1
|
Yang X, Li J, Liang F, Qu J, Dong X, Liu J. Mechanism of secondary renal injury in traumatic hemorrhagic shock model under a dry and heat desert environment. Sci Rep 2025; 15:14833. [PMID: 40295544 PMCID: PMC12037748 DOI: 10.1038/s41598-025-93853-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
We established a swine model of traumatic hemorrhagic shock to assess secondary renal injury under dry-heat conditions to clarify the roles of cell pyroptosis and inflammatory response in traumatic hemorrhagic shock development. Sixty-eight domestic Landrace piglets were divided into normothermic environment, dry-heat sham surgery, and dry-heat environment traumatic hemorrhagic shock groups (four subgroups: 3 h of environmental exposure and 60, 120, and 180 min after inducing traumatic hemorrhagic shock). The kidneys and blood were sampled at various time points. Univariate analysis of variance or non-parametric test was used for intergroup and intragroup comparisons, and the least significant difference test was used for multiple comparisons. The serum lipopolysaccharide, neutrophil gelatinase-associated lipocalin, kidney injury molecule 1, blood urea nitrogen, and creatinine levels, as well as various inflammatory factors, oxidative stress indicators, and Paller score, were significantly higher under dry-heat environment traumatic hemorrhagic shock than under normothermic environment and dry-heat sham surgery at 180 min. The histopathological damage in the dry-heat environment traumatic hemorrhagic shock group increased significantly at 180 min. Immunohistochemistry, western blotting, and terminal deoxynucleotidyl transferase dUTP nick end labeling assays showed that protein expression and apoptosis index values in the renal tissues of all three groups increased but were significantly higher under dry-heat environment traumatic hemorrhagic shock than under normothermic environment and dry-heat sham surgery at 180 min. The combination of dry-heat environment and traumatic hemorrhagic shock induces an aggravation of secondary renal injury, which may be related to cell pyroptosis, inflammatory response, apoptosis, and oxidative stress. Our findings may assist in the development of treatments for acute kidney injury.
Collapse
Affiliation(s)
- Xinyue Yang
- Graduate School of Xinjiang Medical University, Urumqi, 830000, Xinjiang province, China
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, 830000, Xinjiang Province, China
| | - Jiajia Li
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, 830000, Xinjiang Province, China
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830000, Xinjiang province, China
| | - Feixing Liang
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, 830000, Xinjiang Province, China
| | - Jinquan Qu
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, 830000, Xinjiang Province, China
| | - Xiang Dong
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, 830000, Xinjiang Province, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, Urumqi, 830000, Xinjiang Province, China.
| |
Collapse
|
2
|
Chatterjee T, Machado S, Cowen K, Miller M, Zhang Y, Volpicelli-Daley L, Fielding L, Pattanayak R, Rosenblum F, Potor L, Balla G, Balla J, Faul C, Zarjou A. Myeloid FtH Regulates Macrophage Response to Kidney Injury by Modulating Snca and Ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645219. [PMID: 40196511 PMCID: PMC11974884 DOI: 10.1101/2025.03.25.645219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
This study explored the role of myeloid ferritin heavy chain (FtH) in coordinating kidney iron trafficking in health and disease. Synuclein-α (Snca) was the sole iron-binding protein upregulated in response to myeloid FtH deletion (FtH Δ/Δ ). Following kidney injury, FtH Δ/Δ mice showed worsened kidney function. Transcriptome analysis revealed coupling of FtH deficiency with ferroptosis activation, a regulated cell death associated with iron accumulation. Adverse effects of ferroptosis were evidenced by upregulation of ferroptosis-related genes, increased oxidative stress markers, and significant iron deposition in kidney tissues. This iron buildup in FtH Δ/Δ kidneys stemmed from macrophage reprogramming into an iron-recycling phenotype, driven by Spic induction. Mechanistically, we establish that monomeric Snca functions as a ferrireductase catalyst, intensifying oxidative stress and triggering ferroptosis. Additionally, Snca accumulates in kidney diseases distinguished by leukocyte expansion across species. These findings position myeloid FtH as a pivotal orchestrator of the FtH-Snca-Spic axis driving macrophage reprogramming and kidney injury. Highlights Myeloid FtH deficiency drives kidney injury via activation of ferroptosisMΦ FtH deficiency induces Snca, linking iron dysregulation to MΦ function and response to kidney injuryFerrireductase activity of monomeric Snca augments oxidative stress, promoting lipid peroxidation and ferroptosis. In brief MΦ FtH modulates Snca and Spic to coordinate the injury response, linking iron trafficking to ferroptosis-induced kidney injury.
Collapse
|
3
|
Mrug M, Mrug E, Rosenblum F, Chen J, Cui X, Agarwal A, Zarjou A. Distinct developmental reprogramming footprint of macrophages during acute kidney injury across species. Am J Physiol Renal Physiol 2024; 326:F635-F641. [PMID: 38357719 PMCID: PMC11208015 DOI: 10.1152/ajprenal.00013.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
Acute kidney injury (AKI) is a common finding in hospitalized patients, particularly those who are critically ill. The development of AKI is associated with several adverse outcomes including mortality, morbidity, progression to chronic kidney disease, and an increase in healthcare expenditure. Despite the well-established negative impact of AKI and rigorous efforts to better define, identify, and implement targeted therapies, the overall approach to the treatment of AKI continues to principally encompass supportive measures. This enduring challenge is primarily due to the heterogeneous nature of insults that activate many independent and overlapping molecular pathways. Consequently, it is evident that the identification of common mechanisms that mediate the pathogenesis of AKI, independent of etiology and engaged pathophysiological pathways, is of paramount importance and could lead to the identification of novel therapeutic targets. To better distinguish the commonly modulated mechanisms of AKI, we explored the transcriptional characteristics of human kidney biopsies from patients with acute tubular necrosis (ATN), and acute interstitial nephritis (AIN) using a NanoString inflammation panel. Subsequently, we used publicly available single-cell transcriptional resources to better interpret the generated transcriptional findings. Our findings identify robust acute kidney injury (AKI-induced) developmental reprogramming of macrophages (MΦ) with the expansion of C1Q+, CD163+ MΦ that is independent of the etiology of AKI and conserved across mouse and human species. These results would expand the current understanding of the pathophysiology of AKI and potentially offer novel targets for additional studies to enhance the translational transition of AKI research.NEW & NOTEWORTHY Our findings identify robust acute kidney injury (AKI)-induced developmental reprogramming of macrophages (MΦ) with the expansion of C1Q+, CD163+ MΦ that is independent of the etiology of AKI and conserved across mouse and human species.
Collapse
Affiliation(s)
- Michal Mrug
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, United States
| | - Elias Mrug
- Math-Science Department, Alabama School of Fine Arts, Birmingham, Alabama, United States
| | - Frida Rosenblum
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jiandong Chen
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States
| | - Xiangqin Cui
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States
- Department of Veterans Affairs, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
4
|
Xing D, Hage FG, Feng W, Guo Y, Oparil S, Sanders PW. Endothelial cells overexpressing CXCR1/2 are renoprotective in rats with acute kidney injury. Am J Physiol Renal Physiol 2023; 324:F374-F386. [PMID: 36794755 PMCID: PMC10042609 DOI: 10.1152/ajprenal.00238.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/19/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Inflammation that develops with the release of chemokines and cytokines during acute kidney injury (AKI) has been shown to participate in functional renal recovery. Although a major research focus has been on the role of macrophages, the family of C-X-C motif chemokines that promote neutrophil adherence and activation also increases with kidney ischemia-reperfusion (I/R) injury. This study tested the hypothesis that intravenous delivery of endothelial cells (ECs) that overexpress (C-X-C motif) chemokine receptors 1 and 2 (CXCR1 and CXCR2, respectively) improves outcomes in kidney I/R injury. Overexpression of CXCR1/2 enhanced homing of endothelial cells to I/R-injured kidneys and limited interstitial fibrosis, capillary rarefaction, and tissue injury biomarkers (serum creatinine concentration and urinary kidney injury molecule-1) following AKI and also reduced expression of P-selectin and the rodent (C-X-C motif) chemokine cytokine-induced neutrophil chemoattractant (CINC)-2β as well as the number of myeloperoxidase-positive cells in the postischemic kidney. The serum chemokine/cytokine profile, including CINC-1, showed similar reductions. These findings were not observed in rats given endothelial cells transduced with an empty adenoviral vector (null-ECs) or a vehicle alone. These data indicate that extrarenal endothelial cells that overexpress CXCR1 and CXCR2, but not null-ECs or vehicle alone, reduce I/R kidney injury and preserve kidney function in a rat model of AKI.NEW & NOTEWORTHY Inflammation facilitates kidney ischemia-reperfusion (I/R) injury. Endothelial cells (ECs) that were modified to overexpress (C-X-C motif) chemokine receptor (CXCR)1/2 (CXCR1/2-ECs) were injected immediately following kidney I/R injury. The interaction of CXCR1/2-ECs, but not ECs transduced with an empty adenoviral vector, with injured kidney tissue preserved kidney function and reduced production of inflammatory markers, capillary rarefaction, and interstitial fibrosis. The study highlights a functional role for the C-X-C chemokine pathway in kidney damage following I/R injury.
Collapse
Affiliation(s)
- Dongqi Xing
- Division of Pulmonary, Allergy and Critical Care Medicine, Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Fadi G Hage
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States
| | - Wenguang Feng
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Yuanyuan Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States
| |
Collapse
|
5
|
Williams RM, Shah J, Mercer E, Tian HS, Thompson V, Cheung JM, Dorso M, Kubala JM, Gudas LJ, de Stanchina E, Jaimes EA, Heller DA. Kidney-Targeted Redox Scavenger Therapy Prevents Cisplatin-Induced Acute Kidney Injury. Front Pharmacol 2022; 12:790913. [PMID: 35046813 PMCID: PMC8762298 DOI: 10.3389/fphar.2021.790913] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Cisplatin-induced acute kidney injury (CI-AKI) is a significant co-morbidity of chemotherapeutic regimens. While this condition is associated with substantially lower survival and increased economic burden, there is no pharmacological agent to effectively treat CI-AKI. The disease is hallmarked by acute tubular necrosis of the proximal tubular epithelial cells primarily due to increased oxidative stress. We investigated a drug delivery strategy to improve the pharmacokinetics of an approved therapy that does not normally demonstrate appreciable efficacy in CI-AKI, as a preventive intervention. In prior work, we developed a kidney-selective mesoscale nanoparticle (MNP) that targets the renal proximal tubular epithelium. Here, we found that the nanoparticles target the kidneys in a mouse model of CI-AKI with significant damage. We evaluated MNPs loaded with the reactive oxygen species scavenger edaravone, currently used to treat stroke and ALS. We found a marked and significant therapeutic benefit with edaravone-loaded MNPs, including improved renal function, which we demonstrated was likely due to a decrease in tubular epithelial cell damage and death imparted by the specific delivery of edaravone. The results suggest that renal-selective edaravone delivery holds potential for the prevention of acute kidney injury among patients undergoing cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Ryan M. Williams
- The City College of New York Department of Biomedical Engineering, New York, NY, United States
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Janki Shah
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Elizabeth Mercer
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Helen S. Tian
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Vanessa Thompson
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Justin M. Cheung
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Madeline Dorso
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medical College, New York, NY, United States
| | - Jaclyn M. Kubala
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medical College, New York, NY, United States
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, United States
| | | | - Edgar A. Jaimes
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medical College, New York, NY, United States
| | - Daniel A. Heller
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
6
|
Gladytz T, Millward JM, Cantow K, Hummel L, Zhao K, Flemming B, Periquito JS, Pohlmann A, Waiczies S, Seeliger E, Niendorf T. Reliable kidney size determination by magnetic resonance imaging in pathophysiological settings. Acta Physiol (Oxf) 2021; 233:e13701. [PMID: 34089569 DOI: 10.1111/apha.13701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/05/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022]
Abstract
AIM Kidney diseases constitute a major health challenge, which requires noninvasive imaging to complement conventional approaches to diagnosis and monitoring. Several renal pathologies are associated with changes in kidney size, offering an opportunity for magnetic resonance imaging (MRI) biomarkers of disease. This work uses dynamic MRI and an automated bean-shaped model (ABSM) for longitudinal quantification of pathophysiologically relevant changes in kidney size. METHODS A geometry-based ABSM was developed for kidney size measurements in rats using parametric MRI (T2 , T2 * mapping). The ABSM approach was applied to longitudinal renal size quantification using occlusion of the (a) suprarenal aorta or (b) the renal vein, (c) increase in renal pelvis and intratubular pressure and (d) injection of an X-ray contrast medium into the thoracic aorta to induce pathophysiologically relevant changes in kidney size. RESULTS The ABSM yielded renal size measurements with accuracy and precision equivalent to the manual segmentation, with >70-fold time savings. The automated method could detect a ~7% reduction (aortic occlusion) and a ~5%, a ~2% and a ~6% increase in kidney size (venous occlusion, pelvis and intratubular pressure increase and injection of X-ray contrast medium, respectively). These measurements were not affected by reduced image quality following administration of ferumoxytol. CONCLUSION Dynamic MRI in conjunction with renal segmentation using an ABSM supports longitudinal quantification of changes in kidney size in pathophysiologically relevant experimental setups mimicking realistic clinical scenarios. This can potentially be instrumental for developing MRI-based diagnostic tools for various kidney disorders and for gaining new insight into mechanisms of renal pathophysiology.
Collapse
Affiliation(s)
- Thomas Gladytz
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jason M Millward
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kathleen Cantow
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Luis Hummel
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Kaixuan Zhao
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Bert Flemming
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Joāo S Periquito
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Institute of Physiology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sonia Waiczies
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Erdmann Seeliger
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
7
|
Packialakshmi B, Stewart IJ, Burmeister DM, Chung KK, Zhou X. Large animal models for translational research in acute kidney injury. Ren Fail 2021; 42:1042-1058. [PMID: 33043785 PMCID: PMC7586719 DOI: 10.1080/0886022x.2020.1830108] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
While extensive research using animal models has improved the understanding of acute kidney injury (AKI), this knowledge has not been translated into effective treatments. Many promising interventions for AKI identified in mice and rats have not been validated in subsequent clinical trials. As a result, the mortality rate of AKI patients remains high. Inflammation plays a fundamental role in the pathogenesis of AKI, and one reason for the failure to translate promising therapeutics may lie in the profound difference between the immune systems of rodents and humans. The immune systems of large animals such as swine, nonhuman primates, sheep, dogs and cats, more closely resemble the human immune system. Therefore, in the absence of a basic understanding of the pathophysiology of human AKI, large animals are attractive models to test novel interventions. However, there is a lack of reviews on large animal models for AKI in the literature. In this review, we will first highlight differences in innate and adaptive immunities among rodents, large animals, and humans in relation to AKI. After illustrating the potential merits of large animals in testing therapies for AKI, we will summarize the current state of the evidence in terms of what therapeutics have been tested in large animal models. The aim of this review is not to suggest that murine models are not valid to study AKI. Instead, our objective is to demonstrate that large animal models can serve as valuable and complementary tools in translating potential therapeutics into clinical practice.
Collapse
Affiliation(s)
| | - Ian J Stewart
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - David M Burmeister
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kevin K Chung
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
8
|
Periquito JS, Gladytz T, Millward JM, Delgado PR, Cantow K, Grosenick D, Hummel L, Anger A, Zhao K, Seeliger E, Pohlmann A, Waiczies S, Niendorf T. Continuous diffusion spectrum computation for diffusion-weighted magnetic resonance imaging of the kidney tubule system. Quant Imaging Med Surg 2021; 11:3098-3119. [PMID: 34249638 DOI: 10.21037/qims-20-1360] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
Background The use of rigid multi-exponential models (with a priori predefined numbers of components) is common practice for diffusion-weighted MRI (DWI) analysis of the kidney. This approach may not accurately reflect renal microstructure, as the data are forced to conform to the a priori assumptions of simplified models. This work examines the feasibility of less constrained, data-driven non-negative least squares (NNLS) continuum modelling for DWI of the kidney tubule system in simulations that include emulations of pathophysiological conditions. Methods Non-linear least squares (LS) fitting was used as reference for the simulations. For performance assessment, a threshold of 5% or 10% for the mean absolute percentage error (MAPE) of NNLS and LS results was used. As ground truth, a tri-exponential model using defined volume fractions and diffusion coefficients for each renal compartment (tubule system: Dtubules , ftubules ; renal tissue: Dtissue , ftissue ; renal blood: Dblood , fblood ;) was applied. The impact of: (I) signal-to-noise ratio (SNR) =40-1,000, (II) number of b-values (n=10-50), (III) diffusion weighting (b-rangesmall =0-800 up to b-rangelarge =0-2,180 s/mm2), and (IV) fixation of the diffusion coefficients Dtissue and Dblood was examined. NNLS was evaluated for baseline and pathophysiological conditions, namely increased tubular volume fraction (ITV) and renal fibrosis (10%: grade I, mild) and 30% (grade II, moderate). Results NNLS showed the same high degree of reliability as the non-linear LS. MAPE of the tubular volume fraction (ftubules ) decreased with increasing SNR. Increasing the number of b-values was beneficial for ftubules precision. Using the b-rangelarge led to a decrease in MAPE ftubules compared to b-rangesmall. The use of a medium b-value range of b=0-1,380 s/mm2 improved ftubules precision, and further bmax increases beyond this range yielded diminishing improvements. Fixing Dblood and Dtissue significantly reduced MAPE ftubules and provided near perfect distinction between baseline and ITV conditions. Without constraining the number of renal compartments in advance, NNLS was able to detect the (fourth) fibrotic compartment, to differentiate it from the other three diffusion components, and to distinguish between 10% vs. 30% fibrosis. Conclusions This work demonstrates the feasibility of NNLS modelling for DWI of the kidney tubule system and shows its potential for examining diffusion compartments associated with renal pathophysiology including ITV fraction and different degrees of fibrosis.
Collapse
Affiliation(s)
- Joāo S Periquito
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Institute of Physiology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany.,Experimental and Clinical Research Center, a Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thomas Gladytz
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jason M Millward
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Paula Ramos Delgado
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Experimental and Clinical Research Center, a Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kathleen Cantow
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Dirk Grosenick
- Physikalisch-Technische Bundesanstalt (PTB), Berlin, Germany
| | - Luis Hummel
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Ariane Anger
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Kaixuan Zhao
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Erdmann Seeliger
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sonia Waiczies
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Experimental and Clinical Research Center, a Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
9
|
Curtis LM, George J, Vallon V, Barnes S, Darley-Usmar V, Vaingankar S, Cutter GR, Gutierrez OM, Seifert M, Ix JH, Mehta RL, Sanders PW, Agarwal A. UAB-UCSD O'Brien Center for Acute Kidney Injury Research. Am J Physiol Renal Physiol 2021; 320:F870-F882. [PMID: 33779316 DOI: 10.1152/ajprenal.00661.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Acute kidney injury (AKI) remains a significant clinical problem through its diverse etiologies, the challenges of robust measurements of injury and recovery, and its progression to chronic kidney disease (CKD). Bridging the gap in our knowledge of this disorder requires bringing together not only the technical resources for research but also the investigators currently endeavoring to expand our knowledge and those who might bring novel ideas and expertise to this important challenge. The University of Alabama at Birmingham-University of California-San Diego O'Brien Center for Acute Kidney Injury Research brings together technical expertise and programmatic and educational efforts to advance our knowledge in these diverse issues and the required infrastructure to develop areas of novel exploration. Since its inception in 2008, this O'Brien Center has grown its impact by providing state-of-the-art resources in clinical and preclinical modeling of AKI, a bioanalytical core that facilitates measurement of critical biomarkers, including serum creatinine via LC-MS/MS among others, and a biostatistical resource that assists from design to analysis. Through these core resources and with additional educational efforts, our center has grown its investigator base to include >200 members from 51 institutions. Importantly, this center has translated its pilot and catalyst funding program with a $37 return per dollar invested. Over 500 publications have resulted from the support provided with a relative citation ratio of 2.18 ± 0.12 (iCite). Through its efforts, this disease-centric O'Brien Center is providing the infrastructure and focus to help the development of the next generation of researchers in the basic and clinical science of AKI. This center creates the promise of the application at the bedside of the advances in AKI made by current and future investigators.
Collapse
Affiliation(s)
- Lisa M Curtis
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - James George
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Volker Vallon
- Division of Nephrology, Department of Medicine, University of California-San Diego, San Diego, California
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sucheta Vaingankar
- Division of Pediatric Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gary R Cutter
- School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Orlando M Gutierrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael Seifert
- Division of Pediatric Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joachim H Ix
- Division of Nephrology, Department of Medicine, University of California-San Diego, San Diego, California
| | - Ravindra L Mehta
- Division of Nephrology, Department of Medicine, University of California-San Diego, San Diego, California
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Veterans Affairs, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Veterans Affairs, Birmingham, Alabama
| |
Collapse
|
10
|
Nath M, Agarwal A. New insights into the role of heme oxygenase-1 in acute kidney injury. Kidney Res Clin Pract 2020; 39:387-401. [PMID: 33184238 PMCID: PMC7770992 DOI: 10.23876/j.krcp.20.091] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022] Open
Abstract
Acute kidney injury (AKI) is attended by injury-related biomarkers appearing in the urine and serum, decreased urine output, and impaired glomerular filtration rate. AKI causes increased morbidity and mortality and can progress to chronic kidney disease and end-stage kidney failure. AKI is without specific therapies and is managed by supported care. Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. HO-1 induction protects against assorted renal insults as demonstrated by in vitro and preclinical models. This review summarizes the advances in understanding of the protection conferred by HO-1 in AKI, how HO-1 can be induced including via its transcription factor Nrf2, and HO-1 induction as a therapeutic strategy.
Collapse
Affiliation(s)
- Meryl Nath
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Veterans Affairs, Birmingham Veterans Administration Medical Center, Birmingham, AL, USA
| |
Collapse
|
11
|
El-Rashid M, Nguyen-Ngo D, Minhas N, Meijles DN, Li J, Ghimire K, Julovi S, Rogers NM. Repurposing of metformin and colchicine reveals differential modulation of acute and chronic kidney injury. Sci Rep 2020; 10:21968. [PMID: 33319836 PMCID: PMC7738483 DOI: 10.1038/s41598-020-78936-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is a major health problem affecting millions of patients globally. There is no effective treatment for AKI and new therapies are urgently needed. Novel drug development, testing and progression to clinical trials is overwhelmingly expensive. Drug repurposing is a more cost-effective measure. We identified 2 commonly used drugs (colchicine and metformin) that alter inflammatory cell function and signalling pathways characteristic of AKI, and tested them in models of acute and chronic kidney injury to assess therapeutic benefit. We assessed the renoprotective effects of colchicine or metformin in C57BL/6 mice challenged with renal ischemia reperfusion injury (IRI), treated before or after injury. All animals underwent analysis of renal function and biomolecular phenotyping at 24 h, 48 h and 4 weeks after injury. Murine renal tubular epithelial cells were studied in response to in vitro mimics of IRI. Pre-emptive treatment with colchicine or metformin protected against AKI, with lower serum creatinine, improved histological changes and decreased TUNEL staining. Pro-inflammatory cytokine profile and multiple markers of oxidative stress were not substantially different between groups. Metformin augmented expression of multiple autophagic proteins which was reversed by the addition of hydroxychloroquine. Colchicine led to an increase in inflammatory cells within the renal parenchyma. Chronic exposure after acute injury to either therapeutic agent in the context of reduced renal mass did not mitigate the development of fibrosis, with colchicine significantly worsening an ischemic phenotype. These data indicate that colchicine and metformin affect acute and chronic kidney injury differently. This has significant implications for potential drug repurposing, as baseline renal disease must be considered when selecting medication.
Collapse
Affiliation(s)
- Maryam El-Rashid
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Danny Nguyen-Ngo
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Sohel Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia. .,Westmead Clinical Medical School, University of Sydney, Camperdown, NSW, Australia. .,Renal Division, Westmead Hospital, Sydney, NSW, Australia. .,Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Georgiadis G, Zisis IE, Docea AO, Tsarouhas K, Fragkiadoulaki I, Mavridis C, Karavitakis M, Stratakis S, Stylianou K, Tsitsimpikou C, Calina D, Sofikitis N, Tsatsakis A, Mamoulakis C. Current Concepts on the Reno-Protective Effects of Phosphodiesterase 5 Inhibitors in Acute Kidney Injury: Systematic Search and Review. J Clin Med 2020; 9:jcm9051284. [PMID: 32365529 PMCID: PMC7287956 DOI: 10.3390/jcm9051284] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) is associated with increased morbidity, prolonged hospitalization, and mortality, especially in high risk patients. Phosphodiesterase 5 inhibitors (PDE5Is), currently available as first-line therapy of erectile dysfunction in humans, have shown a beneficial potential of reno-protection through various reno-protective mechanisms. The aim of this work is to provide a comprehensive overview of the available literature on the reno-protective properties of PDE5Is in the various forms of AKI. Medline was systematically searched from 1946 to November 2019 to detect all relevant animal and human studies in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. In total, 83 studies were included for qualitative synthesis. Sildenafil is the most widely investigated compound (42 studies), followed by tadalafil (20 studies), icariin (10 studies), vardenafil (7 studies), zaprinast (4 studies), and udenafil (2 studies). Even though data are limited, especially in humans with inconclusive or negative results of only two clinically relevant studies available at present, the results of animal studies are promising. The reno-protective action of PDE5Is was evident in the vast majority of studies, independently of the AKI type and the agent applied. PDE5Is appear to improve the renal functional/histopathological alternations of AKI through various mechanisms, mainly by affecting regional hemodynamics, cell expression, and mitochondrial response to oxidative stress and inflammation.
Collapse
Affiliation(s)
- Georgios Georgiadis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
| | - Ioannis-Erineos Zisis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Crete 71003, Greece;
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | | | - Irene Fragkiadoulaki
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Crete 71003, Greece;
| | - Charalampos Mavridis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
| | - Markos Karavitakis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
| | - Stavros Stratakis
- Department of Nephrology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (S.S.); (K.S.)
| | - Kostas Stylianou
- Department of Nephrology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (S.S.); (K.S.)
| | - Christina Tsitsimpikou
- Department of Hazardous Substances, Mixtures and Articles, General Chemical State Laboratory of Greece, Ampelokipi, Athens, Greece;
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Nikolaos Sofikitis
- Department of Urology, School of Medicine, Ioannina University, Ioannina, Greece;
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Crete 71003, Greece;
| | - Charalampos Mamoulakis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
- Correspondence:
| |
Collapse
|
13
|
Fiorani M, Guidarelli A, Cantoni O. Mitochondrial reactive oxygen species: the effects of mitochondrial ascorbic acid vs untargeted and mitochondria-targeted antioxidants. Int J Radiat Biol 2020; 97:1055-1062. [PMID: 31976796 DOI: 10.1080/09553002.2020.1721604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/16/2019] [Accepted: 01/07/2020] [Indexed: 12/20/2022]
Abstract
PREMISE Mitochondria represent critical sites for reactive oxygen species (ROS) production, which dependent on concentration is responsible for the regulation of both physiological and pathological processes. PURPOSE Antioxidants in mitochondria regulate the redox balance, prevent mitochondrial damage and dysfunction and maintain a physiological ROS-dependent signaling. The aim of the present review is to provide critical elements for addressing this issue in the context of various pharmacological approaches using antioxidants targeted or non-targeted to mitochondria. Furthermore, this review focuses on the mitochondrial antioxidant effects of ascorbic acid (AA), providing clues on the complexities associated with the cellular uptake and subcellular distribution of the vitamin. CONCLUSIONS Antioxidants that are not specifically targeted to mitochondria fail to accumulate in significant amounts in critical sites of mitochondrial ROS production and may eventually interfere with the ensuing physiological signaling. Mitochondria-targeted antioxidants are more effective, but are expected to interfere with the mitochondrial ROS-dependent physiologic signaling. AA promotes multiple beneficial effects in mitochondria. The complex regulation of vitamin C uptake in these organelles likely contributes to its versatile antioxidant response, thereby providing a central role to the vitamin for adequate control of mitochondrial dysfunction associated with increased mitochondrial ROS production.
Collapse
Affiliation(s)
- Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
14
|
Zhou Z, Ni K, Deng H, Chen X. Dancing with reactive oxygen species generation and elimination in nanotheranostics for disease treatment. Adv Drug Deliv Rev 2020; 158:73-90. [PMID: 32526453 DOI: 10.1016/j.addr.2020.06.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) play important roles in cell signaling and tissue homeostasis, in which the level of ROS is critical through the equilibrium between ROS generating and eliminating events. A disruption of the balance leads to disease development either by a surplus or a dearth of ROS, which requires ROS-modulating strategies to overturn the defect for disease treatment. Over the past decade, there have been tremendous advances in nanomedicine centering ROS generation and/or elimination as major mechanisms to treat a variety of diseases. In this review, we will discuss the research achievements on two opposite approaches of ROS-generating and ROS-eliminating strategies for treating cancer and other related diseases. Importantly, we will highlight the conceptual and strategic advances of ROS-mediated immunomodulation, including macrophage polarization, immunogenic cell death and T cell activation, which are currently rising as one of the mainstreams of cancer therapy. At the end, the future challenges and opportunities of mediating ROS-based mechanisms are envisioned. In light of the pleiotropic roles of ROS in different diseases, we hope this review is timely to deliver a clear logic of designing principles on ROS generation and elimination for different disease treatments.
Collapse
|
15
|
Diffusion-weighted Renal MRI at 9.4 Tesla Using RARE to Improve Anatomical Integrity. Sci Rep 2019; 9:19723. [PMID: 31873155 PMCID: PMC6928203 DOI: 10.1038/s41598-019-56184-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 10/23/2019] [Indexed: 12/29/2022] Open
Abstract
Diffusion-weighted magnetic resonance imaging (DWI) is a non-invasive imaging technique sensitive to tissue water movement. By enabling a discrimination between tissue properties without the need of contrast agent administration, DWI is invaluable for probing tissue microstructure in kidney diseases. DWI studies commonly make use of single-shot Echo-Planar Imaging (ss-EPI) techniques that are prone to suffering from geometric distortion. The goal of the present study was to develop a robust DWI technique tailored for preclinical magnetic resonance imaging (MRI) studies that is free of distortion and sensitive to detect microstructural changes. Since fast spin-echo imaging techniques are less susceptible to B0 inhomogeneity related image distortions, we introduced a diffusion sensitization to a split-echo Rapid Acquisition with Relaxation Enhancement (RARE) technique for high field preclinical DWI at 9.4 T. Validation studies in standard liquids provided diffusion coefficients consistent with reported values from the literature. Split-echo RARE outperformed conventional ss-EPI, with ss-EPI showing a 3.5-times larger border displacement (2.60 vs. 0.75) and a 60% higher intra-subject variability (cortex = 74%, outer medulla = 62% and inner medulla = 44%). The anatomical integrity provided by the split-echo RARE DWI technique is an essential component of parametric imaging on the way towards robust renal tissue characterization, especially during kidney disease.
Collapse
|
16
|
Zarjou A, Black LM, Bolisetty S, Traylor AM, Bowhay SA, Zhang MZ, Harris RC, Agarwal A. Dynamic signature of lymphangiogenesis during acute kidney injury and chronic kidney disease. J Transl Med 2019; 99:1376-1388. [PMID: 31019289 PMCID: PMC6716993 DOI: 10.1038/s41374-019-0259-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/07/2019] [Accepted: 03/29/2019] [Indexed: 11/09/2022] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected syndromes with significant attributable morbidity and mortality. The disturbing trend of increasing incidence and prevalence of these clinical disorders highlights the urgent need for better understanding of the underlying mechanisms that are involved in pathogenesis of these conditions. Lymphangiogenesis and its involvement in various inflammatory conditions is increasingly recognized while its role in AKI and CKD remains to be fully elucidated. Here, we studied lymphangiogenesis in three models of kidney injury. Our results demonstrate that the main ligands for lymphangiogenesis, VEGF-C and VEGF-D, are abundantly present in tubules at baseline conditions and the expression pattern of these ligands is significantly altered following injury. In addition, we show that both of these ligands increase in serum and urine post-injury and suggest that such increment may serve as novel urinary biomarkers of AKI as well as in progression of kidney disease. We also provide evidence that irrespective of the nature of initial insult, lymphangiogenic pathways are rapidly and robustly induced as evidenced by higher expression of lymphatic markers within the kidney.
Collapse
Affiliation(s)
- Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Laurence M Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sarah A Bowhay
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ming-Zhi Zhang
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, USA
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Nashville Veterans Affairs Hospital, Nashville, TN, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA.
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Veterans Affairs, Birmingham, AL, USA.
| |
Collapse
|
17
|
Balla J, Balla G, Zarjou A. Ferritin in Kidney and Vascular Related Diseases: Novel Roles for an Old Player. Pharmaceuticals (Basel) 2019; 12:E96. [PMID: 31234273 PMCID: PMC6630272 DOI: 10.3390/ph12020096] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is at the forefront of a number of pivotal biological processes due to its ability to readily accept and donate electrons. However, this property may also catalyze the generation of free radicals with ensuing cellular and tissue toxicity. Accordingly, throughout evolution numerous pathways and proteins have evolved to minimize the potential hazardous effects of iron cations and yet allow for readily available iron cations in a wide variety of fundamental metabolic processes. One of the extensively studied proteins in the context of systemic and cellular iron metabolisms is ferritin. While clinicians utilize serum ferritin to monitor body iron stores and inflammation, it is important to note that the vast majority of ferritin is located intracellularly. Intracellular ferritin is made of two different subunits (heavy and light chain) and plays an imperative role as a safe iron depot. In the past couple of decades our understanding of ferritin biology has remarkably improved. Additionally, a significant body of evidence has emerged describing the significance of the kidney in iron trafficking and homeostasis. Here, we briefly discuss some of the most important findings that relate to the role of iron and ferritin heavy chain in the context of kidney-related diseases and, in particular, vascular calcification, which is a frequent complication of chronic kidney disease.
Collapse
Affiliation(s)
- József Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary.
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - György Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary.
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
18
|
Ischia J, Bolton DM, Patel O. Why is it worth testing the ability of zinc to protect against ischaemia reperfusion injury for human application. Metallomics 2019; 11:1330-1343. [PMID: 31204765 DOI: 10.1039/c9mt00079h] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ischaemia (interruption in the blood/oxygen supply) and subsequent damage induced by reperfusion (restoration of blood/oxygen supply) ultimately leads to cell death, tissue injury and permanent organ dysfunction. The impact of ischaemia reperfusion injury (IRI) is not limited to heart attack and stroke but can be extended to patients undergoing surgeries such as partial nephrectomy for renal cancer, liver resection for colorectal cancer liver metastasis, cardiopulmonary bypass, and organ transplantation. Unfortunately, there are no drugs that can protect organs against the inevitable peril of IRI. Recent data show that a protocol incorporating specific Zn formulation, dosage, number of dosages, time of injection, and mode of Zn delivery (intravenous) and testing of efficacy in a large preclinical sheep model of IRI strongly supports human trials of Zn preconditioning. No doubt, scepticism still exists among funding bodies and research fraternity on whether Zn, a naturally occurring metal, will work where everything else has failed. Therefore, in this article, we review the conflicting evidence on the promoter and protector role of Zn in the case of IRI and highlight factors that may help explain the contradictory evidence. Finally, we review the literature related to the knowledge of Zn's mechanism of action on ROS generation, apoptosis, HIF activation, inflammation, and signal transduction pathways, which highlight Zn's likelihood of success compared to various other interventions targeting IRI.
Collapse
Affiliation(s)
- Joseph Ischia
- Department of Surgery, The University of Melbourne, Austin Health, Studley Rd., Heidelberg, Victoria 3084, Australia. and Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Damien M Bolton
- Department of Surgery, The University of Melbourne, Austin Health, Studley Rd., Heidelberg, Victoria 3084, Australia. and Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Oneel Patel
- Department of Surgery, The University of Melbourne, Austin Health, Studley Rd., Heidelberg, Victoria 3084, Australia.
| |
Collapse
|
19
|
O'Kane D, Baldwin GS, Bolton DM, Ischia JJ, Patel O. Preconditioning against renal ischaemia reperfusion injury: the failure to translate to the clinic. J Nephrol 2019; 32:539-547. [PMID: 30635875 DOI: 10.1007/s40620-019-00582-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/03/2019] [Indexed: 12/22/2022]
Abstract
Acute kidney injury (AKI) as a result of ischaemia-reperfusion represents a major healthcare burden worldwide. Mortality rates from AKI in hospitalized patients are extremely high and have changed little despite decades of research and medical advances. In 1986, Murry et al. demonstrated for the first time the phenomenon of ischaemic preconditioning to protect against ischaemia-reperfusion injury (IRI). This seminal finding paved the way for a broad body of research, which attempted to understand and ultimately harness this phenomenon for human application. The ability of preconditioning to limit renal IRI has now been demonstrated in multiple different animal models. However, more than 30 years later, a safe and consistent method of protecting human organs, including the kidneys, against IRI is still not available. This review highlights agents which, despite strong preclinical data, have recently failed to reduce AKI in human trials. The multiple reasons which may have contributed to the failure to translate some of the promising findings to clinical therapies are discussed. Agents which hold promise in the clinic because of their recent efficacy in preclinical large animal models are also reviewed.
Collapse
Affiliation(s)
- Dermot O'Kane
- Department of Surgery, Austin Health, The University of Melbourne, Studley Rd., Heidelberg, VIC, 3084, Australia
- Department of Urology, Austin Health, Heidelberg, VIC, Australia
| | - Graham S Baldwin
- Department of Surgery, Austin Health, The University of Melbourne, Studley Rd., Heidelberg, VIC, 3084, Australia
| | - Damien M Bolton
- Department of Surgery, Austin Health, The University of Melbourne, Studley Rd., Heidelberg, VIC, 3084, Australia
- Department of Urology, Austin Health, Heidelberg, VIC, Australia
| | - Joseph J Ischia
- Department of Surgery, Austin Health, The University of Melbourne, Studley Rd., Heidelberg, VIC, 3084, Australia
- Department of Urology, Austin Health, Heidelberg, VIC, Australia
| | - Oneel Patel
- Department of Surgery, Austin Health, The University of Melbourne, Studley Rd., Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
20
|
Sethi K, Rao K, Bolton D, Patel O, Ischia J. Targeting HIF-1 α to Prevent Renal Ischemia-Reperfusion Injury: Does It Work? Int J Cell Biol 2018; 2018:9852791. [PMID: 30595695 PMCID: PMC6286753 DOI: 10.1155/2018/9852791] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/14/2018] [Indexed: 12/26/2022] Open
Abstract
Partial nephrectomy (open or minimally invasive) usually requires temporary renal arterial occlusion to limit intraoperative bleeding and improve access to intrarenal structures. This is a time-critical step due to the critical ischemia period of renal tissue. Prolonged renal ischemia may lead to irreversible nephron damage in the remaining tissue and, ultimately, chronic kidney disease. This is potentiated by the incompletely understood ischemia-reperfusion injury (IRI). A key mechanism in IRI prevention appears to be the upregulation of an intracellular transcription protein, Hypoxia-Inducible Factor (HIF). HIF mediates metabolic adaptation, angiogenesis, erythropoiesis, cell growth, survival, and apoptosis. Upregulating HIF-1α via ischemic preconditioning (IPC) or drugs that simulate hypoxia (hypoxia-mimetics) has been investigated as a method to reduce IRI. While many promising chemical agents have been trialed for the prevention of IRI in small animal studies, all have failed in human trials. The aim of this review is to highlight the techniques and drugs that target HIF-1α and ameliorate IRI associated with renal ischemia. Developing a technique or drug that could reduce the risk of acute kidney injury associated with renal IRI would have an immediate worldwide impact on multisystem surgeries that would otherwise risk ischemic tissue injury.
Collapse
Affiliation(s)
- Kapil Sethi
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
- Urology Unit, Austin Health, Heidelberg, VIC, Australia
| | - Kenny Rao
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
- Urology Unit, Austin Health, Heidelberg, VIC, Australia
| | - Damien Bolton
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
- Urology Unit, Austin Health, Heidelberg, VIC, Australia
| | - Oneel Patel
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | - Joseph Ischia
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
- Urology Unit, Austin Health, Heidelberg, VIC, Australia
| |
Collapse
|
21
|
O'Kane D, Gibson L, May CN, du Plessis J, Shulkes A, Baldwin GS, Bolton D, Ischia J, Patel O. Zinc preconditioning protects against renal ischaemia reperfusion injury in a preclinical sheep large animal model. Biometals 2018; 31:821-834. [PMID: 29974287 DOI: 10.1007/s10534-018-0125-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/26/2018] [Indexed: 01/11/2023]
Abstract
Ischaemia-reperfusion injury (IRI) during various surgical procedures, including partial nephrectomy for kidney cancer or renal transplantation, is a major cause of acute kidney injury and chronic kidney disease. Currently there are no drugs or methods for protecting human organs, including the kidneys, against the peril of IRI. The aim of this study was therefore to investigate the reno-protective effect of Zn2+ preconditioning in a clinically relevant large animal sheep model of IRI. Further the reno-protective effectiveness of Zn2+ preconditioning was tested on normal human kidney cell lines HK-2 and HEK293. Anaesthetised sheep were subjected to uninephrectomy and 60 min of renal ischaemia followed by reperfusion. Sheep were preconditioned with intravenous injection of zinc chloride prior to occlusion. Serum creatinine and urea were measured before ischaemia and for 7 days after reperfusion. HK-2 and HEK293 cells were subjected to in vitro IRI using the oxygen- and glucose-deprivation model. Zn2+ preconditioning reduced ischaemic burden determined by creatinine and urea rise over time by ~ 70% in sheep. Zn2+ preconditioning also increased the survival of normal human kidney cells subjected to cellular stress such as hypoxia, hydrogen peroxide injury, and serum starvation. Overall, our protocol incorporating specific Zn2+ dosage, number of dosages (two), time of injection (24 and 4 h prior), mode of Zn2+ delivery (IV) and testing of efficacy in a rat model, a large preclinical sheep model of IRI and cells of human origin has laid the foundation for assessment of the benefit of Zn2+ preconditioning for human applications.
Collapse
Affiliation(s)
- Dermot O'Kane
- The University of Melbourne Department of Surgery, Austin Health, Studley Rd., Heidelberg, VIC, 3084, Australia
- Department of Urology, Austin Health, Heidelberg, VIC, Australia
| | - Luke Gibson
- The University of Melbourne Department of Surgery, Austin Health, Studley Rd., Heidelberg, VIC, 3084, Australia
- Department of Urology, Austin Health, Heidelberg, VIC, Australia
| | - Clive N May
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Justin du Plessis
- Australian Clinical Laboratories, Austin Health, Heidelberg, VIC, Australia
| | - Arthur Shulkes
- The University of Melbourne Department of Surgery, Austin Health, Studley Rd., Heidelberg, VIC, 3084, Australia
| | - Graham S Baldwin
- The University of Melbourne Department of Surgery, Austin Health, Studley Rd., Heidelberg, VIC, 3084, Australia
| | - Damien Bolton
- The University of Melbourne Department of Surgery, Austin Health, Studley Rd., Heidelberg, VIC, 3084, Australia
- Department of Urology, Austin Health, Heidelberg, VIC, Australia
| | - Joseph Ischia
- The University of Melbourne Department of Surgery, Austin Health, Studley Rd., Heidelberg, VIC, 3084, Australia
- Department of Urology, Austin Health, Heidelberg, VIC, Australia
| | - Oneel Patel
- The University of Melbourne Department of Surgery, Austin Health, Studley Rd., Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
22
|
Fiorentino M, Kellum JA. Improving Translation from Preclinical Studies to Clinical Trials in Acute Kidney Injury. Nephron Clin Pract 2018; 140:81-85. [PMID: 29791911 DOI: 10.1159/000489576] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 04/22/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Several cellular and molecular targets and mechanisms have been investigated in preclinical studies of acute kidney injury (AKI), but translation in successful clinical studies has failed to date. This article reviews many issues that have limited this and the potential future perspectives in AKI prevention and treatment. SUMMARY Preclinical models of AKI should closely mimic the complexity of human AKI, considering the importance of several comorbidities in determining the clinical course and outcomes in the human disease. Moreover, studies should test novel interventions in models where AKI is already established, instead of focusing only at primary prevention. AKI definitions and endpoints in animal studies should be similar to those applied in clinical studies; in particular, AKI biomarkers should be implemented to guide patient selection for clinical trials and monitor intervention efficacy. In this scenario, cell-cycle arrest biomarkers have been widely investigated as AKI predictors in both preclinical and clinical studies and they serve as useful tools for future interventional studies. A better understanding of human AKI through a large collection of biological samples and kidney biopsies and omics applications, and an iterative relationship between preclinical and clinical studies are critical steps to improve future preclinical models and clinical trials. Finally, given the great variability in clinical manifestation of AKI, a strong collaboration between research centers and industry is recommended. Key messages: Several methodological issues have hampered the translation of basic research findings in clinical studies, and overcoming these obstacles is necessary to achieve success.
Collapse
Affiliation(s)
- Marco Fiorentino
- Center for Critical Care Nephrology, Department of Critical Care Medicine, CRISMA (Clinical Research, Investigation, and System Modeling of Acute Illness) Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari, Bari, Italy
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, CRISMA (Clinical Research, Investigation, and System Modeling of Acute Illness) Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
23
|
Pohlmann A, Cantow K, Huelnhagen T, Grosenick D, Dos Santos Periquito J, Boehmert L, Gladytz T, Waiczies S, Flemming B, Seeliger E, Niendorf T. Experimental MRI Monitoring of Renal Blood Volume Fraction Variations En Route to Renal Magnetic Resonance Oximetry. ACTA ACUST UNITED AC 2017; 3:188-200. [PMID: 30042981 PMCID: PMC6024389 DOI: 10.18383/j.tom.2017.00012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diagnosis of early-stage acute kidney injury (AKI) will benefit from a timely identification of local tissue hypoxia. Renal tissue hypoxia is an early feature in AKI pathophysiology, and renal oxygenation is increasingly being assessed through T2*-weighted magnetic resonance imaging (MRI). However, changes in renal blood volume fraction (BVf) confound renal T2*. The aim of this study was to assess the feasibility of intravascular contrast-enhanced MRI for monitoring renal BVf during physiological interventions that are concomitant with variations in BVf and to explore the possibility of correcting renal T2* for BVf variations. A dose-dependent study of the contrast agent ferumoxytol was performed in rats. BVf was monitored throughout short-term occlusion of the renal vein, which is known to markedly change renal blood partial pressure of O2 and BVf. BVf calculated from MRI measurements was used to estimate oxygen saturation of hemoglobin (SO2). BVf and SO2 were benchmarked against cortical data derived from near-infrared spectroscopy. As estimated from magnetic resonance parametric maps of T2 and T2*, BVf was shown to increase, whereas SO2 was shown to decline during venous occlusion (VO). This observation could be quantitatively reproduced in test–retest scenarios. Changes in BVf and SO2 were in good agreement with data obtained from near-infrared spectroscopy. Our findings provide motivation to advance multiparametric MRI for studying AKIs, with the ultimate goal of translating MRI-based renal BVf mapping into clinical practice en route noninvasive renal magnetic resonance oximetry as a method of assessing AKI and progression to chronic damage.
Collapse
Affiliation(s)
- Andreas Pohlmann
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
| | - Kathleen Cantow
- Institute of Physiology and Center for Cardiovascular Research, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Till Huelnhagen
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
| | - Dirk Grosenick
- Physikalisch-Technische-Bundesanstalt (PTB), Berlin, Germany
| | - Joāo Dos Santos Periquito
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
| | - Laura Boehmert
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
| | - Thomas Gladytz
- Physikalisch-Technische-Bundesanstalt (PTB), Berlin, Germany
| | - Sonia Waiczies
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
| | - Bert Flemming
- Institute of Physiology and Center for Cardiovascular Research, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Erdmann Seeliger
- Institute of Physiology and Center for Cardiovascular Research, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Thoralf Niendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany.,Experimental and Clinical Research Center, Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; and.,Deutsches Zentrum für Herz- Kreislauf-Forschung (DZHK; German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
24
|
Williams RM, Shah J, Tian HS, Chen X, Geissmann F, Jaimes EA, Heller DA. Selective Nanoparticle Targeting of the Renal Tubules. Hypertension 2017; 71:87-94. [PMID: 29133360 DOI: 10.1161/hypertensionaha.117.09843] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 06/25/2017] [Accepted: 09/21/2017] [Indexed: 01/18/2023]
Abstract
Direct targeting to the kidneys is a promising strategy to improve drug therapeutic index for the treatment of kidney diseases. We sought to investigate the renal selectivity and safety of kidney-targeted mesoscale nanoparticle technology. We found that direct intravenous administration of these particles resulted in 26-fold renal selectivity and localized negligibly in the liver or other organs. The nanoparticles targeted the renal proximal tubular epithelial cells, as evidenced by intravital microscopy and ex vivo imaging. Mice treated with the nanoparticles exhibited no negative systemic consequences, immune reaction, liver impairment, or renal impairment. The localization of material selectively to the renal tubules is uncommon, and this work portends the development of renal-targeted drugs for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Ryan M Williams
- From the Memorial Sloan Kettering Cancer Center, New York (R.M.W., J.S., H.S.T., X.C., F.G., E.A.J., D.A.H.); University of Massachusetts Medical School, Worcester (H.S.T.); and Weill Cornell Medical College, New York (X.C., F.G., E.A.J., D.A.H.)
| | - Janki Shah
- From the Memorial Sloan Kettering Cancer Center, New York (R.M.W., J.S., H.S.T., X.C., F.G., E.A.J., D.A.H.); University of Massachusetts Medical School, Worcester (H.S.T.); and Weill Cornell Medical College, New York (X.C., F.G., E.A.J., D.A.H.)
| | - Helen S Tian
- From the Memorial Sloan Kettering Cancer Center, New York (R.M.W., J.S., H.S.T., X.C., F.G., E.A.J., D.A.H.); University of Massachusetts Medical School, Worcester (H.S.T.); and Weill Cornell Medical College, New York (X.C., F.G., E.A.J., D.A.H.)
| | - Xi Chen
- From the Memorial Sloan Kettering Cancer Center, New York (R.M.W., J.S., H.S.T., X.C., F.G., E.A.J., D.A.H.); University of Massachusetts Medical School, Worcester (H.S.T.); and Weill Cornell Medical College, New York (X.C., F.G., E.A.J., D.A.H.)
| | - Frederic Geissmann
- From the Memorial Sloan Kettering Cancer Center, New York (R.M.W., J.S., H.S.T., X.C., F.G., E.A.J., D.A.H.); University of Massachusetts Medical School, Worcester (H.S.T.); and Weill Cornell Medical College, New York (X.C., F.G., E.A.J., D.A.H.)
| | - Edgar A Jaimes
- From the Memorial Sloan Kettering Cancer Center, New York (R.M.W., J.S., H.S.T., X.C., F.G., E.A.J., D.A.H.); University of Massachusetts Medical School, Worcester (H.S.T.); and Weill Cornell Medical College, New York (X.C., F.G., E.A.J., D.A.H.)
| | - Daniel A Heller
- From the Memorial Sloan Kettering Cancer Center, New York (R.M.W., J.S., H.S.T., X.C., F.G., E.A.J., D.A.H.); University of Massachusetts Medical School, Worcester (H.S.T.); and Weill Cornell Medical College, New York (X.C., F.G., E.A.J., D.A.H.).
| |
Collapse
|
25
|
Hull TD, Agarwal A, Hoyt K. New Ultrasound Techniques Promise Further Advances in AKI and CKD. J Am Soc Nephrol 2017; 28:3452-3460. [PMID: 28923914 DOI: 10.1681/asn.2017060647] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AKI and CKD are important clinical problems because they affect many patients and the associated diagnostic and treatment paradigms are imperfect. Ultrasound is a cost-effective, noninvasive, and simple imaging modality that offers a multitude of means to improve the diagnosis, monitoring, and treatment of both AKI and CKD, especially considering recent advances in this technique. Ultrasound alone can attenuate AKI and prevent CKD by stimulating the splenic cholinergic anti-inflammatory pathway. Additionally, microbubble contrast agents are improving the sensitivity and specificity of ultrasound for diagnosing kidney disease, especially when these agents are conjugated to ligand-specific mAbs or peptides, which make the dynamic assessment of disease progression and response to treatment possible. More recently, drug-loaded microbubbles have been developed and the load release by ultrasound exposure has been shown to be a highly specific treatment modality, making the potential applications of ultrasound even more promising. This review focuses on the multiple strategies for using ultrasound with and without microbubble technology for enhancing our understanding of the pathophysiology of AKI and CKD.
Collapse
Affiliation(s)
- Travis D Hull
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas; and .,Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
26
|
Bolisetty S, Zarjou A, Agarwal A. Heme Oxygenase 1 as a Therapeutic Target in Acute Kidney Injury. Am J Kidney Dis 2017; 69:531-545. [PMID: 28139396 DOI: 10.1053/j.ajkd.2016.10.037] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/22/2016] [Indexed: 01/06/2023]
Abstract
A common clinical condition, acute kidney injury (AKI) significantly influences morbidity and mortality, particularly in critically ill patients. The pathophysiology of AKI is complex and involves multiple pathways, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Recent evidence suggests that a single insult to the kidney significantly enhances the propensity to develop chronic kidney disease. Therefore, the generation of effective therapies against AKI is timely. In this context, the cytoprotective effects of heme oxygenase 1 (HO-1) in animal models of AKI are well documented. HO-1 modulates oxidative stress, autophagy, and inflammation and regulates the progression of cell cycle via direct and indirect mechanisms. These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). This review highlights recent advances in the molecular mechanisms of HO-1-mediated cytoprotection and discusses the translational potential of HO-1 induction in AKI.
Collapse
Affiliation(s)
- Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL; Birmingham Veterans Administration Medical Center, Birmingham, AL.
| |
Collapse
|
27
|
Affiliation(s)
- S. Reuter
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| | - R. Mrowka
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| |
Collapse
|
28
|
Ortiz A. Translational nephrology: what translational research is and a bird's-eye view on translational research in nephrology. Clin Kidney J 2015; 8:14-22. [PMID: 25713705 PMCID: PMC4310441 DOI: 10.1093/ckj/sfu142] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 12/16/2014] [Indexed: 01/05/2023] Open
Abstract
The ultimate aim of biomedical research is to preserve health and improve patient outcomes. However, by a variety of measures, preservation of kidney health and patient outcomes in kidney disease are suboptimal. Severe acute kidney injury has been treated solely by renal replacement therapy for over 50 years and mortality still hovers at around 50%. Worldwide deaths from chronic kidney disease (CKD) increased by 80% in 20 years--one of the greatest increases among major causes of death. This dramatic data concur with huge advances in the cellular and molecular pathophysiology of kidney disease and its consequences. The gap appears to be the result of sequential roadblocks that impede an adequate flow from basic research to clinical development [translational research type 1 (T1), bench-to-bed and back] and from clinical development to clinical practice and widespread implementation (translational research T2) that supported by healthcare policy-making reaches all levels of society throughout the globe (sometimes called translational research T3). Thus, it is more than 10 years since the introduction of the last new-concept drug for CKD patients, cinacalcet; and 30 years since the introduction of reninangiotensin system (RAS) blockade, the current mainstay to prevent progression of CKD, illustrating the basic science-clinical practice disconnect. Roadblocks from clinical advances to widespread implementation, together with lag time-to-benefit may underlie the 20 years since the description of the antiproteinuric effect of RAS blockade to the observation of decreased age-adjusted incidence of endstage renal disease due to diabetic kidney disease. Only a correct understanding of the roadblocks in translational medicine and a full embracement of a translational research culture will spread the benefits of the biomedical revolution to its ultimate destinatary, the society.
Collapse
Affiliation(s)
- Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, School of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
- Fundacion Renal Iñigo Alvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| |
Collapse
|
29
|
Niendorf T, Pohlmann A, Arakelyan K, Flemming B, Cantow K, Hentschel J, Grosenick D, Ladwig M, Reimann H, Klix S, Waiczies S, Seeliger E. How bold is blood oxygenation level-dependent (BOLD) magnetic resonance imaging of the kidney? Opportunities, challenges and future directions. Acta Physiol (Oxf) 2015; 213:19-38. [PMID: 25204811 DOI: 10.1111/apha.12393] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/04/2014] [Accepted: 09/04/2014] [Indexed: 12/11/2022]
Abstract
Renal tissue hypoperfusion and hypoxia are key elements in the pathophysiology of acute kidney injury and its progression to chronic kidney disease. Yet, in vivo assessment of renal haemodynamics and tissue oxygenation remains a challenge. Many of the established approaches are invasive, hence not applicable in humans. Blood oxygenation level-dependent (BOLD) magnetic resonance imaging (MRI) offers an alternative. BOLD-MRI is non-invasive and indicative of renal tissue oxygenation. Nonetheless, recent (pre-) clinical studies revived the question as to how bold renal BOLD-MRI really is. This review aimed to deliver some answers. It is designed to inspire the renal physiology, nephrology and imaging communities to foster explorations into the assessment of renal oxygenation and haemodynamics by exploiting the powers of MRI. For this purpose, the specifics of renal oxygenation and perfusion are outlined. The fundamentals of BOLD-MRI are summarized. The link between tissue oxygenation and the oxygenation-sensitive MR biomarker T2∗ is outlined. The merits and limitations of renal BOLD-MRI in animal and human studies are surveyed together with their clinical implications. Explorations into detailing the relation between renal T2∗ and renal tissue partial pressure of oxygen (pO2 ) are discussed with a focus on factors confounding the T2∗ vs. tissue pO2 relation. Multi-modality in vivo approaches suitable for detailing the role of the confounding factors that govern T2∗ are considered. A schematic approach describing the link between renal perfusion, oxygenation, tissue compartments and renal T2∗ is proposed. Future directions of MRI assessment of renal oxygenation and perfusion are explored.
Collapse
Affiliation(s)
- T. Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin Germany
| | - A. Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin Germany
| | - K. Arakelyan
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin Germany
- Institute of Physiology and Center for Cardiovascular Research (CCR); Charité - Universitätsmedizin Berlin; Berlin Germany
| | - B. Flemming
- Institute of Physiology and Center for Cardiovascular Research (CCR); Charité - Universitätsmedizin Berlin; Berlin Germany
| | - K. Cantow
- Institute of Physiology and Center for Cardiovascular Research (CCR); Charité - Universitätsmedizin Berlin; Berlin Germany
| | - J. Hentschel
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin Germany
| | - D. Grosenick
- Physikalisch-Technische Bundesanstalt (PTB); Berlin Germany
| | - M. Ladwig
- Institute of Physiology and Center for Cardiovascular Research (CCR); Charité - Universitätsmedizin Berlin; Berlin Germany
| | - H. Reimann
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin Germany
| | - S. Klix
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin Germany
| | - S. Waiczies
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin Germany
| | - E. Seeliger
- Institute of Physiology and Center for Cardiovascular Research (CCR); Charité - Universitätsmedizin Berlin; Berlin Germany
| |
Collapse
|
30
|
Abstract
Establishing a programme for the prevention and treatment of acute kidney injury, chronic kidney disease and end-stage renal disease in a developing country involves unique challenges. We became involved in a collaborative effort to improve nephrology care in Haiti after participating in the emergency response to the 2010 earthquake. The focus of this ongoing project is overcoming barriers to implementation with the goal of improving training and resources for Haitian health-care workers and developing programmes for renal disease prevention and treatment in a setting of limited resources. Here, we offer practical advice for nephrologists who would like to help to advance medical care in developing countries. Rather than technical issues related to the prevention and treatment of renal disease, we focus on collaboration, education and the building of partnerships.
Collapse
|
31
|
Oron U, Tuby H, Maltz L, Sagi-Assif O, Abu-Hamed R, Yaakobi T, Doenyas-Barak K, Efrati S. Autologous bone-marrow stem cells stimulation reverses post-ischemic-reperfusion kidney injury in rats. Am J Nephrol 2014; 40:425-33. [PMID: 25413586 DOI: 10.1159/000368721] [Citation(s) in RCA: 257] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Low-level laser therapy (LLLT) has been found to modulate biological activity. The aim of the present study was to investigate the possible beneficial effects of LLLT application to stem cells in the bone marrow (BM), on the kidneys of rats that had undergone acute ischemia-reperfusion injury (IRI). METHODS Injury to the kidneys was induced by the excision of the left kidney and 60 min of IRI to the right kidney in each rat. Rats were then divided randomly into 2 groups: non-laser-treated and laser-treated. LLLT was applied to the BM 10 min and 24 h post-IRI and rats were sacrificed 4 days post-IRI. Blood was collected before the sacrifice and the kidney processed for histology. RESULTS Histological evaluation of kidney sections revealed the restored structural integrity of the renal tubules, and a significant reduction of 66% of pathological score in the laser-treated rats as compared to the non-laser-treated ones. C-kit positive cell density in kidneys post-IRI and laser-treatment was (p = 0.05) 2.4-fold higher compared to that of the non-laser treated group. Creatinine, blood urea nitrogen, and cystatin-C levels were significantly 55, 48, and 25% lower respectively in the laser-treated rats as compared to non-treated ones. CONCLUSION LLLT application to the BM causes induction of stem cells, which subsequently migrate and home in on the injured kidney. Consequently, a significant reduction in pathological features and improved kidney function post-IRI are evident. The results demonstrate a novel approach in cell-based therapy for acute ischemic injured kidneys.
Collapse
Affiliation(s)
- Uri Oron
- Department of Zoology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Chawla LS, Eggers PW, Star RA, Kimmel PL. Acute kidney injury and chronic kidney disease as interconnected syndromes. N Engl J Med 2014; 371:58-66. [PMID: 24988558 PMCID: PMC9720902 DOI: 10.1056/nejmra1214243] [Citation(s) in RCA: 1425] [Impact Index Per Article: 129.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lakhmir S Chawla
- From the Department of Medicine, Division of Intensive Care Medicine, and the Division of Nephrology, Washington, DC, Veterans Affairs Medical Center (L.S.C.), and the Department of Anesthesiology and Critical Care Medicine (L.S.C.) and Department of Medicine, Division of Renal Diseases and Hypertension (L.S.C., P.L.K.), George Washington University Medical Center - both in Washington, DC; and the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD (P.W.E., R.A.S., P.L.K.)
| | | | | | | |
Collapse
|
33
|
Pohlmann A, Cantow K, Hentschel J, Arakelyan K, Ladwig M, Flemming B, Hoff U, Persson PB, Seeliger E, Niendorf T. Linking non-invasive parametric MRI with invasive physiological measurements (MR-PHYSIOL): towards a hybrid and integrated approach for investigation of acute kidney injury in rats. Acta Physiol (Oxf) 2013; 207:673-89. [PMID: 23336404 DOI: 10.1111/apha.12065] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/17/2012] [Accepted: 01/16/2013] [Indexed: 01/11/2023]
Abstract
Acute kidney injury of various origins shares a common link in the pathophysiological chain of events: imbalance between renal medullary oxygen delivery and oxygen demand. For in vivo assessment of kidney haemodynamics and oxygenation in animals, quantitative but invasive physiological methods are established. A very limited number of studies attempted to link these invasive methods with parametric Magnetic Resonance Imaging (MRI) of the kidney. Moreover, the validity of parametric MRI (pMRI) as a surrogate marker for renal tissue perfusion and renal oxygenation has not been systematically examined yet. For this reason, we set out to combine invasive techniques and non-invasive MRI in an integrated hybrid setup (MR-PHYSIOL) with the ultimate goal to calibrate, monitor and interpret parametric MR and physiological parameters by means of standardized interventions. Here we present a first report on the current status of this multi-modality approach. For this purpose, we first highlight key characteristics of renal perfusion and oxygenation. Second, concepts for in vivo characterization of renal perfusion and oxygenation are surveyed together with the capabilities of MRI for probing blood oxygenation-dependent tissue stages. Practical concerns evoked by the use of strong magnetic fields in MRI and interferences between MRI and invasive physiological probes are discussed. Technical solutions that balance the needs of in vivo physiological measurements together with the constraints dictated by small bore MR scanners are presented. An early implementation of the integrated MR-PHYSIOL approach is demonstrated including brief interventions of hypoxia and hyperoxia.
Collapse
Affiliation(s)
- A. Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin; Germany
| | - K. Cantow
- Institute of Physiology; Charité - Universitätsmedizin Berlin; Campus Mitte, and Center for Cardiovascular Research; Berlin; Germany
| | - J. Hentschel
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max Delbrück Center for Molecular Medicine; Berlin; Germany
| | | | - M. Ladwig
- Institute of Physiology; Charité - Universitätsmedizin Berlin; Campus Mitte, and Center for Cardiovascular Research; Berlin; Germany
| | - B. Flemming
- Institute of Physiology; Charité - Universitätsmedizin Berlin; Campus Mitte, and Center for Cardiovascular Research; Berlin; Germany
| | - U. Hoff
- Nephrology and Intensive Care Medicine; Charité - Universitätsmedizin Berlin; Campus Virchow-Klinikum, and Center for Cardiovascular Research; Berlin; Germany
| | - P. B. Persson
- Institute of Physiology; Charité - Universitätsmedizin Berlin; Campus Mitte, and Center for Cardiovascular Research; Berlin; Germany
| | - E. Seeliger
- Institute of Physiology; Charité - Universitätsmedizin Berlin; Campus Mitte, and Center for Cardiovascular Research; Berlin; Germany
| | | |
Collapse
|
34
|
Bolisetty S, Jaimes EA. Mitochondria and reactive oxygen species: physiology and pathophysiology. Int J Mol Sci 2013; 14:6306-44. [PMID: 23528859 PMCID: PMC3634422 DOI: 10.3390/ijms14036306] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/08/2013] [Accepted: 03/11/2013] [Indexed: 02/06/2023] Open
Abstract
The air that we breathe contains nearly 21% oxygen, most of which is utilized by mitochondria during respiration. While we cannot live without it, it was perceived as a bane to aerobic organisms due to the generation of reactive oxygen and nitrogen metabolites by mitochondria and other cellular compartments. However, this dogma was challenged when these species were demonstrated to modulate cellular responses through altering signaling pathways. In fact, since this discovery of a dichotomous role of reactive species in immune function and signal transduction, research in this field grew at an exponential pace and the pursuit for mechanisms involved began. Due to a significant number of review articles present on the reactive species mediated cell death, we have focused on emerging novel pathways such as autophagy, signaling and maintenance of the mitochondrial network. Despite its role in several processes, increased reactive species generation has been associated with the origin and pathogenesis of a plethora of diseases. While it is tempting to speculate that anti-oxidant therapy would protect against these disorders, growing evidence suggests that this may not be true. This further supports our belief that these reactive species play a fundamental role in maintenance of cellular and tissue homeostasis.
Collapse
Affiliation(s)
- Subhashini Bolisetty
- Nephrology Division, University of Alabama at Birmingham, Birmingham, AL 35294, USA; E-Mail:
| | - Edgar A. Jaimes
- Nephrology Division, University of Alabama at Birmingham, Birmingham, AL 35294, USA; E-Mail:
- Veterans Affairs Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
35
|
Poureetezadi SJ, Wingert RA. Congenital and Acute Kidney Disease: Translational Research Insights from Zebrafish Chemical Genetics. ACTA ACUST UNITED AC 2013; 1:112. [PMID: 24653992 DOI: 10.4172/2327-5146.1000112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Today, acute kidney injury (AKI) and congenital anomalies of the kidney and urinary tract (CAKUT) represent major issues in healthcare. Both AKI and CAKUT can lead to end stage renal disease (ESRD) that requires life-long medical care with renal replacement therapy. Renal replacement by dialysis is intensive, and kidney transplantation is restricted by organ availability. These limitations, along with the growing epidemic of patients affected by kidney disease, highlight the significant need to identify alternative ways to treat renal injury and birth defects. Drug discovery is one promising avenue of current research. Here, we discuss zebrafish chemical genetics and its latent potency as a method to rapidly identify small molecule therapeutics to accelerate recovery after AKI. Specifically, we review two groundbreaking studies that have recently provided a template to screen for compounds that expand the renal progenitor field in development that were capable of treating AKI in both the zebrafish and the mouse. These new findings demonstrate that drug discovery using zebrafish can be used for relevant translational research to identify clinical interventions for renal conditions in humans.
Collapse
Affiliation(s)
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
36
|
|