigens, Differentiation, Myelomonocytic/analysis
- Cell Movement/drug effects
- Cells, Cultured
- Chemokine CCL2/biosynthesis
- Chemokine CCL2/genetics
- Cytokines/biosynthesis
- Cytokines/genetics
- Denervation
- Gene Expression Regulation/drug effects
- Hypertension/etiology
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/pathology
- Hypertension, Renal/etiology
- Hypertension, Renal/genetics
- Hypertension, Renal/metabolism
- Hypertension, Renal/pathology
- Immunophenotyping
- Integrin alpha4beta1/biosynthesis
- Integrin alpha4beta1/genetics
- Kidney/innervation
- Kidney/pathology
- Lectins/biosynthesis
- Lectins/genetics
- Macrophages/classification
- Macrophages/drug effects
- Macrophages/pathology
- Male
- NK Cell Lectin-Like Receptor Subfamily B/analysis
- Nephritis/chemically induced
- Nephritis/physiopathology
- Nicotine/pharmacology
- Nicotine/toxicity
- Norepinephrine/metabolism
- Prehypertension/etiology
- Prehypertension/genetics
- Prehypertension/pathology
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Receptor, Angiotensin, Type 1/biosynthesis
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 2/biosynthesis
- Receptor, Angiotensin, Type 2/genetics
- alpha7 Nicotinic Acetylcholine Receptor/biosynthesis
- alpha7 Nicotinic Acetylcholine Receptor/genetics
Collapse
Affiliation(s)
- Sailesh C Harwani
- From the Department of Internal Medicine (S.C.H., J.R., F.S.S., Z.K.B., M.W.C., F.M.A.), Departments of Molecular Physiology and Biophysics (M.W.C., F.M.A.), and Veterans Affairs Medical Center (F.S.S., Z.K.B., M.W.C.), Iowa City; and Department of Pathology (D.K.M.), Inflammation Program, Department of Internal Medicine (F.S.S.), Center for Immunology and Immune Mediated Diseases (S.C.H., F.S.S., F.M.A.), and Abboud Cardiovascular Research Center (S.C.H., J.R., M.W.C., F.M.A.), University of Iowa Carver College of Medicine, Iowa City.
| | - Jason Ratcliff
- From the Department of Internal Medicine (S.C.H., J.R., F.S.S., Z.K.B., M.W.C., F.M.A.), Departments of Molecular Physiology and Biophysics (M.W.C., F.M.A.), and Veterans Affairs Medical Center (F.S.S., Z.K.B., M.W.C.), Iowa City; and Department of Pathology (D.K.M.), Inflammation Program, Department of Internal Medicine (F.S.S.), Center for Immunology and Immune Mediated Diseases (S.C.H., F.S.S., F.M.A.), and Abboud Cardiovascular Research Center (S.C.H., J.R., M.W.C., F.M.A.), University of Iowa Carver College of Medicine, Iowa City
| | - Fayyaz S Sutterwala
- From the Department of Internal Medicine (S.C.H., J.R., F.S.S., Z.K.B., M.W.C., F.M.A.), Departments of Molecular Physiology and Biophysics (M.W.C., F.M.A.), and Veterans Affairs Medical Center (F.S.S., Z.K.B., M.W.C.), Iowa City; and Department of Pathology (D.K.M.), Inflammation Program, Department of Internal Medicine (F.S.S.), Center for Immunology and Immune Mediated Diseases (S.C.H., F.S.S., F.M.A.), and Abboud Cardiovascular Research Center (S.C.H., J.R., M.W.C., F.M.A.), University of Iowa Carver College of Medicine, Iowa City
| | - Zuhair K Ballas
- From the Department of Internal Medicine (S.C.H., J.R., F.S.S., Z.K.B., M.W.C., F.M.A.), Departments of Molecular Physiology and Biophysics (M.W.C., F.M.A.), and Veterans Affairs Medical Center (F.S.S., Z.K.B., M.W.C.), Iowa City; and Department of Pathology (D.K.M.), Inflammation Program, Department of Internal Medicine (F.S.S.), Center for Immunology and Immune Mediated Diseases (S.C.H., F.S.S., F.M.A.), and Abboud Cardiovascular Research Center (S.C.H., J.R., M.W.C., F.M.A.), University of Iowa Carver College of Medicine, Iowa City
| | - David K Meyerholz
- From the Department of Internal Medicine (S.C.H., J.R., F.S.S., Z.K.B., M.W.C., F.M.A.), Departments of Molecular Physiology and Biophysics (M.W.C., F.M.A.), and Veterans Affairs Medical Center (F.S.S., Z.K.B., M.W.C.), Iowa City; and Department of Pathology (D.K.M.), Inflammation Program, Department of Internal Medicine (F.S.S.), Center for Immunology and Immune Mediated Diseases (S.C.H., F.S.S., F.M.A.), and Abboud Cardiovascular Research Center (S.C.H., J.R., M.W.C., F.M.A.), University of Iowa Carver College of Medicine, Iowa City
| | - Mark W Chapleau
- From the Department of Internal Medicine (S.C.H., J.R., F.S.S., Z.K.B., M.W.C., F.M.A.), Departments of Molecular Physiology and Biophysics (M.W.C., F.M.A.), and Veterans Affairs Medical Center (F.S.S., Z.K.B., M.W.C.), Iowa City; and Department of Pathology (D.K.M.), Inflammation Program, Department of Internal Medicine (F.S.S.), Center for Immunology and Immune Mediated Diseases (S.C.H., F.S.S., F.M.A.), and Abboud Cardiovascular Research Center (S.C.H., J.R., M.W.C., F.M.A.), University of Iowa Carver College of Medicine, Iowa City
| | - Francois M Abboud
- From the Department of Internal Medicine (S.C.H., J.R., F.S.S., Z.K.B., M.W.C., F.M.A.), Departments of Molecular Physiology and Biophysics (M.W.C., F.M.A.), and Veterans Affairs Medical Center (F.S.S., Z.K.B., M.W.C.), Iowa City; and Department of Pathology (D.K.M.), Inflammation Program, Department of Internal Medicine (F.S.S.), Center for Immunology and Immune Mediated Diseases (S.C.H., F.S.S., F.M.A.), and Abboud Cardiovascular Research Center (S.C.H., J.R., M.W.C., F.M.A.), University of Iowa Carver College of Medicine, Iowa City
| |
Collapse
|
284
|
Liu KD, Siew ED, Reeves WB, Himmelfarb J, Go AS, Hsu CY, Bennett MR, Devarajan P, Ikizler TA, Kaufman JS, Kimmel PL, Chinchilli VM, Parikh CR, for the ASSESS-AKI Study Investigators. Storage Time and Urine Biomarker Levels in the ASSESS-AKI Study. PLoS One 2016; 11:e0164832. [PMID: 27788160 PMCID: PMC5082822 DOI: 10.1371/journal.pone.0164832] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 09/30/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Although stored urine samples are often used in biomarker studies focused on acute and chronic kidney disease, how storage time impacts biomarker levels is not well understood. METHODS 866 subjects enrolled in the NIDDK-sponsored ASsessment, Serial Evaluation, and Subsequent Sequelae in Acute Kidney Injury (ASSESS-AKI) Study were included. Samples were processed under standard conditions and stored at -70°C until analyzed. Kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), interleukin-18 (IL-18), and liver fatty acid binding protein (L-FABP) were measured in urine samples collected during the index hospitalization or an outpatient visit 3 months later. Mixed effects models were used to determine the effect of storage time on biomarker levels and stratified by visit. RESULTS Median storage was 17.8 months (25-75% IQR 10.6-23.7) for samples from the index hospitalization and 14.6 months (IQR 7.3-20.4) for outpatient samples. In the mixed effects models, the only significant association between storage time and biomarker concentration was for KIM-1 in outpatient samples, where each month of storage was associated with a 1.7% decrease (95% CI -3% to -0.3%). There was no relationship between storage time and KIM-1 levels in samples from the index hospitalization. CONCLUSION There was no significant impact of storage time over a median of 18 months on urine KIM-1, NGAL, IL-18 or L-FABP in hospitalized samples; a statistically significant effect towards a decrease over time was noted for KIM-1 in outpatient samples. Additional studies are needed to determine whether longer periods of storage at -70°C systematically impact levels of these analytes.
Collapse
Affiliation(s)
- Kathleen D. Liu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Edward D. Siew
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - W. Brian Reeves
- Department of Medicine, Division of Nephrology, Penn State College of Medicine, Hershey, PA, United States of America
| | - Jonathan Himmelfarb
- Kidney Research Institute, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Alan S. Go
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, United States of America
| | - Chi-yuan Hsu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, United States of America
| | - Michael R. Bennett
- Division of Nephrology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Prasad Devarajan
- Division of Nephrology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - T. Alp Ikizler
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - James S. Kaufman
- Research Service and Renal Section, VA New York Harbor Healthcare System and New York University School of Medicine, New York, NY, United States of America
| | - Paul L. Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, NIDDK, NIH, Bethesda, MD, United States of America
| | - Vernon M. Chinchilli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, United States of America
| | - Chirag R. Parikh
- Section of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States of America
- Program of Applied Translational Research, Yale University School of Medicine, New Haven, CT, United States of America
| | | |
Collapse
|
285
|
Abstract
Kidney injury molecule-1(KIM-1) is a type I membrane protein, comprising an extracellular portion and a cytoplasmic portion, which is expressed at very low levels in the normal kidney. The extracellular portion can cleave and rapidly enter tubule lumens after kidney injury, and can then be detected in the urine. It has been confirmed that the urine KIM-1 level is closely related to tissue KIM-1 level and correlated with kidney tissue damage. Not only is KIM-1 proven to be an early biomarker of acute kidney injury but it also has a potential role in predicting long-term renal outcome. This review summarizes the relationships between KIM-1 and kidney injury, especially in chronic kidney disease.
Collapse
Affiliation(s)
- Caixia Yin
- a Department of Nephrology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , Jiangsu Province , People's Republic of China
| | - Ningning Wang
- a Department of Nephrology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , Jiangsu Province , People's Republic of China
| |
Collapse
|
286
|
AbdulHameed MDM, Ippolito DL, Stallings JD, Wallqvist A. Mining kidney toxicogenomic data by using gene co-expression modules. BMC Genomics 2016; 17:790. [PMID: 27724849 PMCID: PMC5057266 DOI: 10.1186/s12864-016-3143-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 09/29/2016] [Indexed: 12/15/2022] Open
Abstract
Background Acute kidney injury (AKI) caused by drug and toxicant ingestion is a serious clinical condition associated with high mortality rates. We currently lack detailed knowledge of the underlying molecular mechanisms and biological networks associated with AKI. In this study, we carried out gene co-expression analyses using DrugMatrix—a large toxicogenomics database with gene expression data from rats exposed to diverse chemicals—and identified gene modules associated with kidney injury to probe the molecular-level details of this disease. Results We generated a comprehensive set of gene co-expression modules by using the Iterative Signature Algorithm and found distinct clusters of modules that shared genes and were associated with similar chemical exposure conditions. We identified two module clusters that showed specificity for kidney injury in that they 1) were activated by chemical exposures causing kidney injury, 2) were not activated by other chemical exposures, and 3) contained known AKI-relevant genes such as Havcr1, Clu, and Tff3. We used the genes in these AKI-relevant module clusters to develop a signature of 30 genes that could assess the potential of a chemical to cause kidney injury well before injury actually occurs. We integrated AKI-relevant module cluster genes with protein-protein interaction networks and identified the involvement of immunoproteasomes in AKI. To identify biological networks and processes linked to Havcr1, we determined genes within the modules that frequently co-express with Havcr1, including Cd44, Plk2, Mdm2, Hnmt, Macrod1, and Gtpbp4. We verified this procedure by showing that randomized data did not identify Havcr1 co-expression genes and that excluding up to 10 % of the data caused only minimal degradation of the gene set. Finally, by using an external dataset from a rat kidney ischemic study, we showed that the frequently co-expressed genes of Havcr1 behaved similarly in a model of non-chemically induced kidney injury. Conclusions Our study demonstrated that co-expression modules and co-expressed genes contain rich information for generating novel biomarker hypotheses and constructing mechanism-based molecular networks associated with kidney injury. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3143-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohamed Diwan M AbdulHameed
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, 504 Scott Street, Fort Detrick, MD, 21702, USA
| | - Danielle L Ippolito
- U.S. Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
| | - Jonathan D Stallings
- U.S. Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, 504 Scott Street, Fort Detrick, MD, 21702, USA.
| |
Collapse
|
287
|
DCoH: A novel biomarker for diagnosing acute kidney injury. Med Hypotheses 2016; 95:27-30. [PMID: 27692160 DOI: 10.1016/j.mehy.2016.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 08/14/2016] [Indexed: 11/21/2022]
Abstract
Initial diagnosis of acute kidney injury (AKI) is usually based on measuring serum-creatinine and blood urea nitrogen levels; however such measurements are still poor in identifying renal injuries at initial stages. These standard matrices are not enough to monitor the outcome and progression of AKI. The prognosis prevents proper treatment and timely delay in providing putative therapeutic agents. The cost effective therapies to get delayed, patient health gets compromised and ultimately requires renal transplant due to end-stage renal disease, which is another major problematic factor due to shortage kidney donors. To establish effective therapies for AKI the need will be facilitated by developing and identifying reliable, sensitive biomarkers which can be detected early during all stages of AKI, even during preclinical and clinical studies. Although reaching to human clinical trials takes years of thorough evaluations, preliminary studies should be carried out effectively by: (a) Employing cell culture analysis, (b) use of AKI animal models, studying various gene regulated networks, and biomarkers, and (c) patient serum sampling and testing. As elevated phenylalanine are indicative of AKI onset within 4h, its levels is controlled, 4a-Hydroxy-tetrahydrobiopterin dehydratase/dimerization cofactor of HNF-1 (DCoH). There is a possibility of targeting DCoH to the current bedside list of biomarkers involved in AKI onset.
Collapse
|
288
|
Chu X, Bleasby K, Chan GH, Nunes I, Evers R. The Complexities of Interpreting Reversible Elevated Serum Creatinine Levels in Drug Development: Does a Correlation with Inhibition of Renal Transporters Exist? Drug Metab Dispos 2016; 44:1498-509. [PMID: 26825641 DOI: 10.1124/dmd.115.067694] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/28/2016] [Indexed: 12/19/2022] Open
Abstract
In humans, creatinine is formed by a multistep process in liver and muscle and eliminated via the kidney by a combination of glomerular filtration and active transport. Based on current evidence, creatinine can be taken up into renal proximal tubule cells by the basolaterally localized organic cation transporter 2 (OCT2) and the organic anion transporter 2, and effluxed into the urine by the apically localized multidrug and toxin extrusion protein 1 (MATE1) and MATE2K. Drug-induced elevation of serum creatinine (SCr) and/or reduced creatinine renal clearance is routinely used as a marker for acute kidney injury. Interpretation of elevated SCr can be complex, because such increases can be reversible and explained by inhibition of renal transporters involved in active secretion of creatinine or other secondary factors, such as diet and disease state. Distinction between these possibilities is important from a drug development perspective, as increases in SCr can result in the termination of otherwise efficacious drug candidates. In this review, we discuss the challenges associated with using creatinine as a marker for kidney damage. Furthermore, to evaluate whether reversible changes in SCr can be predicted prospectively based on in vitro transporter inhibition data, an in-depth in vitro-in vivo correlation (IVIVC) analysis was conducted for 16 drugs with in-house and literature in vitro transporter inhibition data for OCT2, MATE1, and MATE2K, as well as total and unbound maximum plasma concentration (Cmax and Cmax,u) data measured in the clinic.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Kelly Bleasby
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Grace Hoyee Chan
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Irene Nunes
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Raymond Evers
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| |
Collapse
|
289
|
Moyake N, Buchmann E, Crowther NJ. Neutrophil gelatinase-associated lipocalin as a diagnostic marker of acute kidney injury in pre-eclampsia. J Obstet Gynaecol Res 2016; 42:1483-1488. [DOI: 10.1111/jog.13088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/28/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Naledi Moyake
- Department of Chemical Pathology, National Health Laboratory Service; University of the Witwatersrand Faculty of Health Sciences; Johannesburg South Africa
| | - Eckhart Buchmann
- Department of Obstetrics and Gynaecology, Chris Hani Baragwanath Academic Hospital; University of the Witwatersrand; Johannesburg South Africa
| | - Nigel J. Crowther
- Department of Chemical Pathology, National Health Laboratory Service; University of the Witwatersrand Faculty of Health Sciences; Johannesburg South Africa
| |
Collapse
|
290
|
Pavkovic M, Robinson-Cohen C, Chua AS, Nicoara O, Cárdenas-González M, Bijol V, Ramachandran K, Hampson L, Pirmohamed M, Antoine DJ, Frendl G, Himmelfarb J, Waikar SS, Vaidya VS. Detection of Drug-Induced Acute Kidney Injury in Humans Using Urinary KIM-1, miR-21, -200c, and -423. Toxicol Sci 2016; 152:205-13. [PMID: 27122240 PMCID: PMC5009468 DOI: 10.1093/toxsci/kfw077] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Drug-induced acute kidney injury (AKI) is often encountered in hospitalized patients. Although serum creatinine (SCr) is still routinely used for assessing AKI, it is known to be insensitive and nonspecific. Therefore, our objective was to evaluate kidney injury molecule 1 (KIM-1) in conjunction with microRNA (miR)-21, -200c, and -423 as urinary biomarkers for drug-induced AKI in humans. In a cross-sectional cohort of patients (n = 135) with acetaminophen (APAP) overdose, all 4 biomarkers were significantly (P < .004) higher not only in APAP-overdosed (OD) patients with AKI (based on SCr increase) but also in APAP-OD patients without clinical diagnosis of AKI compared with healthy volunteers. In a longitudinal cohort of patients with malignant mesothelioma receiving intraoperative cisplatin (Cp) therapy (n = 108) the 4 biomarkers increased significantly (P < .0014) over time after Cp administration, but could not be used to distinguish patients with or without AKI. Evidence for human proximal tubular epithelial cells (HPTECs) being the source of miRNAs in urine was obtained first, by in situ hybridization based confirmation of increase in miR-21 expression in the kidney sections of AKI patients and second, by increased levels of miR-21, -200c, and -423 in the medium of cultured HPTECs treated with Cp and 4-aminophenol (APAP degradation product). Target prediction analysis revealed 1102 mRNA targets of miR-21, -200c, and -423 that are associated with pathways perturbed in diverse pathological kidney conditions. In summary, we report noninvasive detection of AKI in humans by combining the sensitivity of KIM-1 along with mechanistic potentials of miR-21, -200c, and -423.
Collapse
Affiliation(s)
- Mira Pavkovic
- *Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts 02115 Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | | | - Alicia S Chua
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Oana Nicoara
- Boston Children's Hospital, Nephrology, Boston, Massachusetts 02115
| | | | - Vanesa Bijol
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | | | - Lucy Hampson
- Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Munir Pirmohamed
- Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Daniel J Antoine
- Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Gyorgy Frendl
- Department of Anesthesiology, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Jonathan Himmelfarb
- Kidney Research Institute, University of Washington, Seattle, Washington 98195
| | - Sushrut S Waikar
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Vishal S Vaidya
- *Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts 02115 Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115 Harvard School of Public Health, Environmental Health, Boston, MA, 02115
| |
Collapse
|
291
|
Lee SJ, Park S. Usefulness of the Neutrophil Gelatinase-Associated Lipocalin (NGAL) Kit for Acute Kidney Injury Patients at the Emergency Medical Center in Daegu. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2016. [DOI: 10.15324/kjcls.2016.48.2.49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Seung-Jin Lee
- Department of Laboratory Medicine, Daegu Catholic University Medical Center, Daegu 42472, Korea
| | - Sangwook Park
- Department of Biomedical Laboratory Science, College of Health, Kyungwoon University, Gumi 39160, Korea
| |
Collapse
|
292
|
Zulu M, Kaile T, Kantenga T, Chileshe C, Nkhoma P, Sinkala M. Kidney injury molecule-1 and microalbuminuria levels in Zambian population: biomarkers of kidney injury. Pan Afr Med J 2016; 24:54. [PMID: 27642395 PMCID: PMC5012817 DOI: 10.11604/pamj.2016.24.54.8759] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/05/2016] [Indexed: 01/18/2023] Open
Abstract
Introduction Kidney injury affects renal excretion of plasma analytes and metabolic waste products with grave pathologic consequences. Early detection, thus of kidney injury is essential for injury specific intervention that may avert permanent renal damage and delay progression of kidney injury. We aimed to evaluate Kidney Injury Molecule-1 (KIM-1) and Microalbuminuria (MAU), as biomarkers of kidney injury, in comparison with creatinine. Methods We compared the levels of urine MAU, urine KIM-1 and other plasma biochemical tests in specimens from 80 individuals with and without kidney disease. Results We found no difference in KIM-1 levels between the kidney disease group (2.82± 1.36ng/mL) and controls (3.29 ± 1.14ng/mL), p = 0.122. MAU was higher in participants with kidney disease (130.809± 84.744 µg/mL) than the controls (15.983± 20.442µg/mL), p ?0.001. KIM-1 showed a weak negative correlation with creatinine (r = -0.279, p = 0.09), whereas MAU was positively correlated with creatinine in participants with kidney disease with statistical significance (r = 0.556, p = 0.001). Conclusion The study demonstrated that in Zambian setting MAU and creatinine are sensitive biomarkers in the diagnosis of kidney damage. We moreover propose further evaluation of KIM-1 as a biomarker of kidney injury.
Collapse
Affiliation(s)
- Mildred Zulu
- University of Zambia, School of Medicine, Department of Pathology and Microbiology, Kalundu, Lusaka, Zambia
| | - Trevor Kaile
- University of Zambia, School of Medicine, Department of Pathology and Microbiology, Kalundu, Lusaka, Zambia
| | - Timothy Kantenga
- University Teaching Hospital, Department of Pathology and Microbiology, Kalundu, Lusaka, Zambia
| | - Chisanga Chileshe
- University of Zambia, School of Medicine, KS-HHV8 Research and Diagnostic Laboratory, Kalundu, Lusaka, Zambia
| | - Panji Nkhoma
- University of Zambia, School of Medicine, Department of Biomedical Sciences, Kalundu, Lusaka, Zambia
| | - Musalula Sinkala
- University of Zambia, School of Medicine, Department of Biomedical Sciences, Kalundu, Lusaka, Zambia
| |
Collapse
|
293
|
Husi H, Human C. Molecular determinants of acute kidney injury. J Inj Violence Res 2016; 7:75-86. [PMID: 26104320 PMCID: PMC4522318 DOI: 10.5249/jivr.v7i2.615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022] Open
Abstract
Background: Acute kidney injury (AKI) is a condition that leads to a rapid deterioration of renal function associated with impairment to maintain electrolyte and acid balance, and, if left untreated, ultimately irreversible kidney damage and renal necrosis. There are a number of causes that can trigger AKI, ranging from underlying conditions as well as trauma and surgery. Specifically, the global rise in surgical procedures led to a substantial increase of AKI incidence rates, which in turn impacts on mortality rates, quality of life and economic costs to the healthcare system. However, no effective therapy for AKI exists. Current approaches, such as pharmacological intervention, help in alleviating symptoms in slowing down the progression, but do not prevent or reverse AKI-induced organ damage. Methods: An in-depth understanding of the molecular machinery involved in and modulated by AKI induction and progression is necessary to specifically pharmacologically target key molecules. A major hurdle to devise a successful strategy is the multifactorial and complex nature of the disorder itself, whereby the activation of a number of seemingly independent molecular pathways in the kidney leads to apoptotic and necrotic events. Results: The renin-angiotensin-aldosterone-system (RAAS) axis appears to be a common element, leading to downstream events such as triggers of immune responses via the NFB pathway. Other pathways intricately linked with AKI-induction and progression are the tumor necrosis factor alpha (TNF α) and transforming growth factor beta (TGF β) signaling cascades, as well as a number of other modulators. Surprisingly, it has been shown that the involvement of the glutamatergic axis, believed to be mainly a component of the neurological system, is also a major contributor. Conclusions: Here we address the current understanding of the molecular pathways evoked in AKI, their interplay, and the potential to pharmacologically intervene in the effective prevention and/or progression of AKI.
Collapse
Affiliation(s)
- Holger Husi
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK.
| | | |
Collapse
|
294
|
Renal markers cystatin C and neutrophil gelatinase-associated lipocalin (NGAL) in postmortem samples. Forensic Sci Med Pathol 2016; 12:189-92. [PMID: 26972903 DOI: 10.1007/s12024-016-9760-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2016] [Indexed: 10/22/2022]
Abstract
PURPOSE Renal markers may provide valuable information for cause of death (CoD) investigation when assessing the influence of impaired kidney function. A commonly used marker, creatinine (Cr), increases due to kidney injury and is known to be reasonably stable in postmortem (PM) samples. More sensitive markers are needed, as the increase of serum Cr level only occurs after relatively severe renal damage. We evaluated two markers, cystatin C and neutrophil gelatinase-associated lipocalin (NGAL), in addition to Cr. To the best of our knowledge this was the first study to investigate cystatin C and NGAL in a postmortem (PM) context. METHODS Cr, cystatin C, and NGAL were measured from PM blood in 39 autopsy cases. NGAL was also measured from urine in 16 cases. Cystatin C and NGAL were analyzed using ELISA, Cr measurements were performed with Jaffe method. Correlations of these markers were evaluated. RESULTS AND CONCLUSIONS Both, blood cystatin C and NGAL, levels showed significant correlation with Cr (p = 0.05 and p = 0.01, respectively). Cystatin C and NGAL in blood are promising markers for further studies with PM samples.
Collapse
|
295
|
Fisch S, Liao R, Hsiao LL, Lu T. Early Detection of Drug-Induced Renal Hemodynamic Dysfunction Using Sonographic Technology in Rats. J Vis Exp 2016. [PMID: 27022768 PMCID: PMC4828235 DOI: 10.3791/52409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The kidney normally functions to maintain hemodynamic homeostasis and is a major site of damage caused by drug toxicity. Drug-induced nephrotoxicity is estimated to contribute to 19- 25% of all clinical cases of acute kidney injury (AKI) in critically ill patients. AKI detection has historically relied on metrics such as serum creatinine (sCr) or blood urea nitrogen (BUN) which are demonstrably inadequate in full assessment of nephrotoxicity in the early phase of renal dysfunction. Currently, there is no robust diagnostic method to accurately detect hemodynamic alteration in the early phase of AKI while such alterations might actually precede the rise in serum biomarker levels. Such early detection can help clinicians make an accurate diagnosis and help in in decision making for therapeutic strategy. Rats were treated with Cisplatin to induce AKI. Nephrotoxicity was assessed for six days using high-frequency sonography, sCr measurement and upon histopathology of kidney. Hemodynamic evaluation using 2D and Color-Doppler images were used to serially study nephrotoxicity in rats, using the sonography. Our data showed successful drug-induced kidney injury in adult rats by histological examination. Color-Doppler based sonographic assessment of AKI indicated that resistive-index (RI) and pulsatile-index (PI) were increased in the treatment group; and peak-systolic velocity (mm/s), end-diastolic velocity (mm/s) and velocity-time integral (VTI, mm) were decreased in renal arteries in the same group. Importantly, these hemodynamic changes evaluated by sonography preceded the rise of sCr levels. Sonography-based indices such as RI or PI can thus be useful predictive markers of declining renal function in rodents. From our sonography-based observations in the kidneys of rats that underwent AKI, we showed that these noninvasive hemodynamic measurements may consider as an accurate, sensitive and robust method in detecting early stage kidney dysfunction. This study also underscores the importance of ethical issues associated with animal use in research.
Collapse
Affiliation(s)
- Sudeshna Fisch
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Ronglih Liao
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Li-Li Hsiao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School
| | - Tzongshi Lu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School;
| |
Collapse
|
296
|
Zhybak M, Beni V, Vagin M, Dempsey E, Turner A, Korpan Y. Creatinine and urea biosensors based on a novel ammonium ion-selective copper-polyaniline nano-composite. Biosens Bioelectron 2016; 77:505-11. [DOI: 10.1016/j.bios.2015.10.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/29/2015] [Accepted: 10/03/2015] [Indexed: 11/16/2022]
|
297
|
Severin MJ, Trebucobich MS, Buszniez P, Brandoni A, Torres AM. The urinary excretion of an organic anion transporter as an early biomarker of methotrexate-induced kidney injury. Toxicol Res (Camb) 2016; 5:530-538. [PMID: 30090367 PMCID: PMC6062349 DOI: 10.1039/c5tx00436e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/06/2016] [Indexed: 11/21/2022] Open
Abstract
Methotrexate (MTX) belongs to a group of medicines known as antimetabolites. It is commonly used in the treatment of malignant diseases and is prescribed in autoimmune and chronic inflammatory disorders. Along with its effective therapeutic power, MTX has adverse effects on several organs, including the kidney. The organic anion transporter 5 (Oat5) is exclusively localized in the renal apical membrane. Oat5 urinary excretion was proposed as an early biomarker in ischemic and nephrotoxic-induced kidney injury and in renal damage due to vascular calcification in preclinical models. The aim of this study was to evaluate Oat5 renal expression and urinary excretion in rats 48 h after the exposure to different doses of MTX, in comparison with traditional markers of renal injury, such as creatinine and urea plasma levels, protein urinary levels, urinary alkaline phosphatase (AP) activity, fractional excretion of water (FEWater) and renal histology. Male Wistar rats were treated with a single intraperitoneal injection of MTX at different dosages: 40-80-120-180-360 mg per kg b.w. (M40, M80, M120, M180, M360, n = 4, respectively) and experiments were carried out 48 h after MTX administration. Oat5 renal expression was evaluated by western blotting and immunohistochemistry. Traditional parameters were only modified at the higher MTX dose (M360). Conversely, Oat5 urinary excretion was elevated at the middle dose of 80 mg per kg b.w. Oat5 renal expression was modified at the highest dose as well, both in homogenates and in apical membranes. These results suggest that Oat5 urinary excretion might serve as an early biomarker of MTX-induced kidney injury.
Collapse
Affiliation(s)
- María J Severin
- Área Farmacología , Facultad de Ciencias Bioquímicas y Farmacéuticas. Universidad Nacional de Rosario. CONICET , Argentina . ; Tel: +0054/341/4393400
| | - Mara S Trebucobich
- Área Farmacología , Facultad de Ciencias Bioquímicas y Farmacéuticas. Universidad Nacional de Rosario. CONICET , Argentina . ; Tel: +0054/341/4393400
| | - Patricia Buszniez
- Área Farmacología , Facultad de Ciencias Bioquímicas y Farmacéuticas. Universidad Nacional de Rosario. CONICET , Argentina . ; Tel: +0054/341/4393400
| | - Anabel Brandoni
- Área Farmacología , Facultad de Ciencias Bioquímicas y Farmacéuticas. Universidad Nacional de Rosario. CONICET , Argentina . ; Tel: +0054/341/4393400
| | - Adriana M Torres
- Área Farmacología , Facultad de Ciencias Bioquímicas y Farmacéuticas. Universidad Nacional de Rosario. CONICET , Argentina . ; Tel: +0054/341/4393400
| |
Collapse
|
298
|
Yang CH, Chang CH, Chen TH, Fan PC, Chang SW, Chen CC, Chu PH, Chen YT, Yang HY, Yang CW, Chen YC. Combination of Urinary Biomarkers Improves Early Detection of Acute Kidney Injury in Patients With Heart Failure. Circ J 2016; 80:1017-23. [PMID: 26888148 DOI: 10.1253/circj.cj-15-0886] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) is associated with morality and repeated hospitalization, and is frequently encountered in patients with acute decompensated heart failure (ADHF). However, few effective tools exist for early AKI identification and risk stratification. METHODS AND RESULTS This was a prospective observational study conducted in the coronary care unit (CCU) of a tertiary care university hospital. Patients with a diagnosis of ADHF and who were using diuretics were enrolled.Samples collected between December 2013 and February 2015 were tested for serum cystatin C (Cys-C), urinary neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1 (KIM-1). Demographic, clinical, and laboratory data were evaluated. A total of 103 adult patients with a mean age of 68 years were investigated. AKI was diagnosed in 49 patients (47.6%). For predicting intrinsic AKI on the first day of CCU admission, a combination of Cys-C and urine KIM-1 yielded an excellent area under the receiver operating characteristic curve of 0.828, a sensitivity of 71.0%, and specificity of 43.0%, for an overall accuracy of 78%. CONCLUSIONS In this study, we found that combinations of the biomarker (Cys-C and KIM-1) were an effective clinical model for predicting AKI in patients with ADHF. The biomarker was also useful for differentiating subclinical AKI in patients with ADHF.
Collapse
Affiliation(s)
- Chia-Hung Yang
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
299
|
Segawa T, Kobayashi Y, Inamoto S, Suzuki M, Endoh T, Itou T. Identification and Expression Profiles of microRNA in Dolphin. Zoolog Sci 2016; 33:92-7. [DOI: 10.2108/zs150090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Takao Segawa
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Yuki Kobayashi
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Satoko Inamoto
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Miwa Suzuki
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Tomoko Endoh
- Shinagawa Aquarium, 3-2-1 Katsushima, Shinagawa-ku, Tokyo 140-0012, Japan
| | - Takuya Itou
- Nihon University Veterinary Research Center, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| |
Collapse
|
300
|
Jacobo-Estrada T, Cardenas-Gonzalez M, Santoyo-Sánchez M, Parada-Cruz B, Uria-Galicia E, Arreola-Mendoza L, Barbier O. Evaluation of kidney injury biomarkers in rat amniotic fluid after gestational exposure to cadmium. J Appl Toxicol 2016; 36:1183-93. [PMID: 26815315 DOI: 10.1002/jat.3286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/11/2015] [Accepted: 12/12/2015] [Indexed: 12/24/2022]
Abstract
Cadmium is a well-characterized nephrotoxic agent that is also capable of accumulating and diffusing across the placenta; however, only a few studies have addressed its effects over fetal kidneys and none of them has used a panel of sensitive and specific biomarkers for the detection of kidney injury. The goal of this study was to determine cadmium renal effects in rat fetuses by the quantification of early kidney injury biomarkers. Pregnant Wistar rats were exposed by inhalation to an isotonic saline solution or to CdCl2 solution (DDel =1.48 mg Cd kg(-1) day(-1) ) during gestational days (GD) 8-20. On GD 21, dams were euthanized and samples obtained. Kidney injury biomarkers were quantified in amniotic fluid samples and fetal kidneys were microscopically evaluated to search for histological alterations. Our results showed that cadmium exposure significantly raised albumin, osteopontin, vascular endothelial growth factor and tissue inhibitor of metalloproteinases-1 levels in amniotic fluid, whereas it decreased creatinine. Clusterin, calbindin and IFN-inducible protein 10 did not show any change. Accordingly, histological findings showed tubular damage and precipitations in the renal pelvis. In conclusion, gestational exposure to cadmium induces structural alterations in fetal renal tissue that can be detected by some kidney injury biomarkers in amniotic fluid samples. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tania Jacobo-Estrada
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Mariana Cardenas-Gonzalez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Mitzi Santoyo-Sánchez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Benjamín Parada-Cruz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Esther Uria-Galicia
- Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomas, CP 11340, México, D.F., México
| | - Laura Arreola-Mendoza
- Departamento de Biociencias e Ingeniería, Centro Interdisciplinario de Investigaciones y Estudios sobre Medio Ambiente y Desarrollo, Instituto Politécnico Nacional, 30 de Junio de 1520 s/n, Col. Barrio la Laguna Ticomán, CP 07340, México, D.F., México
| | - Olivier Barbier
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| |
Collapse
|