1
|
Lichtenberger FB, Xu M, Erdoğan C, Fei L, Mathar I, Dietz L, Sandner P, Seeliger E, Boral S, Bonk JS, Sieckmann T, Persson PB, Patzak A, Cantow K, Khedkar PH. Activating soluble guanylyl cyclase attenuates ischemic kidney damage. Kidney Int 2025; 107:476-491. [PMID: 39571904 DOI: 10.1016/j.kint.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/21/2024] [Accepted: 10/31/2024] [Indexed: 12/22/2024]
Abstract
Can direct activation of soluble guanylyl cyclase (sGC) provide kidney-protection? To answer this, we tested the kidney-protective effects of a sGC activator, which functions independent of nitric oxide and with oxidized sGC, in an acute kidney injury (AKI) model with transition to chronic kidney disease (CKD). We hypothesize this treatment would provide protection of kidney microvasculature, kidney blood flow, fibrosis, inflammation, and kidney damage. Assessment took place on days three, seven, 14 (acute phase) and 84 (late phase) after unilateral ischemia reperfusion injury (IRI) in rats. Post-ischemia, animals received vehicle or the sGC activator BAY 60-2770 orally. In the vehicle group, medullary microvessels narrowed and cortical microvessels showed hypertrophic inward remodeling. The mRNA levels of acute injury markers (Kim-1, Ngal) were high in the acute phase but declined in the late phase. Kidney weight decreased after the acute phase, while fibrosis started after day seven. Abundance of fibrotic (Col1a, Tgf-β1) and inflammatory markers (Il-6, Tnf-α) remained elevated throughout, along with mononuclear cell invasion, with elevated plasma cystatin C and creatinine. BAY 60-2770 treatment increased tissue cGMP concentration, dilated kidney microvasculature, and enhanced blood flow and oxygenation. This intervention significantly attenuated kidney weight loss, cell damage, fibrosis, and inflammation. Plasma cystatin C and creatinine improved significantly with sGC activator treatment indicating functional recovery, though possible GFR increase above kidney reserve in uninjured kidneys could not be excluded. In cultured human tubular cells (HK-2 cells) exposed to hypoxia or profibrotic TGF-β, BAY 60-2770 improved abundance patterns of pathologically relevant genes. Overall, our results show that sGC activation may provide effective kidney-protection and attenuate the AKI-to-CKD transition.
Collapse
Affiliation(s)
- Falk-Bach Lichtenberger
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Minze Xu
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Cem Erdoğan
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lingyan Fei
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Nephrology, Center of Kidney and Urology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ilka Mathar
- Cardiovascular Research, Bayer AG Pharmaceuticals, Wuppertal, Germany
| | - Lisa Dietz
- Cardiovascular Research, Bayer AG Pharmaceuticals, Wuppertal, Germany
| | - Peter Sandner
- Cardiovascular Research, Bayer AG Pharmaceuticals, Wuppertal, Germany
| | - Erdmann Seeliger
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sengül Boral
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Sophie Bonk
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tobias Sieckmann
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pontus B Persson
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Patzak
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kathleen Cantow
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pratik H Khedkar
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
2
|
Su CY, Chang KF, Hsiao CY, Tsai NM. Neng-Jing-Huo Essential Oil Blend Inhibits Lipopolysaccharide-Induced Intracellular Reactive Oxygen Species Accumulation, Inflammation, and Apoptosis in Renal Tubular Epithelial Cells. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:57-66. [PMID: 39846315 DOI: 10.4103/ejpi.ejpi-d-24-00096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025]
Abstract
ABSTRACT Acute kidney injury (AKI) is a common serious complication of sepsis that is characterized by the rapid deterioration of kidney function. Neng-Jing-Huo (NJH) is an essential oil blend, including Gaultheria procumbens, Zingiber officinale, Bulnesia sarmientoi, Artemisia vulgaris , and Styrax benzoin oils, with antimicrobial, antioxidant, and anti-inflammatory activities. Here, we investigated the effects of NJH on oxidative stress, inflammatory response, and apoptosis in an in vitro septic AKI model and explored the underlying mechanisms. A cellular model of septic AKI was established using lipopolysaccharide (LPS). Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Action mechanisms of NJH were analyzed using the Griess reagent, 2',7'-dichlorodihydrofluorescein diacetate, 5,5',6,6' tetrachloro-1,1'3,3' tetraethylbenzimidazolcarbocyanine iodide, annexin V, caspase activity, western blotting, and semi-quantitative reverse transcription polymerase chain reaction assays. Results showed that pretreatment with NJH significantly improved cell survival and suppressed nitric oxide (NO) production in LPS-stimulated NRK-52E renal tubular epithelial cells. NJH also decreased the levels of intracellular reactive oxygen species and maintained the mitochondrial membrane potential by upregulating the nuclear factor (NF) erythroid 2-related factor 2/heme oxygenase-1 levels and downregulating the NADPH oxidase 4 levels. In addition, NJH suppressed the activation of the toll-like receptor 4/NF-κB and NLRP3/caspase-1 pathways, thereby decreasing the inflammatory response in LPS-stimulated NRK-52E cells. Moreover, NJH decreased the levels of Bax, caspase-9, and caspase-3 but increased those of Bcl-2, which led to a reduction in LPS-induced apoptosis. Overall, our findings revealed that NJH ameliorated LPS-induced damage in NRK-52E cells by inhibiting oxidative stress, inflammation, and apoptosis, highlighting its therapeutic potential for septic AKI.
Collapse
Affiliation(s)
- Chin-Ya Su
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Kai-Fu Chang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Yen Hsiao
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
- Department of Life-and-Death Studies, Nanhua University, Chiayi, Taiwan
| |
Collapse
|
3
|
Pu M, Zhao H, Xu S, Gu X, Feng Q, Huang P. Urine miR-340-5p Predicts the Adverse Prognosis of Sepsis-Associated Acute Kidney Injury and Regulates Renal Tubular Epithelial Cell Injury by Targeting KDM4C. Nephron Clin Pract 2024; 149:197-206. [PMID: 39551047 DOI: 10.1159/000541348] [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: 02/01/2024] [Accepted: 08/19/2024] [Indexed: 11/19/2024] Open
Abstract
INTRODUCTION Sepsis-associated acute kidney injury (SA-AKI) is a common complication of sepsis. miR-340-5p has been identified as an effective biomarker of various human diseases. As the downstream target, the involvement of lysine (K)-specific demethylase 4C (KDM4C) in SA-AKI would help interpret the regulatory mechanism of miR-340-5p. The significance of miR-340-5p in the onset and progression of SA-AKI was evaluated to provide a potential therapeutic target for SA-AKI. METHODS This study enrolled 64 healthy individuals (control) and 159 sepsis patients (92 SA-AKI and 67 non-AKI) and collected urine samples. The urine level of miR-340-5p was analyzed by PCR, and a series of statistical analyses were conducted to assess the clinical significance of miR-340-5p in the occurrence and development of SA-AKI. The injured renal tubular epithelial cells were established with LPS induction. The roles of miR-340-5p in cellular processes were evaluated. RESULTS Increasing urine miR-340-5p discriminated SA-AKI patients from healthy individuals (AUC = 0.934) and non-AKI sepsis patients (AUC = 0.806) sensitively. Additionally, elevated miR-340-5p could predict the adverse prognosis (HR = 5.128, 95% CI = 1.259-20.892) and malignant development of SA-AKI patients. In vitro, lipopolysaccharide (LPS) also induced an increased level of miR-340-5p and significant cell injury in the renal tubular epithelial cell; silencing miR-340-5p could alleviate the suppressed proliferation, migration, and invasion caused by LPS. In mechanism, miR-340-5p negatively regulated KDM4C, which mediated the function of miR-340-5p. CONCLUSION miR-340-5p served as a diagnostic and prognostic biomarker of SA-AKI and regulated renal tubular epithelial cell injury via modulating KDM4C.
Collapse
Affiliation(s)
- Mengmeng Pu
- Department of Nephrology, Xingtai People's Hospital, Xingtai, China
| | - Huanhuan Zhao
- Department of Nephrology, Jinan Weigao Nephrology Hospital, Jinan, China
| | - Silei Xu
- Medical School of University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaohui Gu
- Department of Urinary Surgery, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Qiang Feng
- Department of Urinary Surgery, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Peng Huang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Research on Prevention and Control of High Incidence Diseases in Western Guangxi, Baise, China
| |
Collapse
|
4
|
Çomaklı S, Küçükler S, Değirmençay Ş, Bolat İ, Özdemir S. Quinacrine, a PLA2 inhibitor, alleviates LPS-induced acute kidney injury in rats: Involvement of TLR4/NF-κB/TNF α-mediated signaling. Int Immunopharmacol 2024; 126:111264. [PMID: 38016342 DOI: 10.1016/j.intimp.2023.111264] [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: 09/19/2023] [Revised: 10/30/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023]
Abstract
Acute Kidney Injury (AKI) is a major factor in sepsis-related mortality and may occur due to lipopolysaccharide (LPS), an endotoxin produced by gram-negative bacteria that triggers a systemic acute inflammatory response. Quinacrine's (QC) renoprotective properties in sepsis and the underlying mechanism, however, are still not fully understood. This study was done to investigate the anti-inflammatory, antioxidative, and anti-apoptotic effects of QC, a phospholipase A2 (PLA2) inhibitor, against LPS-induced AKI. Rats were randomly divided into five groups: control group, QC30 group, LPS group, LPS+QC 10 group, and LPS+QC 30 group. The rats were administered intraperitoneally QC (10 and 30 mg/kg) for 3 days (once a day) prior to injection of LPS (3 mg/kg). Six hours after the LPS injection, the histopathological changes, oxidative stress, inflammation, and apoptosis in the collected kidney tissues were detected by hematoxylin and eosin staining, enzyme-linked immunosorbent assay (ELISA), real-time PCR (RT-PCR), and immunohistochemistry staining, respectively. QC pretreatment could successfully attenuate LPS-induced AKI, as evidenced by a decrease in tissue histopathological injury. Meanwhile, QC alleviated LPS-induced kidney oxidative stress; it reduced MDA levels and increased levels of SOD, CAT, GPX, and GSH. LPS-induced elevations in kidney TLR4, NF-κB, TNF-α, IL-1β, IL-6, PLA2, caspase 3, and Bax contents were significantly attenuated in QC-treated groups. Our findings revealed a significant effect of QC: protecting against LPS-induced AKI through inhibition of PLA2 and decreasing inflammation, oxidative stress, and apoptosis. To treat LPS-induced AKI, QC may be an effective substance with an excellent protection profile.
Collapse
Affiliation(s)
- Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Sefa Küçükler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Şükrü Değirmençay
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - İsmail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey; German Center for Neurodegenerative Diseases, DZNE, Bonn, Germany.
| |
Collapse
|
5
|
Almazmomi MA, Esmat A, Naeem A. Acute Kidney Injury: Definition, Management, and Promising Therapeutic Target. Cureus 2023; 15:e51228. [PMID: 38283512 PMCID: PMC10821757 DOI: 10.7759/cureus.51228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Acute kidney injury (AKI) is caused by a sudden loss of renal function, resulting in the build-up of waste products and a significant increase in mortality and morbidity. It is commonly diagnosed in critically ill patients, with its occurrence estimated at up to 50% in patients hospitalized in the intensive critical unit. Despite ongoing efforts, the death rate associated with AKI has remained high over the past half-century. Thus, it is critical to investigate novel therapy options for preventing the epidemic. Many studies have found that inflammation and Toll-like receptor-4 (TLR-4) activation have a significant role in the pathogenesis of AKI. Noteworthy, challenges in the search for efficient pharmacological therapy for AKI have arisen due to the multifaceted origin and complexity of the clinical history of people with the disease. This article focuses on kidney injury's epidemiology, risk factors, and pathophysiological processes. Specifically, it focuses on the role of TLRs especially type 4 in disease development.
Collapse
Affiliation(s)
- Meaad A Almazmomi
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Ahmed Esmat
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Anjum Naeem
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
| |
Collapse
|
6
|
Forsse JS, Richardson KA, Torres R, Lowry C, Taylor JK, Beeson CL, Ward J, Dhillon A, Niceler B, Ismaeel A, Koutakis P. Exploring an Unknown Corner of a Well-Known Topic: HIIE Influence on Renal Health and Filtration in Healthy Individuals Free of Cardiometabolic Diseases. Sports (Basel) 2023; 11:210. [PMID: 37999427 PMCID: PMC10675315 DOI: 10.3390/sports11110210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
Aerobic exercise, specifically high-intensity interval exercise (HIIE), and its effects on renal health and filtration (RHF) are not well understood. Several studies support incorporating contemporary biomarkers serum cystatin C (CyC) and urine epidermal growth factor (uEGF) to combat the volatility of serum creatinine (sCr). Using these biomarkers, we examined the acute influences HIIE has on RHF to determine if there is a ceiling effect in healthy populations. The purpose was to determine the influence of an acute bout of HIIE on RHF. Thirty-six participants (n = 22 males; n = 14 females; age 37.6 ± 12.4 years.; BF% 19.2 ± 7.1%; VO2max 41.8 + 7.4 mL/kg/min) completed 30 min of HIIE on a treadmill (80% and 40% of VO2reserve in 3:2 min ratio). Blood and urine samples were obtained under standardized conditions before, 1 h, and 24 h post-exercise. CyC, sCR, uEGF, urine creatinine (uCr), uCr/uEGF ratio, and multiple estimates of glomerular filtration rate (eGFR) Modification of Diet in Renal Disease (MDRD) and CKD-EPI equations were used. The analysis employed paired sample t-tests and repeated measures ANOVAs. CyC, uEGF, uCr, and uCr/uEGF ratio concentrations were not altered between timepoints. sCr increased 1 h post-exercise (p > 0.002) but not at 24 h post-exercise. eGFR decreased in the MDRD and CKD-EPI equations at 1 h (p > 0.012) with no changes at 24 h post-exercise. CyC and sCr/CyC demonstrated no significant changes. CyC and uEGF are not altered by acute HIIE. The results demonstrate a potential ceiling effect in contemporary and traditional biomarkers of RHF, indicating improvements in RHF may be isolated to populations with reduced kidney function.
Collapse
Affiliation(s)
- Jeffrey S. Forsse
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Kathleen A. Richardson
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Ricardo Torres
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Catherine Lowry
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO 80011, USA
| | - James Kyle Taylor
- Medical & Clinical Laboratory Sciences, Auburn University—Montgomery, Montgomery, AL 36124, USA;
| | - Cassidy L. Beeson
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Jacob Ward
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Southern Illinois University Medical School, Lindegren Hall, 600 Agriculture Dr #132, Carbondale, IL 62901, USA
| | - Anurag Dhillon
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Health Science Center, San Antonio Joe R and Teresa Lozano Long School of Medicine, The University of Texas, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Brock Niceler
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Waco Family Medicine, Waco, TX 76707, USA
| | - Ahmed Ismaeel
- Department of Physiology, University of Kentucky, 780 Rose Street, MS508, Lexington, KY 40536, USA;
- Clinical Muscle Biology Lab, Baylor University, Waco, TX 76706, USA;
| | | |
Collapse
|
7
|
Gleeson PJ, Crippa IA, Sannier A, Koopmansch C, Bienfait L, Allard J, Sexton DJ, Fontana V, Rorive S, Vincent JL, Creteur J, Taccone FS. Critically ill patients with acute kidney injury: clinical determinants and post-mortem histology. Clin Kidney J 2023; 16:1664-1673. [PMID: 37779855 PMCID: PMC10539222 DOI: 10.1093/ckj/sfad113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Indexed: 10/03/2023] Open
Abstract
Background Acute kidney injury (AKI) requiring renal replacement therapy (RRT) in the intensive care unit (ICU) portends a poor prognosis. We aimed to better characterize predictors of survival and the mechanism of kidney failure in these patients. Methods This was a retrospective observational study using clinical and radiological electronic health records, analysed by univariable and multivariable binary logistic regression. Histopathological examination of post-mortem renal tissue was performed. Results Among 157 patients with AKI requiring RRT, higher serum creatinine at RRT initiation associated with increased ICU survival [odds ratio (OR) 0.33, 95% confidence interval (CI) 0.17-0.62, P = .001]; however, muscle mass (a marker of frailty) interacted with creatinine (P = .02) and superseded creatinine as a predictor of survival (OR 0.26, 95% CI 0.08-0.82; P = .02). Achieving lower cumulative fluid balance (mL/kg) predicted ICU survival (OR 1.01, 95% CI 1.00-1.01, P < .001), as supported by sensitivity analyses showing improved ICU survival with the use of furosemide (OR 0.40, 95% CI 0.18-0.87, P = .02) and increasing net ultrafiltration (OR 0.97, 95% CI 0.95-0.99, P = .02). A urine output of >500 mL/24 h strongly predicted successful liberation from RRT (OR 0.125, 95% CI 0.05-0.35, P < .001). Post-mortem reports were available for 32 patients; clinically unrecognized renal findings were described in 6 patients, 1 of whom had interstitial nephritis. Experimental staining of renal tissue from patients with sepsis-associated AKI (S-AKI) showed glomerular loss of synaptopodin (P = .02). Conclusions Confounding of creatinine by muscle mass undermines its use as a marker of AKI severity in clinical studies. Volume management and urine output are key determinants of outcome. Loss of synaptopodin implicates glomerular injury in the pathogenesis of S-AKI.
Collapse
Affiliation(s)
- Patrick James Gleeson
- Department of Intensive Care Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Université de Paris Cité, INSERM UMR1149 & CNRS EMR8252, Centre de Recherche sur l'Inflammation, Inflamex Laboratory of Excellence, Paris, France
- Department of Renal Medicine, Cork University Hospital, Cork, Ireland
| | - Ilaria Alice Crippa
- Department of Intensive Care Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Department of Anesthesiology, San Marco Hospital, San Donato Group, Zingonia, Bergame, Italy
| | - Aurélie Sannier
- AP-HP, Nord/Université de Paris, Hôpital Bichat-Claude Bernard, Service d'Anatomie-Pathologique, Paris, France
| | - Caroline Koopmansch
- Department of Pathology, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Institut de Pathologie et de Génétique, Avenue George Lemaître, Gosselies, Belgium
| | - Lucie Bienfait
- Department of Pathology, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Justine Allard
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles, Gosselies, Belgium
| | - Donal J Sexton
- Trinity Health Kidney Center, Trinity College Dublin, Dublin, Ireland
- Department of Nephrology, St James’ Hospital, Dublin, Ireland
| | - Vito Fontana
- Department of Intensive Care Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Department of Intensive Care Medicine, Clinique Saint-Jean, Brussels, Belgium
| | - Sandrine Rorive
- Department of Pathology, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Louis Vincent
- Department of Intensive Care Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Fabio Silvio Taccone
- Department of Intensive Care Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
8
|
Yang G, Tan L, Yao H, Xiong Z, Wu J, Huang X. Long-Term Effects of Severe Burns on the Kidneys: Research Advances and Potential Therapeutic Approaches. J Inflamm Res 2023; 16:1905-1921. [PMID: 37152866 PMCID: PMC10162109 DOI: 10.2147/jir.s404983] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023] Open
Abstract
Burns are a seriously underestimated form of trauma that not only damage the skin system but also cause various complications, such as acute kidney injury (AKI). Recent clinical studies have shown that the proportion of chronic kidney diseases (CKD) in burn patients after discharge is significantly higher than that in the general population, but the mechanism behind this is controversial. The traditional view is that CKD is associated with hypoperfusion, AKI, sepsis, and drugs administered in the early stages of burns. However, recent studies have shown that burns can cause long-term immune dysfunction, which is a high-risk factor for CKD. This suggests that burns affect the kidneys more than previously recognized. In other words, severe burns are not only an acute injury but also a chronic disease. Neglecting to study long-term kidney function in burn patients also results in a lack of preventive and therapeutic methods being developed. Furthermore, stem cells and their exosomes have shown excellent comprehensive therapeutic properties in the prevention and treatment of CKD, making them increasingly the focus of research attention. Their engineering strategy further improved the therapeutic performance. This review will focus on the research advances in burns on the development of CKD, illustrating the possible mechanism of burn-induced CKD and introducing potential biological treatment options and their engineering strategies.
Collapse
Affiliation(s)
- Guang Yang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518000, People’s Republic of China
- Department of Life Sciences, Yuncheng University, Yuncheng, 044006, People’s Republic of China
| | - Lishan Tan
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518000, People’s Republic of China
| | - Hua Yao
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541004, People’s Republic of China
| | - Zuying Xiong
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518000, People’s Republic of China
| | - Jun Wu
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, People’s Republic of China
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Pediatrics & Gynecology, University of Verona Medical School, Verona, Venetia, 37134, Italy
| | - Xiaoyan Huang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518000, People’s Republic of China
| |
Collapse
|
9
|
Abstract
An increased intraabdominal pressure, particularly when occurring during periods of hemodynamic instability or fluid overload, is regarded as a major contributor to acute kidney injury (AKI) in intensive care units. During abdominal laparoscopic procedures, intraoperative insufflation pressures up to 15 mmHg are applied, to enable visualization and surgical manipulation but with the potential to compromise net renal perfusion. Despite the widely acknowledged renal arterial autoregulation, net arterial perfusion pressure is known to be narrow, and the effective renal medullary perfusion is disproportionately impacted by venous and lymphatic congestion. At present, the potential risk factors, mitigators and risk-stratification of AKI during surgical pneumoperitoneum formation received relatively limited attention among nephrologists and represent an opportunity to look beyond mere blood pressure and intake-output balances. Careful charting and reporting duration and extent of surgical pneumoperitoneum represents an opportunity for anesthesia teams to better communicate intraoperative factors affecting renal outcomes for the postoperative clinical teams. In this current article, the authors are integrating preclinical data and clinical experience to provide a better understanding to optimize renal perfusion during surgeries. Future studies should carefully consider intrabdominal insufflation pressure as a key variable when assessing outcomes and blood pressure goals in these settings.
Collapse
|
10
|
Maciel AT, Vitorio D, Osawa EA. Urine biochemistry assessment in the sequential evaluation of renal function: Time to think outside the box. Front Med (Lausanne) 2022; 9:912877. [PMID: 35957852 PMCID: PMC9360530 DOI: 10.3389/fmed.2022.912877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
Urine biochemistry (UB) remains a controversial tool in acute kidney injury (AKI) monitoring, being considered to be of limited value both in terms of AKI diagnosis and prognosis. However, many criticisms can be made to the studies that have established the so called “pre-renal paradigm” (used for decades as the essential physiological basis for UB assessment in AKI) as well as to more recent studies suggesting that UB has no utility in daily clinical practice. The aim of this article is to describe our hypothesis on how to interpret simple and widely recognized urine biochemical parameters from a novel perspective, propose the rationale for their sequential assessment and demonstrate their usefulness in AKI monitoring, especially in the critical care setting.
Collapse
Affiliation(s)
- Alexandre T. Maciel
- Research Department, Imed Group, São Paulo, Brazil
- Adult Intensive Care Unit, São Camilo Hospital–Pompéia Unit, São Paulo, Brazil
- *Correspondence: Alexandre T. Maciel,
| | - Daniel Vitorio
- Research Department, Imed Group, São Paulo, Brazil
- Adult Intensive Care Unit, São Camilo Hospital–Pompéia Unit, São Paulo, Brazil
| | - Eduardo A. Osawa
- Research Department, Imed Group, São Paulo, Brazil
- Adult Intensive Care Unit, São Camilo Hospital–Pompéia Unit, São Paulo, Brazil
| |
Collapse
|
11
|
Mediators of Regional Kidney Perfusion during Surgical Pneumo-Peritoneum Creation and the Risk of Acute Kidney Injury—A Review of Basic Physiology. J Clin Med 2022; 11:jcm11102728. [PMID: 35628855 PMCID: PMC9142947 DOI: 10.3390/jcm11102728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
Acute kidney injury (AKI), especially if recurring, represents a risk factor for future chronic kidney disease. In intensive care units, increased intra-abdominal pressure is well-recognized as a significant contributor to AKI. However, the importance of transiently increased intra-abdominal pressures procedures is less commonly appreciated during laparoscopic surgery, the use of which has rapidly increased over the last few decades. Unlike the well-known autoregulation of the renal cortical circulation, medulla perfusion is modulated via partially independent regulatory mechanisms and strongly impacted by changes in venous and lymphatic pressures. In our review paper, we will provide a comprehensive overview of this evolving topic, covering a broad range from basic pathophysiology up to and including current clinical relevance and examples. Key regulators of oxidative stress such as ischemia-reperfusion injury, the activation of inflammatory response and humoral changes interacting with procedural pneumo-peritoneum formation and AKI risk will be recounted. Moreover, we present an in-depth review of the interaction of pneumo-peritoneum formation with general anesthetic agents and animal models of congestive heart failure. A better understanding of the relationship between pneumo-peritoneum formation and renal perfusion will support basic and clinical research, leading to improved clinical care and collaboration among specialists.
Collapse
|
12
|
Abstract
The definition of sepsis has evolved significantly over the past three decades. Today, sepsis is defined as a dysregulated host immune response to microbial invasion leading to end organ dysfunction. Septic shock is characterized by hypotension requiring vasopressors after adequate fluid resuscitation with elevated lactate. Early recognition and intervention remain hallmarks for sepsis management. We addressed the current literature and assimilated thought regarding optimum initial resuscitation of the patient with sepsis. A nuanced understanding of the physiology of lactate is provided in our review. Physiologic and practical knowledge of steroid and vasopressor therapy for sepsis is crucial and addressed. As blood purification may interest the nephrologist treating sepsis, we have also added a brief discussion of its status.
Collapse
Affiliation(s)
- Sharad Patel
- Department of Critical Care, Rowan University Cooper Medical School, Camden, New Jersey
| | - Nitin Puri
- Department of Critical Care, Cooper Hospital University Medical Center, Camden, New Jersey
| | - R Phillip Dellinger
- Department of Critical Care, Cooper Hospital University Medical Center, Camden, New Jersey
| |
Collapse
|
13
|
Xie Z, Wei L, Chen J, Chen Z. Calcium dobesilate alleviates renal dysfunction and inflammation by targeting nuclear factor kappa B (NF-κB) signaling in sepsis-associated acute kidney injury. Bioengineered 2022; 13:2816-2826. [PMID: 35038964 PMCID: PMC8974157 DOI: 10.1080/21655979.2021.2024394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is a serious complication of sepsis that increases mortality and the risk of progression to chronic kidney disease. Oxidative stress and apoptosis are reported to exert critical function in the pathogenesis of sepsis-associated AKI. Calcium dobesilate (CaD) was reported to play a protective role in renal diseases. Therefore, we explored the antioxidant effect and potential mechanism of CaD in lipopolysaccharide (LPS)-induced AKI in mice. We evaluated renal function (blood urea nitrogen (BUN) and serum creatinine (SCr)), histopathology, oxidative stress (superoxide dismutase (SOD) and malondialdehyde (MDA)), inflammation cytokines, and apoptosis in kidneys of mice. The effect of CaD on NF-κB signaling was evaluated by Western blot. Our findings showed that CaD alleviated renal dysfunction and kidney injury, and also reversed upregulated MDA concentration and reduced SOD enzyme activity in AKI mice. Moreover, LPS-induced inflammatory response was attenuated by CaD. CaD treatment also reduced the apoptosis evoked by LPS. Additionally, CaD downregulated phosphorylation of nuclear factor kappa B (NF-κB) signaling components in LPS mice. Conclusively, CaD alleviates renal dysfunction and inflammation by targeting NF-κB signaling in sepsis-associated AKI.
Collapse
Affiliation(s)
- Zhijuan Xie
- Department of Nephrology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Lanji Wei
- Department of Health Management Center, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Jianying Chen
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Zhong Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| |
Collapse
|
14
|
Milanez M, Liberatore A, Nishi E, Bergamaschi C, Campos R, Koh I. Patterns of renal and splanchnic sympathetic vasomotor activity in an animal model of survival to experimental sepsis. Braz J Med Biol Res 2022; 55:e11873. [PMID: 35043862 PMCID: PMC8852156 DOI: 10.1590/1414-431x2021e11873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | - E.E. Nishi
- Universidade Federal de São Paulo, Brasil
| | | | | | - I.H.J. Koh
- Universidade Federal de São Paulo, Brasil
| |
Collapse
|
15
|
Rosales TO, Horewicz VV, Ferreira MA, Nardi GM, Assreuy J. Dynamics of GRK2 in the kidney: a putative mechanism for sepsis-associated kidney injury. Clin Sci (Lond) 2021; 135:2341-2356. [PMID: 34622918 DOI: 10.1042/cs20210462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/24/2021] [Accepted: 10/07/2021] [Indexed: 01/09/2023]
Abstract
Renal vascular reactivity to vasoconstrictors is preserved in sepsis in opposition to what happens in the systemic circulation. We studied whether this distinct behavior was related to α1 adrenergic receptor density, G protein-coupled receptor kinase 2 (GRK2) and the putative role of nitric oxide (NO). Sepsis was induced in female mice by cecal ligation and puncture (CLP). Wildtype mice were treated with prazosin 12 h after CLP or nitric oxide synthase 2 (NOS-2) inhibitor, 30 min before and 6 and 12 h after CLP. In vivo experiments and biochemistry assays were performed 24 h after CLP. Sepsis decreased the systemic mean arterial pressure (MAP) and the vascular reactivity to phenylephrine. Sepsis also reduced basal renal blood flow which was normalized by treatment with prazosin. Sepsis led to a substantial decrease in GRK2 level associated with an increase in α1 adrenergic receptor density in the kidney. The disappearance of renal GRK2 was prevented in NOS-2-KO mice or mice treated with 1400 W. Treatment of non-septic mice with an NO donor reduced GRK2 content in the kidney. Therefore, our results show that an NO-dependent reduction in GRK2 level in the kidney leads to the maintenance of a normal α1 adrenergic receptor density. The preservation of the density and/or functionality of this receptor in the kidney together with a higher vasoconstrictor tonus in sepsis lead to vasoconstriction. Thus, the increased concentration of vasoconstrictor mediators together with the preservation (and even increase) of the response to them may help to explain sepsis-induced acute kidney injury.
Collapse
Affiliation(s)
| | | | | | - Geisson Marcos Nardi
- Department of Morphological Sciences, Universidade Federal de Santa Catarina, SC, Brazil
| | - Jamil Assreuy
- Department of Pharmacology, Universidade Federal de Santa Catarina, SC, Brazil
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW This review discusses the macrocirculatory and microcirculatory aspects of renal perfusion, as well as novel methods by which to measure renal blood flow. Finally, therapeutic options are briefly discussed, including renal-specific microcirculatory effects. RECENT FINDINGS The optimal mean arterial pressure (MAP) needed for preservation of renal function has been debated but is most likely a MAP of 60-80 mmHg. In addition, attention should be paid to renal outflow pressure, typically central venous pressure. Heterogeneity in microcirculation can exist and may be mitigated through appropriate use of vasopressors with unique microcirculatory effects. Excessive catecholamines have been shown to be harmful and should be avoided. Both angiotensin II and vasopressin may improve glomerular flow through a number of mechanisms. Macrocirculatory and microcirculatory blood flow can be measured through a number of bedside ultrasound modalities, sublingual microscopy and urinary oxygen measurement, SUMMARY: Acute kidney injury (AKI) is a common manifestation of organ failure in shock, and avoidance of hemodynamic instability can mitigate this risk. Measurement of renal haemodynamics is not routinely performed but may help to guide therapeutic goals. A thorough understanding of pathophysiology, measurement techniques and therapeutic options may allow for a personalized approach to blood pressure management in patients with septic shock and may ultimately mitigate AKI.
Collapse
|
17
|
Bitker L, Patel SK, Bittar I, Eastwood GM, Bellomo R, Burrell LM. Reduced urinary levels of angiotensin-converting enzyme 2 activity predict acute kidney injury in critically ill patients. CRIT CARE RESUSC 2020; 22:344-354. [PMID: 38046883 PMCID: PMC10692539 DOI: 10.51893/2020.4.oa7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective: Angiotensin-converting enzyme 2 activity reflects non-classical renin-angiotensin system upregulation. We assessed the association of urinary angiotensin-converting enzyme 2 (uACE2) activity with acute kidney injury (AKI). Design, setting and participants: A prospective observational study in which we measured uACE2 activity in 105 critically ill patients at risk of AKI. We report AKI stage 2 or 3 at 12 hours of urine collection (AKI12h) and AKI stage 2 or 3 at any time during intensive care unit stay in patients free from any stage of AKI at inclusion (AKIICU). AKI prediction was assessed using area under the receiver-operating characteristics curve (AUROC) and net reclassification indices (NRIs). Main outcome measure: AKI stage 2 or 3 at 12 hours of urine collection. Results: Within 12 hours of inclusion, 32 of 105 patients (30%) had developed AKI12h. Corrected uACE2 activity was significantly higher in patients without AKI12h compared with those with AKI12h (median [interquartile range], 13 [6-24] v 7 [4-10] pmol/min/mL per mmol/L of urine creatinine; P < 0.01). A 10-unit increase in uACE2 was associated with a 28% decrease in AKI12h risk (odds ratio [95% CI], 0.72 [0.46-0.97]). During intensive care unit admission, 39 of 76 patients (51%) developed AKIICU. uACE2 had an AUROC for the prediction of AKI12h of 0.68 (95% CI, 0.57-0.79), and correctly reclassified 28% of patients (positive NRI) to AKI12h. Patients with uACE2 > 8.7 pmol/min/mL per mmol/L of urine creatinine had a significantly lower risk of AKIICU on log-rank analysis (52% v 84%; P < 0.01). Conclusions: Higher uACE2 activity was associated with a decreased risk of AKI stage 2 or 3. Our findings support future evaluations of the role of the non-classical renin-angiotensin system during AKI.
Collapse
Affiliation(s)
- Laurent Bitker
- Department of Intensive Care, Austin Health, Melbourne, VIC, Australia
- Université de Lyon, Lyon, France
| | - Sheila K. Patel
- Department of Medicine, Austin Health, Melbourne, VIC, Australia
| | - Intissar Bittar
- Department of Pathology, Austin Health, Melbourne, VIC, Australia
| | - Glenn M. Eastwood
- Department of Intensive Care, Austin Health, Melbourne, VIC, Australia
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Health, Melbourne, VIC, Australia
- Centre for Integrated Critical Care, University of Melbourne, Melbourne, VIC, Australia
| | | |
Collapse
|
18
|
Wang Z, Wu J, Hu Z, Luo C, Wang P, Zhang Y, Li H. Dexmedetomidine Alleviates Lipopolysaccharide-Induced Acute Kidney Injury by Inhibiting p75NTR-Mediated Oxidative Stress and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5454210. [PMID: 33194004 PMCID: PMC7648709 DOI: 10.1155/2020/5454210] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/22/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023]
Abstract
Oxidative stress and apoptosis play a key role in the pathogenesis of sepsis-associated acute kidney injury (AKI). Dexmedetomidine (DEX) may present renal protective effects in sepsis. Therefore, we studied antioxidant effects and the mechanism of DEX in an inflammatory proximal tubular epithelial cell model and lipopolysaccharide- (LPS-) induced AKI in mice. Methods. We assessed renal function (creatinine, urea nitrogen), histopathology, oxidative stress (malondialdehyde (MDA) and superoxide dismutase (SOD)), and apoptosis (TUNEL staining and Cleaved caspase-3) in mice. In vitro experiments including Cleaved caspase-3 and p75NTR/p38MAPK/JNK signaling pathways were evaluated using western blot. Reactive oxidative species (ROS) production and apoptosis were determined using flow cytometry. Results. DEX significantly improved renal function and kidney injury and also revert the substantially increased level of MDA concentrations as well as the reduction of the SOD enzyme activity found in LPS-induced AKI mice. In parallel, DEX treatment also reduced the apoptosis and Cleaved caspase-3 expression evoked by LPS. The expression of p75NTR was increased in kidney tissues of mice with AKI but decreased after treatment with DEX. In cultured human renal tubular epithelial cell line (HK-2 cells), DEX inhibited LPS-induced apoptosis and generation of ROS, but this was reversed by overexpression of p75NTR. Furthermore, pretreatment with DEX significantly downregulated phosphorylation of JNK and p38MAPK in LPS-stimulated HK-2 cells, and this effect was abolished by overexpression of p75NTR. Conclusion. DEX ameliorated AKI in mice with sepsis by partially reducing oxidative stress and apoptosis through regulation of p75NTR/p38MAPK/JNK signaling pathways.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiali Wu
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaolan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cong Luo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanling Zhang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Plasma Renin Concentration is Associated With Hemodynamic Deficiency and Adverse Renal Outcome in Septic Shock. Shock 2020; 52:e22-e30. [PMID: 30407370 DOI: 10.1097/shk.0000000000001285] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND In septic shock, both systemic vasodilatation and glomerular arteriole dilatation are responsible for the drop in glomerular filtration observed in early acute kidney injury. Angiotensin II has been shown to act on both mechanisms. Our objective was to evaluate the impact of renin angiotensin system activation, on hemodynamic deficiency and renal outcome in patient with septic shock and to assess whether urinary sodium could be a reliable test for high plasma renin concentration screening. METHODS This was a prospective and observational study. Inclusion criteria were early septic shock (first episode), dose of norepinephrine ≥ 0.25 μg/kg/min, before the start of substitutive corticosteroids. Plasma renin concentration, plasma aldosterone concentration, and urinary sodium were measured at inclusion. Renal outcome, organ deficiency, and 28-day survival were followed. RESULTS Plasma renin concentration was associated with worse hemodynamic deficiency and adverse renal outcome. Natriuresis was associated with shock severity but was not associated with renal outcome. Low natriuresis (< 20 mM) was associated with higher renin concentration. Those two variables were only weakly correlated. CONCLUSION Plasma renin concentration is associated with adverse renal outcome, probably through shock severity and insufficient glomerular efferent arterioles vasoconstriction. An association was observed between low natriuresis and high plasma renin concentration.
Collapse
|
20
|
Busse LW, Ostermann M. Vasopressor Therapy and Blood Pressure Management in the Setting of Acute Kidney Injury. Semin Nephrol 2020; 39:462-472. [PMID: 31514910 DOI: 10.1016/j.semnephrol.2019.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Acute kidney injury (AKI) is common in the setting of shock. Hemodynamic instability is a risk factor for the development of AKI, and pathophysiological mechanisms include loss of renal perfusion pressure and impaired microcirculation. Although restoration of mean arterial pressure (MAP) may mitigate the risk of AKI to some extent, evidence on this is conflicting. Also debatable is the optimal blood pressure needed to minimize the risk of kidney injury. A MAP of 65 mm Hg traditionally has been considered adequate to maintain renal perfusion pressure, and studies have failed to consistently show improved outcomes at higher levels of MAP. Therapeutic options to support renal perfusion consist of catecholamines, vasopressin, and angiotensin II. Although catecholamines are the most studied, they are associated with adverse events at higher doses, including AKI. Vasopressin and angiotensin II are noncatecholamine options to support blood pressure and may improve microcirculatory hemodynamics through unique mechanisms, including differential vasoconstriction of efferent and afferent arterioles within the nephron. Future areas of study include methods by which clinicians can measure renal blood flow in a macrocirculatory and microcirculatory way, a personalized approach to blood pressure management in septic shock using patient-specific measures of perfusion adequacy, and novel agents that may improve the microcirculation within the kidneys without causing adverse microcirculatory effects in other organs.
Collapse
Affiliation(s)
- Laurence W Busse
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA.
| | - Marlies Ostermann
- Department of Critical Care, King's College London, Guy's and St Thomas' National Health Service Foundation Hospital, London, United Kingdom
| |
Collapse
|
21
|
Grisk O. The sympathetic nervous system in acute kidney injury. Acta Physiol (Oxf) 2020; 228:e13404. [PMID: 31610091 DOI: 10.1111/apha.13404] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/23/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is frequently accompanied by activation of the sympathetic nervous system (SNS). This may result from pre-exisiting chronic diseases associated with sympathetic activation prior to AKI or it may be induced by stressors that ultimately lead to AKI such as endotoxins and arterial hypotension in circulatory shock. Conversely, sympathetic activation may also result from acute renal injury. Focusing on studies in experimental renal ischaemia and reperfusion (IR), this review summarizes the current knowledge on how the SNS is activated in IR-induced AKI and on the consequences of sympathetic activation for the development of acute renal damage. Experimental studies show beneficial effects of sympathoinhibitory interventions on renal structure and function in response to IR. However, few clinical trials obtained in scenarios that correspond to experimental IR, namely major elective surgery, showed that peri-operative treatment with centrally acting sympatholytics reduced the incidence of AKI. Apparently, discrepant findings on how sympathetic activation influences renal responses to acute IR-induced injury are discussed and future areas of research in this field are identified.
Collapse
Affiliation(s)
- Olaf Grisk
- Institute of Physiology University of Greifswald Greifswald Germany
| |
Collapse
|
22
|
Yue J, Si Y, Zhu T, Yang J, Xu X, Fang Y, Fu W. MicroRNA-187 Reduces Acute Ischemic Renal Podocyte Injury via Targeting Acetylcholinesterase. J Surg Res 2019; 244:302-311. [PMID: 31302329 DOI: 10.1016/j.jss.2019.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/22/2019] [Accepted: 06/04/2019] [Indexed: 12/26/2022]
|
23
|
Barbar SD, Gaudry S, Dreyfuss D, Quenot JP. Renal replacement therapy: Time to give up on early initiation? Yes. Anaesth Crit Care Pain Med 2019; 37:501-503. [PMID: 30573205 DOI: 10.1016/j.accpm.2018.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Saber Davide Barbar
- Department of intensive care medicine, Nîmes University Hospital, place du prof. Robert-Debré, 30029 Nîmes, France.
| | - Stephane Gaudry
- Department of surgical and medical intensive care, Avicenne Hospital, AP-HP, 125, rue de Stalingrad, 93000 Bobigny, France; Remodelling and Repair of Renal Tissue, Tenon Hospital, French national institute of health and medical research (Inserm), UMR_S1155, 75020 Paris, France
| | - Didier Dreyfuss
- Department of surgical and medical intensive care, Avicenne Hospital, AP-HP, 125, rue de Stalingrad, 93000 Bobigny, France; Paris Diderot University, Sorbonne Paris Cité, IAME, UMRS 1137, 75018 Paris, France; Inserm, IAME, U1137, 75018 Paris, France
| | - Jean-Pierre Quenot
- Department of intensive care medicine, Dijon Bourgogne university hospital, 14, rue Paul-Gaffarel, 21000 Dijon, France; Bourgogne Franche-Comté university, Lipness Team UMR 1231 et LabExLipSTIC, 21000 Dijon, France; Inserm CIC 1432 Clinical Epidemiology and clinical trial, Dijon Bourgogne university hospital, 21000 Dijon, France
| |
Collapse
|
24
|
Abstract
Neuroimmune interaction is an emerging concept, wherein the nervous system modulates the immune system and vice versa. This concept is gaining attention as a novel therapeutic target in various inflammatory diseases including acute kidney injury (AKI). Vagus nerve stimulation or treatment with pulsed ultrasound activates the cholinergic anti-inflammatory pathway to prevent AKI in mice. The kidneys are innervated by sympathetic efferent and sensory afferent neurons, and these neurons also may play a role in the modulation of inflammation in AKI. In this review, we discuss several neural circuits with respect to the control of renal inflammation and AKI as well as optogenetics as a novel tool for understanding these complex neural circuits.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA.
| |
Collapse
|
25
|
Maciel AT. Urine electrolyte measurement as a "window" into renal microcirculatory stress assessment in critically ill patients. J Crit Care 2018; 48:90-96. [PMID: 30176529 DOI: 10.1016/j.jcrc.2018.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/07/2018] [Accepted: 08/14/2018] [Indexed: 12/20/2022]
Abstract
Urine electrolyte assessment has long been used in order to understand electrolyte concentration disturbances in blood and as an easy tool for monitoring renal perfusion and structural tubular damage. In the last few years, great improvement in the pathophysiology of acute kidney injury (AKI) has occurred, and the correlation between urine biochemistry (UB) behavior and renal perfusion was frequently questioned. Many authors have suggested abandoning UB monitoring due to its unclear role in AKI monitoring. Our group has been working in this field in the critically ill population, and we believe that, although UB is indeed very useful, a different point of view regarding the interpretation of the data should be used. The aim of this review is to explain the rationale of these new concepts and make suggestions for their adequate use in daily ICU practice, especially in low-income countries where more sophisticated and expensive AKI biomarker assessments are not available.
Collapse
Affiliation(s)
- Alexandre T Maciel
- Imed Research Group, Adult Intensive Care Unit, São Camilo Hospital, São Paulo, Brazil.
| |
Collapse
|
26
|
Berthelsen RE, Perner A, Jensen AK, Jensen JU, Bestle MH. Fluid accumulation during acute kidney injury in the intensive care unit. Acta Anaesthesiol Scand 2018; 62:780-790. [PMID: 29512107 DOI: 10.1111/aas.13105] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 01/31/2018] [Accepted: 02/07/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Fluid therapy is a ubiquitous intervention in patients admitted to the intensive care unit, but positive fluid balance may be associated with poor outcomes and particular in patients with acute kidney injury. Studies describing this have defined fluid overload either at specific time points or considered patients with a positive mean daily fluid balance as fluid overloaded. We wished to detail this further and performed joint model analyses of the association between daily fluid balance and outcome represented by mortality and renal recovery in patients admitted with acute kidney injury. METHOD We did a retrospective cohort study of patients admitted to the intensive care unit with acute kidney injury during a 2-year observation period. We used serum creatinine measurements to identify patients with acute kidney injury and collected sequential daily fluid balance during the first 5 days of admission to the intensive care unit. We used joint modelling techniques to correlate the development of fluid overload with survival and renal recovery adjusted for age, gender and disease severity. RESULTS The cohort contained 863 patients with acute kidney injury of whom 460 (53%) and 254 (29%) developed 5% and 10% fluid overload, respectively. We found that both 5% and 10% fluid overload was correlated with reduced survival and renal recovery. CONCLUSION Joint model analyses of fluid accumulation in patients admitted to the intensive care unit with acute kidney injury confirm that even a modest degree of fluid overload (5%) may be negatively associated with both survival and renal recovery.
Collapse
Affiliation(s)
- R. E. Berthelsen
- Department of Anaesthesiology and Intensive Care; Nordsjaellands Hospital; Hilleroed Denmark
| | - A. Perner
- Department of Intensive Care 4131; Rigshospitalet; Copenhagen Denmark
| | - A. K. Jensen
- Department of Research; Nordsjaellands Hospital; Hilleroed Denmark
- Department of Public Health, Section of Biostatistics; Copenhagen University; Copenhagen Denmark
| | - J.-U. Jensen
- CHIP & PERSIMUNE; Rigshospitalet; Copenhagen Denmark
| | - M. H. Bestle
- Department of Anaesthesiology and Intensive Care; Nordsjaellands Hospital; Hilleroed Denmark
| |
Collapse
|
27
|
Evans RG, Lankadeva YR, Cochrane AD, Marino B, Iguchi N, Zhu MZL, Hood SG, Smith JA, Bellomo R, Gardiner BS, Lee C, Smith DW, May CN. Renal haemodynamics and oxygenation during and after cardiac surgery and cardiopulmonary bypass. Acta Physiol (Oxf) 2018; 222. [PMID: 29127739 DOI: 10.1111/apha.12995] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
Acute kidney injury (AKI) is a common complication following cardiac surgery performed on cardiopulmonary bypass (CPB) and has important implications for prognosis. The aetiology of cardiac surgery-associated AKI is complex, but renal hypoxia, particularly in the medulla, is thought to play at least some role. There is strong evidence from studies in experimental animals, clinical observations and computational models that medullary ischaemia and hypoxia occur during CPB. There are no validated methods to monitor or improve renal oxygenation during CPB, and thus possibly decrease the risk of AKI. Attempts to reduce the incidence of AKI by early transfusion to ameliorate intra-operative anaemia, refinement of protocols for cooling and rewarming on bypass, optimization of pump flow and arterial pressure, or the use of pulsatile flow, have not been successful to date. This may in part reflect the complexity of renal oxygenation, which may limit the effectiveness of individual interventions. We propose a multi-disciplinary pathway for translation comprising three components. Firstly, large-animal models of CPB to continuously monitor both whole kidney and regional kidney perfusion and oxygenation. Secondly, computational models to obtain information that can be used to interpret the data and develop rational interventions. Thirdly, clinically feasible non-invasive methods to continuously monitor renal oxygenation in the operating theatre and to identify patients at risk of AKI. In this review, we outline the recent progress on each of these fronts.
Collapse
Affiliation(s)
- R. G. Evans
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - Y. R. Lankadeva
- Florey Institute of Neuroscience and Mental Health University of Melbourne Melbourne Vic. Australia
| | - A. D. Cochrane
- Department of Cardiothoracic Surgery Monash Health Monash University Melbourne Vic. Australia
- Department of Surgery School of Clinical Sciences at Monash Health Monash University Melbourne Vic. Australia
| | - B. Marino
- Department of Perfusion Services Austin Hospital Heidelberg Vic. Australia
| | - N. Iguchi
- Florey Institute of Neuroscience and Mental Health University of Melbourne Melbourne Vic. Australia
| | - M. Z. L. Zhu
- Department of Cardiothoracic Surgery Monash Health Monash University Melbourne Vic. Australia
- Department of Surgery School of Clinical Sciences at Monash Health Monash University Melbourne Vic. Australia
| | - S. G. Hood
- Florey Institute of Neuroscience and Mental Health University of Melbourne Melbourne Vic. Australia
| | - J. A. Smith
- Department of Cardiothoracic Surgery Monash Health Monash University Melbourne Vic. Australia
- Department of Surgery School of Clinical Sciences at Monash Health Monash University Melbourne Vic. Australia
| | - R. Bellomo
- Department of Intensive Care Austin Hospital Heidelberg Vic. Australia
| | - B. S. Gardiner
- School of Engineering and Information Technology Murdoch University Perth WA Australia
- Faculty of Engineering and Mathematical Sciences The University of Western Australia Perth WA Australia
| | - C.‐J. Lee
- School of Engineering and Information Technology Murdoch University Perth WA Australia
- Faculty of Engineering and Mathematical Sciences The University of Western Australia Perth WA Australia
| | - D. W. Smith
- Faculty of Engineering and Mathematical Sciences The University of Western Australia Perth WA Australia
| | - C. N. May
- Florey Institute of Neuroscience and Mental Health University of Melbourne Melbourne Vic. Australia
| |
Collapse
|
28
|
Determinants of Urinary Output Response to IV Furosemide in Acute Kidney Injury: A Pharmacokinetic/Pharmacodynamic Study. Crit Care Med 2017; 44:e923-9. [PMID: 27183025 DOI: 10.1097/ccm.0000000000001823] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVES This study assessed the determinants of urinary output response to furosemide in acute kidney injury; specifically, whether the response is related to altered pharmacokinetics or pharmacodynamics. DESIGN Prospective cohort. SETTING Tertiary ICU. PATIENTS Thirty critically ill patients with acute kidney injury without preexisting renal impairment or recent diuretic exposure. INTERVENTION A single dose of IV furosemide. MEASUREMENTS AND MAIN RESULTS Baseline markers of intravascular volume status were obtained prior to administering furosemide. Six-hour creatinine clearance, hourly plasma/urinary furosemide concentrations, and hourly urinary output were used to assess furosemide pharmacokinetics/pharmacodynamics parameters. Of 30 patients enrolled, 11 had stage-1 (37%), nine had stage-2 (30%), and 10 had stage-3 (33%) Acute Kidney Injury Network acute kidney injury. Seventy-three percent were septic, 47% required norepinephrine, and 53% were mechanically ventilated. Urinary output doubled in 20 patients (67%) following IV furosemide. Measured creatinine clearance was strongly associated with the amount of urinary furosemide excreted and was the only reliable predictor of the urinary output after furosemide (area under the receiver-operating-characteristic curve, 0.75; 95% CI, 0.57-0.93). In addition to an altered pharmacokinetics (p < 0.01), a reduced pharmacodynamics response to furosemide also became important when creatinine clearance was reduced to less than 40 mL/min/1.73 m (p = 0.01). Acute kidney injury staging and markers of intravascular volume, including central venous pressure, brain-natriuretic-peptide concentration, and fractional urinary sodium excretion were not predictive of urinary output response to furosemide. CONCLUSIONS The severity of acute kidney injury, as reflected by the measured creatinine clearance, alters both pharmacokinetics and pharmacodynamics of furosemide in acute kidney injury, and was the only reliable predictor of the urinary output response to furosemide in acute kidney injury.
Collapse
|
29
|
|
30
|
The exciting “bench to bedside” journey of cell therapies for acute kidney injury and renal transplantation. J Nephrol 2017; 30:319-336. [DOI: 10.1007/s40620-017-0384-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/20/2017] [Indexed: 12/15/2022]
|
31
|
Hering D, Winklewski PJ. R1 autonomic nervous system in acute kidney injury. Clin Exp Pharmacol Physiol 2017; 44:162-171. [DOI: 10.1111/1440-1681.12694] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 10/17/2016] [Accepted: 10/29/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Dagmara Hering
- Dobney Hypertension Centre; School of Medicine and Pharmacology; Royal Perth Hospital Unit; The University of Western Australia; Perth WA Australia
| | - Pawel J Winklewski
- Institute of Human Physiology; Medical University of Gdansk; Gdansk Poland
- Department of Clinical Sciences; Institute of Health Sciences; Pomeranian University of Slupsk; Slupsk Poland
| |
Collapse
|
32
|
Urine biochemistry assessment in critically ill patients: controversies and future perspectives. J Clin Monit Comput 2016; 31:539-546. [PMID: 27038161 DOI: 10.1007/s10877-016-9871-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/30/2016] [Indexed: 12/17/2022]
Abstract
In the past, urine biochemistry was a major tool in acute kidney injury (AKI) management. Classic papers published some decades ago established the values of the urine indices which were thought to distinguish "pre-renal" (functional) AKI attributed to low renal perfusion and "renal" (structural) AKI attributed to acute tubular necrosis (ATN). However, there were a lot of drawbacks and limitations in these studies and some recent articles have questioned the utility of measuring urine electrolytes especially because they do not seem to adequately inform about renal perfusion nor AKI duration (transient vs. persistent). At the same time, the "pre-renal" paradigm has been consistently criticized because hypoperfusion followed by ischemia and ATN does not seem to explain most of the AKI developing in critically ill patients and distinct AKI durations do not seem to be clearly related to different pathophysiological mechanisms or histopathological findings. In this new context, other possible roles for urine biochemistry have emerged. Some studies have suggested standardized changes in the urine electrolyte composition preceding increases in serum creatinine independently of AKI subsequent duration, which might actually be due to intra-renal microcirculatory changes and activation of sodium-retaining mechanisms even in the absence of impaired global renal blood flow. In the present review, the points of controversy regarding urine biochemistry assessment were evaluated as well as future perspectives for its role in AKI monitoring. An alternative approach for the interpretation of measured urine electrolytes is proposed which needs further larger studies to be validated and incorporated in daily ICU practice.
Collapse
|
33
|
Abstract
All components of the endothelin (ET) system are present in renal tubular cells. In this review, we summarize current knowledge about ET and the most common tubular diseases: acute kidney injury (AKI) and polycystic kidney disease. AKI originally was called acute tubular necrosis, pointing to the most prominent morphologic findings. Similarly, cysts in polycystic kidney disease, and especially in autosomal-dominant polycystic kidney disease, are of tubular origin. Preclinical studies have indicated that the ET system and particularly ETA receptors are involved in the pathogenesis of ischemia-reperfusion injury, although these findings have not been translated to clinical studies. The ET system also has been implicated in radiocontrast-dye-induced AKI, however, ET-receptor blockade in a large human study was not successful. The ET system is activated in sepsis models of AKI; the effectiveness of ET blocking agents in preclinical studies is variable depending on the model and the ET-receptor antagonist used. Numerous studies have shown that the ET system plays an important role in the complex pathophysiology associated with cyst formation and disease progression in polycystic kidney disease. However, results from selective targeting of ET-receptor subtypes in animal models of polycystic kidney disease have proved disappointing and do not support clinical trials. These studies have shown that a critical balance between ETA and ETB receptor action is necessary to maintain structure and function in the cystic kidney. In summary, ETs have been implicated in the pathogenesis of several renal tubulointerstitial diseases, however, experimental animal findings have not yet led to use of ET blockers in human beings.
Collapse
Affiliation(s)
- Albert C M Ong
- Kidney Genetics Group, Academic Nephrology Unit, Department of Infection and Immunity, University of Sheffield Medical School, Sheffield, UK
| | | | - Berthold Hocher
- Institute for Nutritional Science, University of Potsdam, Potsdam, Germany.
| |
Collapse
|
34
|
Maciel AT, Nassar AP, Vitorio D. Very Transient Cases of Acute Kidney Injury in the Early Postoperative Period After Cardiac Surgery: The Relevance of More Frequent Serum Creatinine Assessment and Concomitant Urinary Biochemistry Evaluation. J Cardiothorac Vasc Anesth 2016; 30:56-63. [DOI: 10.1053/j.jvca.2015.04.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Indexed: 11/11/2022]
|
35
|
Farías JG, Herrera EA, Carrasco-Pozo C, Sotomayor-Zárate R, Cruz G, Morales P, Castillo RL. Pharmacological models and approaches for pathophysiological conditions associated with hypoxia and oxidative stress. Pharmacol Ther 2015; 158:1-23. [PMID: 26617218 DOI: 10.1016/j.pharmthera.2015.11.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypoxia is the failure of oxygenation at the tissue level, where the reduced oxygen delivered is not enough to satisfy tissue demands. Metabolic depression is the physiological adaptation associated with reduced oxygen consumption, which evidently does not cause any harm to organs that are exposed to acute and short hypoxic insults. Oxidative stress (OS) refers to the imbalance between the generation of reactive oxygen species (ROS) and the ability of endogenous antioxidant systems to scavenge ROS, where ROS overwhelms the antioxidant capacity. Oxidative stress plays a crucial role in the pathogenesis of diseases related to hypoxia during intrauterine development and postnatal life. Thus, excessive ROS are implicated in the irreversible damage to cell membranes, DNA, and other cellular structures by oxidizing lipids, proteins, and nucleic acids. Here, we describe several pathophysiological conditions and in vivo and ex vivo models developed for the study of hypoxic and oxidative stress injury. We reviewed existing literature on the responses to hypoxia and oxidative stress of the cardiovascular, renal, reproductive, and central nervous systems, and discussed paradigms of chronic and intermittent hypobaric hypoxia. This systematic review is a critical analysis of the advantages in the application of some experimental strategies and their contributions leading to novel pharmacological therapies.
Collapse
Affiliation(s)
- Jorge G Farías
- Facultad de Ingeniería y Ciencias, Departamento de Ingeniería Química, Universidad de la Frontera, Casilla 54-D, Temuco, Chile
| | - Emilio A Herrera
- Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Chile
| | | | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile
| | - Paola Morales
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Chile
| | - Rodrigo L Castillo
- Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Chile.
| |
Collapse
|
36
|
Abstract
Acute kidney injury (AKI) is a serious yet potentially reversible complication of sepsis. Several molecular mechanisms involved in the development of septic AKI have been identified. These mechanisms may be important targets in the development of future therapies. This review highlights the role of the innate immune response to sepsis and its downstream effects on kidney structure and function with special reference to the adaptive cellular response and glomerular hemodynamic changes. In addition, current evidence surrounding the management of patients with septic AKI is summarized. Finally, potential novel therapies for septic AKI are presented.
Collapse
Affiliation(s)
- Johan Mårtensson
- Department of Intensive Care, Austin Hospital, 145 Studley Road, Heidelberg, Victoria 3084, Australia; Department of Physiology and Pharmacology, Section of Anaesthesia and Intensive Care Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, 145 Studley Road, Heidelberg, Victoria 3084, Australia; Department of Epidemiology and Preventive Medicine, Australian and New Zealand Intensive Care Research Centre, School of Preventive Medicine and Public Health, Monash University, 99 Commercial Road, Melbourne, Victoria 3004, Australia.
| |
Collapse
|
37
|
Acute Kidney Injury Induced by Systemic Inflammatory Response Syndrome is an Avid and Persistent Sodium-Retaining State. Case Rep Crit Care 2014; 2014:471658. [PMID: 25309760 PMCID: PMC4189521 DOI: 10.1155/2014/471658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/27/2014] [Accepted: 08/29/2014] [Indexed: 12/27/2022] Open
Abstract
Acute kidney injury (AKI) is a frequent complication of the systemic inflammatory response syndrome (SIRS), which is triggered by many conditions in the intensive care unit, including different types of circulatory shock. One under-recognized characteristic of the SIRS-induced AKI is its avidity for sodium retention, with progressive decreases in urinary sodium concentration (NaU) and its fractional excretion (FENa). This phenomenon occurs in parallel with increases in serum creatinine, being only transitorily mitigated by diuretic use. In the present case, we report a situation of two consecutive shocks: the first shock is hemorrhagic in origin and then the second shock is a septic one in the same patient. The SIRS and AKI triggered by the first shock were not completely solved when the second shock occurred. This could be viewed as a persistent avid sodium-retaining state, which may be appreciated even during renal replacement therapy (in the absence of complete anuria) and that usually solves only after complete AKI and SIRS resolution. We suggest that decreases in NaU and FENa are major characteristics of SIRS-induced AKI, irrespective of the primary cause, and may serve as additional monitoring tools in its development and resolution.
Collapse
|