1
|
Yang X, Zhu L, Pan H, Yang Y. Cardiopulmonary bypass associated acute kidney injury: better understanding and better prevention. Ren Fail 2024; 46:2331062. [PMID: 38515271 PMCID: PMC10962309 DOI: 10.1080/0886022x.2024.2331062] [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: 10/17/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
Cardiopulmonary bypass (CPB) is a common technique in cardiac surgery but is associated with acute kidney injury (AKI), which carries considerable morbidity and mortality. In this review, we explore the range and definition of CPB-associated AKI and discuss the possible impact of different disease recognition methods on research outcomes. Furthermore, we introduce the specialized equipment and procedural intricacies associated with CPB surgeries. Based on recent research, we discuss the potential pathogenesis of AKI that may result from CPB, including compromised perfusion and oxygenation, inflammatory activation, oxidative stress, coagulopathy, hemolysis, and endothelial damage. Finally, we explore current interventions aimed at preventing and attenuating renal impairment related to CPB, and presenting these measures from three perspectives: (1) avoiding CPB to eliminate the fundamental impact on renal function; (2) optimizing CPB by adjusting equipment parameters, optimizing surgical procedures, or using improved materials to mitigate kidney damage; (3) employing pharmacological or interventional measures targeting pathogenic factors.
Collapse
Affiliation(s)
- Xutao Yang
- The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Li Zhu
- The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- The Jinhua Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Hong Pan
- The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yi Yang
- The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
2
|
Fogagnolo A, Azzolina D, Taccone FS, Pedarzani E, Pasa G, Marianello D, Valpiani G, Marchesini C, Annoni F, Moureau A, Volta CA, Franchi F, Spadaro S. Oxygen extraction-guided transfusion strategy in critically ill patients: study protocol for a randomised, open-labelled, controlled trial. BMJ Open 2024; 14:e089910. [PMID: 39532348 PMCID: PMC11575245 DOI: 10.1136/bmjopen-2024-089910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION In critically ill patients, individualised strategies for red blood cell transfusion (RBCT) are lacking. The objective of this study is to demonstrate the potential advantages of employing an individualised transfusion strategy compared with a restrictive approach, in unselected intensive care unit (ICU) patients. METHODS This will be a randomised, multicentre, international trial. Two open-label parallel groups will be compared with an allocation ratio of 1:1. The trial is designed to investigate the superiority of the individualised intervention group compared with the standard intervention group. The study will be performed in three mixed, academic ICUs located in two different countries. In the individualised group, prescription of RCBT is restricted to patients who present haemoglobin (Hb) ≤9.0 g/dL and oxygen extraction ratio (O2ER) ≥ 30%, for a minimum Hb value of ≤6.0 g/dL. In the control group, prescription of RBCT is guided by thresholds proposed by recent guidelines, regardless of O2ER values. ETHICS AND DISSEMINATION This trial is approved by the Comitato Etico Area Vasta Centro della Regione Emilia-Romagna (protocol number 350/2023/Sper/AOUFe/PRBCT, date of approval 18/05/2023) and ethic boards at all participating sites. Our results will be published and shared with relevant organisations and healthcare professionals. TRIAL REGISTRATION NUMBER Clinicaltrials.gov NCT06102590.
Collapse
Affiliation(s)
| | - Danila Azzolina
- Dipartimento di Scienze Ambientali e Preventive, University of Ferrara, Ferrara, Emilia-Romagna, Italy
| | - Fabio Silvio Taccone
- Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Emma Pedarzani
- Dipartimento di Scienze Ambientali e Preventive, University of Ferrara, Ferrara, Emilia-Romagna, Italy
- Dipartimento di Scienze Cardiache, Toraciche, Vascolari e Sanità Pubblica, Unità di Biostatistica, Epidemiologia e Sanità Pubblica Salute, University of Padua, Padova, Veneto, Italy
| | - Gianluca Pasa
- Department of Medical Science, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Daniele Marianello
- Department of Medical Science, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Giorgia Valpiani
- Dipartimento di Scienze Ambientali e Preventive, University of Ferrara, Ferrara, Emilia-Romagna, Italy
| | - Chiara Marchesini
- Department of translational medicine, University of Ferrrara, Ferrara, Italy
| | - Filippo Annoni
- Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Anthony Moureau
- Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Federico Franchi
- Department of Medical Science, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Savino Spadaro
- Department of translational medicine, University of Ferrrara, Ferrara, Italy
- Emergency Department, University of Ferrara, Ferrara, Italy
| |
Collapse
|
3
|
Hillaert A, Sanmiguel Serpa LC, Bogaert S, Broeckx BJG, Hesta M, Vandermeulen E, Germonpré J, Stock E, Pullens P, Vanderperren K. Assessment of pharmacologically induced changes in canine kidney function by multiparametric magnetic resonance imaging and contrast enhanced ultrasound. Front Vet Sci 2024; 11:1406343. [PMID: 38966564 PMCID: PMC11223176 DOI: 10.3389/fvets.2024.1406343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024] Open
Abstract
IntroductionDynamic contrast-enhanced (DCE) MRI and arterial spin labeling (ASL) MRI enable non-invasive measurement of renal blood flow (RBF), whereas blood oxygenation level-dependent (BOLD) MRI enables non-invasive measurement of the apparent relaxation rate (R2*), an indicator of oxygenation. This study was conducted to evaluate the potential role of these MRI modalities in assessing RBF and oxygenation in dogs. The correlation between contrast-enhanced ultrasound (CEUS) and the MRI modalities was examined and also the ability of the MRI modalities to detect pharmacologically induced changes.MethodsRBF, using CEUS, ASL- and DCE-MRI, as well as renal oxygenation, using BOLD-MRI of eight adult beagles were assessed at two time-points, 2–3 weeks apart. During each time point, the anesthetized dogs received either a control (0.9% sodium chloride) or a dopamine treatment. For each time point, measurements were carried out over 2 days. An MRI scan at 3 T was performed on day one, followed by CEUS on day two.ResultsUsing the model-free model with caudal placement of the arterial input function (AIF) region of interest (ROI) in the aorta, the DCE results showed a significant correlation with ASL measured RBF and detected significant changes in blood flow during dopamine infusion. Additionally, R2* negatively correlated with ASL measured RBF at the cortex and medulla, as well as with medullary wash-in rate (WiR) and peak intensity (PI). ASL measured RBF, in its turn, showed a positive correlation with cortical WiR, PI, area under the curve (AUC) and fall time (FT), and with medullary WiR and PI, but a negative correlation with medullary rise time (RT). During dopamine infusion, BOLD-MRI observed a significant decrease in R2* at the medulla and entire kidney, while ASL-MRI demonstrated a significant increase in RBF at the cortex, medulla and the entire kidney.ConclusionASL- and BOLD-MRI can measure pharmacologically induced changes in renal blood flow and renal oxygenation in dogs and might allow detection of changes that cannot be observed with CEUS. However, further research is needed to confirm the potential of ASL- and BOLD-MRI in dogs and to clarify which analysis method is most suitable for DCE-MRI in dogs.
Collapse
Affiliation(s)
- Amber Hillaert
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Luis Carlos Sanmiguel Serpa
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Stephanie Bogaert
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, Ghent, Belgium
| | - Bart J. G. Broeckx
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Myriam Hesta
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eva Vandermeulen
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jolien Germonpré
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Emmelie Stock
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Pim Pullens
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, Ghent, Belgium
- Institute of Biomedical Engineering and Technology, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Katrien Vanderperren
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
4
|
Milne B, Gilbey T, De Somer F, Kunst G. Adverse renal effects associated with cardiopulmonary bypass. Perfusion 2024; 39:452-468. [PMID: 36794518 PMCID: PMC10943608 DOI: 10.1177/02676591231157055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Cardiac surgery on cardiopulmonary bypass (CPB) is associated with postoperative renal dysfunction, one of the most common complications of this surgical cohort. Acute kidney injury (AKI) is associated with increased short-term morbidity and mortality and has been the focus of much research. There is increasing recognition of the role of AKI as the key pathophysiological state leading to the disease entities acute and chronic kidney disease (AKD and CKD). In this narrative review, we will consider the epidemiology of renal dysfunction after cardiac surgery on CPB and the clinical manifestations across the spectrum of disease. We will discuss the transition between different states of injury and dysfunction, and, importantly, the relevance to clinicians. The specific facets of kidney injury on extracorporeal circulation will be described and the current evidence evaluated for the use of perfusion-based techniques to reduce the incidence and mitigate the complications of renal dysfunction after cardiac surgery.
Collapse
Affiliation(s)
- Benjamin Milne
- Department of Anaesthesia & Pain Medicine, King’s College Hospital NHS Foundation Trust, London, UK
| | - Tom Gilbey
- Department of Anaesthesia & Pain Medicine, King’s College Hospital NHS Foundation Trust, London, UK
- Nuffield Department of Anaesthesia, John Radcliffe Hospital, Oxford, UK
| | - Filip De Somer
- Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University Hospital, Ghent, Belgium
| | - Gudrun Kunst
- Department of Anaesthesia & Pain Medicine, King’s College Hospital NHS Foundation Trust, London, UK
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London British Heart Foundation Centre of Excellence, London, UK
| |
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
|
Suieubekov B, Sepbayeva A, Yeshmanova A, Kusainov A. Cardiac surgery-associated acute kidney injury in newborns: A meta-analysis. ELECTRONIC JOURNAL OF GENERAL MEDICINE 2023. [DOI: 10.29333/ejgm/12805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
<b>Introduction</b>: Acute kidney injury is a common complication following pediatric heart surgery, and it has been linked to an increased risk of morbidity and fatality.<br />
<b>Methods</b>: The PubMed and Medline databases were combed for relevant research until May 2022. The terms [Cardiac surgery] AND [acute renal injury] AND [newborns OR children OR neonates] AND [randomized control studies OR randomized control trials] were used as search criteria. The studies that met the inclusion criteria were considered qualified using the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines.<br />
<b>Results</b>: A total of 2,941 newborns or children were enrolled in 14 studies, with 931 developing acute renal damage. 2,095 of the enrolled infants and children received steroid, aminophylline, dexmedetomidine, and acetaminophen therapies. In seven studies, the odds ratio for steroids was not significantly different from control. In contrast, two studies comparing aminophylline to a control group found no statistically significant change. Two studies found no significant difference in dexmedetomidine therapy compared to control. Three trials, however, found a significant difference between the acetaminophen treatment and control groups.<br />
<b>Conclusion</b>: Acetaminophen was linked to a decreased risk of postoperative acute renal injury, while steroids had no benefit and aminophylline treatment could be justified.
Collapse
Affiliation(s)
| | - Anar Sepbayeva
- Asfendiyarov Kazakh National Medical University, Almaty, KAZAKHSTAN
| | - Ainur Yeshmanova
- Asfendiyarov Kazakh National Medical University, Almaty, KAZAKHSTAN
| | | |
Collapse
|
7
|
Zhou Y, Feng J, Mei S, Zhong H, Tang R, Xing S, Gao Y, Xu Q, He Z. MACHINE LEARNING MODELS FOR PREDICTING ACUTE KIDNEY INJURY IN PATIENTS WITH SEPSIS-ASSOCIATED ACUTE RESPIRATORY DISTRESS SYNDROME. Shock 2023; 59:352-359. [PMID: 36625493 DOI: 10.1097/shk.0000000000002065] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
ABSTRACT Background: Acute kidney injury (AKI) is a prevalent and serious complication among patients with sepsis-associated acute respiratory distress syndrome (ARDS). Prompt and accurate prediction of AKI has an important role in timely intervention, ultimately improving the patients' survival rate. This study aimed to establish machine learning models to predict AKI via thorough analysis of data derived from electronic medical records. Method: The data of eligible patients were retrospectively collected from the Medical Information Mart for Intensive Care III database from 2001 to 2012. The primary outcome was the development of AKI within 48 hours after intensive care unit admission. Four different machine learning models were established based on logistic regression, support vector machine, random forest, and extreme gradient boosting (XGBoost). The performance of all predictive models was evaluated using the area under receiver operating characteristic curve, precision-recall curve, confusion matrix, and calibration plot. Moreover, the discrimination ability of the machine learning models was compared with that of the Sequential Organ Failure Assessment (SOFA) model. Results; Among 1,085 sepsis-associated ARDS patients included in this research, 375 patients (34.6%) developed AKI within 48 hours after intensive care unit admission. Twelve predictive variables were selected and further used to establish the machine learning models. The XGBoost model yielded the most accurate predictions with the highest area under receiver operating characteristic curve (0.86) and accuracy (0.81). In addition, a novel shiny application based on the XGBoost model was established to predict the probability of developing AKI among patients with sepsis-associated ARDS. Conclusions: Machine learning models could be used for predicting AKI in patients with sepsis-associated ARDS. Accordingly, a user-friendly shiny application based on the XGBoost model with reliable predictive performance was released online to predict the probability of developing AKI among patients with sepsis-associated ARDS.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Brignone J, Jensen M, Jensen BL, Assersen KB, Goetze JP, Jødal L, Andersen TB, Magnusdottir SO, Kloster B, Jønler M, Lund L. Protective effect of sacubitril/valsartan (Entresto) on kidney function and filtration barrier injury in a porcine model of partial nephrectomy. Nephrol Dial Transplant 2023; 38:80-92. [PMID: 35704678 DOI: 10.1093/ndt/gfac200] [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: 02/02/2022] [Indexed: 01/26/2023] Open
Abstract
Kidney surgery often includes organ ischaemia with a risk of acute kidney injury. The present study tested if treatment with the combined angiotensin II-angiotensin II receptor type 1 and neprilysin blocker Entresto (LCZ696, sacubitril/valsartan) protects filtration barrier and kidney function after ischaemia and partial nephrectomy (PN) in pigs. Single kidney glomerular filtration rate (GFR) by technetium-99m diethylene-triamine-pentaacetate clearance was validated (n = 6). Next, four groups of pigs were followed for 15 days (n = 24) after PN (one-third right kidney, 60 min ischaemia) + Entresto (49/51 mg/day; n = 8), PN + vehicle (n = 8), sham + Entresto (49/51 mg/day; n = 4) and sham + vehicle (n = 4). GFR, diuresis and urinary albumin were measured at baseline and from each kidney after 15 days. The sum of single-kidney GFR (right 25 ± 6 mL/min, left 31 ± 7 mL/min) accounted for the total GFR (56 ± 14 mL/min). Entresto had no effect on baseline blood pressure, p-creatinine, mid-regional pro-atrial natriuretic peptide (MR-proANP), heart rate and diuresis. After 15 days, Entresto increased GFR in the uninjured kidney (+23 ± 6 mL/min, P < .05) and reduced albuminuria from both kidneys. In the sham group, plasma MR-proANP was not altered by Entresto; it increased to similar levels 2 h after surgery with and without Entresto. Fractional sodium excretion increased with Entresto. Kidney histology and kidney injury molecule-1 in cortex tissue were not different. In conclusion, Entresto protects the filtration barrier and increases the functional adaptive response of the uninjured kidney.
Collapse
Affiliation(s)
- Juan Brignone
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Mia Jensen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.,Department of Urology, Odense University Hospital, Odense, Denmark
| | - Kasper Bostlund Assersen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry, University of Copenhagen, Copenhagen, Denmark
| | - Lars Jødal
- Department of Nuclear Medicine, Aalborg University Hospital, Aalborg, Denmark
| | | | | | - Brian Kloster
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark
| | - Morten Jønler
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark
| | - Lars Lund
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark.,Department of Urology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
9
|
Youth versus adult-onset type 2 diabetic kidney disease: Insights into currently known structural differences and the potential underlying mechanisms. Clin Sci (Lond) 2022; 136:1471-1483. [PMID: 36326718 PMCID: PMC10175439 DOI: 10.1042/cs20210627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 11/05/2022]
Abstract
Abstract
Type 2 diabetes (T2D) is a global health pandemic with significant humanitarian, economic, and societal implications, particularly for youth and young adults who are experiencing an exponential rise in incident disease. Youth-onset T2D has a more aggressive phenotype than adult-onset T2D, and this translates to important differences in rates of progression of diabetic kidney disease (DKD). We hypothesize that youth-onset DKD due to T2D may exhibit morphometric, metabolic, and molecular characteristics that are distinct from adult-onset T2D and develop secondary to inherent differences in renal energy expenditure and substrate metabolism, resulting in a central metabolic imbalance. Kidney structural changes that are evident at the onset of puberty also serve to exacerbate the organ’s baseline high rates of energy expenditure. Additionally, the physiologic state of insulin resistance seen during puberty increases the risk for kidney disease and is exacerbated by both concurrent diabetes and obesity. A metabolic mismatch in renal energetics may represent a novel target for pharmacologic intervention, both for prevention and treatment of DKD. Further investigation into the underlying molecular mechanisms resulting in DKD in youth-onset T2D using metabolomics and RNA sequencing of kidney tissue obtained at biopsy is necessary to expand our understanding of early DKD and potential targets for therapeutic intervention. Furthermore, large-scale clinical trials evaluating the duration of kidney protective effects of pharmacologic interventions that target a metabolic mismatch in kidney energy expenditure are needed to help mitigate the risk of DKD in youth-onset T2D.
Collapse
|
10
|
Liu X, Du H, Sun Y, Shao L. Role of abnormal energy metabolism in the progression of chronic kidney disease and drug intervention. Ren Fail 2022; 44:790-805. [PMID: 35535500 PMCID: PMC9103584 DOI: 10.1080/0886022x.2022.2072743] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) is a severe clinical syndrome with significant socioeconomic impact worldwide. Orderly energy metabolism is essential for normal kidney function and energy metabolism disorders are increasingly recognized as an important player in CKD. Energy metabolism disorders are characterized by ATP deficits and reactive oxygen species increase. Oxygen and mitochondria are essential for ATP production, hypoxia and mitochondrial dysfunction both affect the energy production process. Renin-angiotensin and adenine signaling pathway also play important regulatory roles in energy metabolism. In addition, disturbance of energy metabolism is a key factor in the development of hereditary nephropathy such as autosomal dominant polycystic kidney disease. Currently, drugs with clinically clear renal function protection, such as Angiotensin II Type 1 receptor blockers and fenofibrate, have been proven to improve energy metabolism disorders. The sodium-glucose co-transporter inhibitors 2 that can mediate glucose metabolism disorders not only delay the progress of diabetic nephropathy, but also have significant protective effects in non-diabetic nephropathy. Hypoxia-inducible factor enhances ATP production to the kidney by improving renal oxygen supply and increasing glycolysis, and the mitochondria targeted peptides (SS-31) plays a protective role by stabilizing the mitochondrial inner membrane. Moreover, several drugs are being studied and are predicted to have potential renal protective properties. We propose that the regulation of energy metabolism represents a promising strategy to delay the progression of CKD.
Collapse
Affiliation(s)
- Xuyan Liu
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Huasheng Du
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Yan Sun
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Leping Shao
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Davidson JA, Robison J, Khailova L, Frank BS, Jaggers J, Ing RJ, Lawson S, Iguidbashian J, Ali E, Treece A, Soranno DE, Osorio-Lujan S, Klawitter J. Metabolomic profiling demonstrates evidence for kidney and urine metabolic dysregulation in a piglet model of cardiac surgery-induced acute kidney injury. Am J Physiol Renal Physiol 2022; 323:F20-F32. [PMID: 35532069 PMCID: PMC9236877 DOI: 10.1152/ajprenal.00039.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Acute kidney injury (AKI) is a common cause of morbidity after congenital heart disease surgery. Progress on diagnosis and therapy remains limited, however, in part due to poor mechanistic understanding and a lack of relevant translational models. Metabolomic approaches could help identify novel mechanisms of injury and potential therapeutic targets. In the present study, we used a piglet model of cardiopulmonary bypass with deep hypothermic circulatory arrest (CPB/DHCA) and targeted metabolic profiling of kidney tissue, urine, and serum to evaluate metabolic changes specific to animals with histological acute kidney injury. CPB/DHCA animals with acute kidney injury were compared with those without acute kidney injury and mechanically ventilated controls. Acute kidney injury occurred in 10 of 20 CPB/DHCA animals 4 h after CPB/DHCA and 0 of 7 control animals. Injured kidneys showed a distinct tissue metabolic profile compared with uninjured kidneys (R2 = 0.93, Q2 = 0.53), with evidence of dysregulated tryptophan and purine metabolism. Nine urine metabolites differed significantly in animals with acute kidney injury with a pattern suggestive of increased aerobic glycolysis. Dysregulated metabolites in kidney tissue and urine did not overlap. CPB/DHCA strongly affected the serum metabolic profile, with only one metabolite that differed significantly with acute kidney injury (pyroglutamic acid, a marker of oxidative stress). In conclusion, based on these findings, kidney tryptophan and purine metabolism are candidates for further mechanistic and therapeutic investigation. Urine biomarkers of aerobic glycolysis could help diagnose early acute kidney injury after CPB/DHCA and warrant further evaluation. The serum metabolites measured at this early time point did not strongly differentiate based on acute kidney injury. NEW & NOTEWORTHY This project explored the metabolic underpinnings of postoperative acute kidney injury (AKI) following pediatric cardiac surgery in a translationally relevant large animal model of cardiopulmonary bypass with deep hypothermic circulatory arrest. Here, we present novel evidence for dysregulated tryptophan catabolism and purine catabolism in kidney tissue and increased urinary glycolysis intermediates in animals who developed histological AKI. These pathways represent potential diagnostic and therapeutic targets for postoperative AKI in this high-risk population.
Collapse
Affiliation(s)
- Jesse A Davidson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Justin Robison
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, United States
| | - Ludmila Khailova
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Benjamin S Frank
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - James Jaggers
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Richard J Ing
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Scott Lawson
- Heart Institute, Children's Hospital Colorado, Aurora, CO, United States
| | - John Iguidbashian
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Eiman Ali
- Heart Institute, Children's Hospital Colorado, Aurora, CO, United States
| | - Amy Treece
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Danielle E Soranno
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Suzanne Osorio-Lujan
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jelena Klawitter
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
12
|
Kitzerow O, Zucker IH, Lisco SJ, Wang HJ. Timeline of Multi-Organ Plasma Extravasation After Bleomycin-Induced Acute Lung Injury. Front Physiol 2022; 13:777072. [PMID: 35173628 PMCID: PMC8841715 DOI: 10.3389/fphys.2022.777072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/10/2022] [Indexed: 12/30/2022] Open
Abstract
Acute lung injury (ALI) is characterized by the abrupt onset of clinically significant hypoxemia in the context of non-hydrostatic pulmonary edema. Acute lung injury is associated with cytokine release and plasma extravasation (PEx) that can cause pulmonary edema and subsequently acute respiratory distress syndrome (ARDS). Therefore, it is critical we understand the relationship between ALI and lung PEx. In addition, it is also important to assess PEx in the lungs and other organs post-ALI since ALI/ARDS often causes multi-organ failure. We hypothesized that ALI induces time-dependent lung PEx, which promotes extravasation in the heart, liver, kidney, spleen, pancreas, and gastrointestinal (GI) tract, in a time-dependent manner. To test our hypothesis, we administered bleomycin or saline via tracheal intubation in 8-week-old Sprague Dawley rats. At the terminal experiments, Evans Blue was injected (IV) through the femoral vein to allow for the visualization of PEx. Plasma extravasation of desired organs was evaluated at 3-, 7-, 14-, 21-, and 28-days after bleomycin or saline treatment by evaluating Evans Blue concentrations calorimetrically at fluorescence excitation wavelength of 620 nm (bandwidth 10 nm) and an emission wavelength of 680 nm (bandwidth 40 nm). Data show that ALI induces lung PEx beginning at day 3 and peaking between 7 and 21 days. Extravasation was also seen in all organs at varying degrees beginning at day 3 and peaking between days 7 and 14. Resolution appears to start after day 21 and continues past day 28. We conclude that ALI caused by bleomycin incites a time-dependent PEx of the lungs and multiple other organs.
Collapse
Affiliation(s)
- Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
- Deptrtment of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Steven J. Lisco
- Deptrtment of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Han-Jun Wang
- Deptrtment of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Han-Jun Wang,
| |
Collapse
|
13
|
Imai E, Morohashi Y, Mishima K, Ozaki T, Igarashi K, Wakabayashi G. A goal-directed therapy protocol for preventing acute kidney injury after laparoscopic liver resection: a retrospective observational cohort study. Surg Today 2022; 52:1262-1274. [PMID: 35044519 DOI: 10.1007/s00595-022-02453-3] [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: 09/07/2021] [Accepted: 11/26/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Postoperative acute kidney injury (AKI) remains a serious complication of liver resection with restrictive fluid therapy. However, unlike open hepatectomy, laparoscopic liver resection (LLR) does not have established anesthesia management strategies. We compared our goal-directed therapy (GDT) protocol for LLR with/without carperitide and the conventional restrictive method regarding AKI prevention. METHODS The GDT thresholds in this retrospective observational cohort study were as follows: stroke volume variation, ≤ 15%; pulse pressure variation, ≤ 13%; oxygen delivery index, ≥ 600 mL/min/m2; and mean arterial pressure (MAP), ≥ 55 mmHg. If the thresholds were not achieved, a 250 mL infusion fluid bolus was administered. The MAP target was changed to > 65 mmHg if the urine output was < 0.3 mL/kg/h. Postoperative AKI within 48 h and perioperative outcomes within 90 days were analyzed. RESULTS Forty-seven propensity score-matched pairs from 127 patients were investigated. We adjusted for AKI risk factors and surgical difficulty; 46.8% of the GDT group received carperitide. The GDT group had a lower postoperative AKI rate (10.6% vs. 27.7%, P = 0.04) and shorter overall (P = 0.04) and postoperative (P < 0.01) hospital stays than the conventional group. Furthermore, the GDT group received more intraoperative fluid (P = 0.001) and phenylephrine (P = 0.02), without significant increases in blood loss and transfusion volume, than the conventional group. CONCLUSIONS GDT reduced the AKI rates post-LLR.
Collapse
Affiliation(s)
- Eriya Imai
- Division of Anesthesia, Mitsui Memorial Hospital, Kanda-Izumi-cho-1, Chiyoda-ku, Tokyo, 101-8643, Japan.
| | | | - Kohei Mishima
- Department of Surgery, Ageo Central General Hospital, Saitama, Japan
| | - Takahiro Ozaki
- Department of Surgery, Ageo Central General Hospital, Saitama, Japan
| | - Kazuharu Igarashi
- Department of Surgery, Ageo Central General Hospital, Saitama, Japan
| | - Go Wakabayashi
- Department of Surgery, Ageo Central General Hospital, Saitama, Japan
| |
Collapse
|
14
|
Chen A, Yin L, Lee K, He JC. Similarities and Differences between COVID-19-Associated Nephropathy and HIV-Associated Nephropathy. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:1-12. [PMID: 35127839 PMCID: PMC8805054 DOI: 10.1159/000520235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/11/2021] [Indexed: 12/13/2022]
Abstract
Kidney disease is a major complication of viral infection, which can cause both acute and chronic kidney diseases via different mechanisms such as immune-mediated injury, kidney cell injury from a direct viral infection, systemic effects, and antiviral drug-induced nephrotoxicity. HIV-associated nephropathy (HIVAN), characterized by collapsing focal segmental glomerulosclerosis (cFSGS), has been described 2 decades ago as a major complication of acquired-immunodeficiency syndrome. The pathogenesis of HIVAN has been well studied, including viral entry, host response, and genetic factors. The incidence of this disease has been dramatically dropped with current antiretroviral therapy. In the recent severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic, acute kidney injury was also found to be a major complication in patients with (coronavirus disease) COVID-19. These patients also developed glomerular disease such as cFSGS in African Americans with apolipoprotein L1 risk alleles, similar to HIVAN. Whether SARS-CoV-2 can infect kidney cells locally remains controversial, but both local infection and systemic effects are likely involved in the pathogenesis of this disease. In this review, we present a comparison of the clinical presentations, pathological findings, disease mechanisms, and potential treatments between HIVAN and COVID-19. Leveraging the knowledge in HIVAN and experimental approaches used to study HIVAN will facilitate the exploration in the pathogenesis of COVID-19-associated kidney disease and improve our management of COVID-19 patients.
Collapse
Affiliation(s)
- Anqun Chen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Lijun Yin
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicineat Mount Sinai, New York, New York, USA
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicineat Mount Sinai, New York, New York, USA
- Renal Program, James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
15
|
Toro-Pérez J, Rodrigo R. Contribution of oxidative stress in the mechanisms of postoperative complications and multiple organ dysfunction syndrome. Redox Rep 2021; 26:35-44. [PMID: 33622196 PMCID: PMC7906620 DOI: 10.1080/13510002.2021.1891808] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND The extent of the damage following surgery has been subject of study for several years. Numerous surgical complications can impact postoperative quality of life of patients and even can cause mortality. Although these complications are generally due to multifactorial mechanisms, oxidative stress plays a key pathophysiological role. Moreover, oxidative stress could be an unavoidable effect derived even from the surgical procedure itself. METHODS A systematic review was performed following an electronic search of Pubmed and ScienceDirect databases. Keywords such as sepsis, oxidative stress, organ dysfunction, antioxidants, outcomes in postoperative complications, among others, were used. Review articles were preferably used between the years 2015 onwards, not excluding older ones. RESULTS The vast majority point to the role of oxidative stress in generating greater damage and worse prognosis in postoperative patients without the necessary care and precautions, taking importance on the use of antioxidants to prevent this problem. DISCUSSIONS Oxidative stress represents a common final pathway related to pathological processes such as inflammation or ischemia-reperfusion, among others. The expression of greater severity of these complications can result in multiple organ dysfunction or sepsis. The aim of this study was to present an update of the role of oxidative stress on surgical postoperative complications.
Collapse
Affiliation(s)
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|
16
|
Segar J, Jetton JG. Diuretic use, acute kidney injury, and premature infants: the call for evidence-based guidelines. Pediatr Nephrol 2021; 36:3807-3811. [PMID: 34258644 DOI: 10.1007/s00467-021-05201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Jeffrey Segar
- Departments of Pediatrics and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jennifer G Jetton
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
17
|
Jayaram A, Deer E, Amaral LM, Campbell N, Vaka VR, Cunningham M, Ibrahim T, Cornelius DC, LaMarca BB. The role of tumor necrosis factor in triggering activation of natural killer cell, multi-organ mitochondrial dysfunction and hypertension during pregnancy. Pregnancy Hypertens 2021; 24:65-72. [PMID: 33677421 PMCID: PMC8681863 DOI: 10.1016/j.preghy.2021.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/13/2020] [Accepted: 02/04/2021] [Indexed: 11/24/2022]
Abstract
Pre-eclampsia (PE) is a hypertensive disorder of pregnancy associated with chronic inflammation, mitochondrial (mt) dysfunction and fetal demise. Natural Killer cells (NK cells) are critical for the innate immune response against tumors or infection by disrupting cellular mt function and causing cell death. Although NK cells can be stimulated by Tumor necrosis factor alpha (TNF-α), we don't know the role of TNF-α on NK cell mediated mt dysfunction during PE. Our objective was to determine if mechanisms of TNF-α induced hypertension included activation of NK cells and multi-organ mt dysfunction during pregnancy. Pregnant rats were divided into 2 groups: normal pregnant (NP) (n = 18) and NP + TNF-α (n = 18). On gestational day 14, TNF-α (50 ng/ml) was infused via mini-osmotic pump and on day 18, carotid artery catheters were inserted. Blood pressure (MAP) and samples were collected on day 19. TNF-α increased MAP (109 ± 2 vs 100 ± 2, p < 0.05), circulating cytolytic NK cells (0.771 ± 0.328 vs.0.008 ± 0.003% gated, <0.05) and fetal reabsorptions compared to NP rats. Moreover, TNF-α caused mtROS in the placenta (12976 ± 7038 vs 176.9 ± 68.04% fold, p < 0.05) and in the kidney (2191 ± 1027 vs 816 ± 454.7% fold, p < 0.05) compared to NP rats. TNF-α induced hypertension is associated fetal demise, activation of NK cells and multi-organ mt dysfunction which could be mechanisms for fetal demise and hypertension. Understanding of the mechanisms by which TNF-α causes pathology is important for the use of anti-TNF-α therapeutic agents in pregnancies complicated by PE.
Collapse
Affiliation(s)
- Aswathi Jayaram
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Lorena M Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Venkata Ramana Vaka
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Mark Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States
| | - Babbette B LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, N State St, Jackson, MS 39216, United States.
| |
Collapse
|
18
|
Abstract
To characterize current evidence and current foci of perioperative clinical trials, we systematically reviewed Medline and identified perioperative trials involving 100 or more adult patients undergoing surgery and reporting renal end points that were published in high-impact journals since 2004. We categorized the 101 trials identified based on the nature of the intervention and summarized major trial findings from the five categories most applicable to perioperative management of patients. Trials that targeted ischemia suggested that increasing perioperative renal oxygen delivery with inotropes or blood transfusion does not reliably mitigate acute kidney injury (AKI), although goal-directed therapy with hemodynamic monitors appeared beneficial in some trials. Trials that have targeted inflammation or oxidative stress, including studies of nonsteroidal anti-inflammatory drugs, steroids, N-acetylcysteine, and sodium bicarbonate, have not shown renal benefits, and high-dose perioperative statin treatment increased AKI in some patient groups in two large trials. Balanced crystalloid intravenous fluids appear safer than saline, and crystalloids appear safer than colloids. Liberal compared with restrictive fluid administration reduced AKI in a recent large trial in open abdominal surgery. Remote ischemic preconditioning, although effective in several smaller trials, failed to reduce AKI in two larger trials. The translation of promising preclinical therapies to patients undergoing surgery remains poor, and most interventions that reduced perioperative AKI compared novel surgical management techniques or existing processes of care rather than novel pharmacologic interventions.
Collapse
Affiliation(s)
- David R McIlroy
- Division of Cardiothoracic Anesthesiology, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Marcos G Lopez
- Division of Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Frederic T Billings
- Division of Cardiothoracic Anesthesiology, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; Division of Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
19
|
Brignone J, Assersen KB, Jensen M, Jensen BL, Kloster B, Jønler M, Lund L. Protection of kidney function and tissue integrity by pharmacologic use of natriuretic peptides and neprilysin inhibitors. Pflugers Arch 2021; 473:595-610. [PMID: 33844072 DOI: 10.1007/s00424-021-02555-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/03/2021] [Accepted: 03/06/2021] [Indexed: 12/11/2022]
Abstract
With variable potencies atrial-, brain-type and c-type natriuretic peptides (NP)s, best documented for ANP and its analogues, promote sodium and water excretion, renal blood flow, lipolysis, lower blood pressure, and suppress renin and aldosterone secretion through interaction predominantly with cGMP-coupled NPR-A receptor. Infusion of especially ANP and its analogues up to 50 ng/kg/min in patients with high risk of acute kidney injury (cardiac vascular bypass surgery, intraabdominal surgery, direct kidney surgery) protects kidney function (GFR, plasma flow, medullary flow, albuminuria, renal replacement therapy, tissue injury) at short term and also long term and likely additively with the diuretic furosemide. This documents a pharmacologic potential for the pathway. Neprilysin (NEP, neutral endopeptidase) degrades NPs, in particular ANP, and angiotensin II. The drug LCZ696, a mixture of the neprilysin inhibitor sacubitril and the ANGII-AT1 receptor blocker valsartan, was FDA approved in 2015 and marketed as Entresto®. In preclinical studies of kidney injury, LCZ696 and NPs lowered plasma creatinine, countered hypoxia and oxidative stress, suppressed proinflammatory cytokines, and inhibited fibrosis. Few randomized clinical studies exist and were designed with primary cardiac outcomes. The studies showed that LCZ696/entresto stabilized and improved glomerular filtration rate in patients with chronic kidney disease. LCZ696 is safe to use concerning kidney function and stabilizes or increases GFR. In perspective, combined AT1 and neprilysin inhibition is a promising approach for long-term renal protection in addition to AT1 receptor blockers in acute kidney injury and chronic kidney disease.
Collapse
Affiliation(s)
- Juan Brignone
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark. .,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | - Kasper Bostlund Assersen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Mia Jensen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Brian Kloster
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark
| | - Morten Jønler
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark
| | - Lars Lund
- Department of Urology, Aalborg University Hospital, Aalborg, Denmark.,Department of Urology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
20
|
Chen J, Wang W, Tang Y, Huang XR, Yu X, Lan HY. Inflammatory stress in SARS-COV-2 associated Acute Kidney Injury. Int J Biol Sci 2021; 17:1497-1506. [PMID: 33907513 PMCID: PMC8071761 DOI: 10.7150/ijbs.58791] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 01/08/2023] Open
Abstract
Increasing clinical evidence shows that acute kidney injury (AKI) is a common and severe complication in critically ill COVID-19 patients. The older age, the severity of COVID-19 infection, the ethnicity, and the history of smoking, diabetes, hypertension, and cardiovascular disease are the risk factor for AKI in COVID-19 patients. Of them, inflammation may be a key player in the pathogenesis of AKI in patients with COVID-19. It is highly possible that SARS-COV-2 infection may trigger the activation of multiple inflammatory pathways including angiotensin II, cytokine storm such as interleukin-6 (IL-6), C-reactive protein (CRP), TGF-β signaling, complement activation, and lung-kidney crosstalk to cause AKI. Thus, treatments by targeting these inflammatory molecules and pathways with a monoclonal antibody against IL-6 (Tocilizumab), C3 inhibitor AMY-101, anti-C5 antibody, anti-TGF-β OT-101, and the use of CRRT in critically ill patients may represent as novel and specific therapies for AKI in COVID-19 patients.
Collapse
Affiliation(s)
- Junzhe Chen
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Nephrology, The Third Affiliated hospital, Southern Medical university, Guangzhou, China
| | - Wenbiao Wang
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Ying Tang
- Department of Nephrology, The Third Affiliated hospital, Southern Medical university, Guangzhou, China
| | - Xiao-ru Huang
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Science, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xueqing Yu
- Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Science, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Hui-Yao Lan
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
21
|
Ricksten SE, Bragadottir G, Lannemyr L, Redfors B, Skytte J. Renal Hemodynamics, Function, and Oxygenation in Critically Ill Patients and after Major Surgery. KIDNEY360 2021; 2:894-904. [PMID: 35373068 PMCID: PMC8791344 DOI: 10.34067/kid.0007012020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/02/2021] [Indexed: 02/04/2023]
Abstract
This review outlines the available data from the work of our group on renal hemodynamics, function, and oxygenation in patients who are critically ill with acute renal dysfunction, such as those with postoperative AKI, those in early clinical septic shock, in patients undergoing cardiac surgery with cardiopulmonary bypass, or in patients undergoing liver transplantation. We also provide information on renal hemodynamics, function, and oxygenation in patients with chronic renal impairment due to congestive heart failure. This review will argue that, for all of these groups of patients, the common denominator is that renal oxygenation is impaired due to a lower renal oxygen delivery or a pronounced increase in renal oxygen consumption.
Collapse
|
22
|
Chong WH, Saha BK. Relationship Between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and the Etiology of Acute Kidney Injury (AKI). Am J Med Sci 2021; 361:287-296. [PMID: 33358501 PMCID: PMC7590839 DOI: 10.1016/j.amjms.2020.10.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/23/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since it was first recognized in December 2019, it has resulted in the ongoing worldwide pandemic. Although acute hypoxic respiratory failure (AHRF) and acute respiratory distress syndrome (ARDS) are the main features of the disease, the involvement of other organs needs to be explored. There has been a growing concern regarding the association between acute kidney injury (AKI) and poor outcomes in SARS-CoV-2 patients. Based on current observational data, AKI is the 2nd most common cause of morbidity and mortality behind ARDS in SARS-CoV-2 patients. Angiotensin-converting enzyme 2 (ACE2) receptor has been shown to be the cornerstone of SARS-CoV-2 infection and possibly plays a significant role in the occurrence of renal injury. The pathogenesis of AKI is likely multifactorial that involves not only direct viral invasion but also dysregulated immune response in the form of cytokine storm, ischemia to kidneys, hypercoagulable state, and rhabdomyolysis, among others. We performed a literature search of the Pubmed and Google Scholar database from 1996 to 2020 using the following keywords: severe acute respiratory syndrome coronavirus 2, coronavirus disease 2019, angiotensin-converting enzyme 2 receptor, and acute kidney injury to find the most pertinent and highest-quality of evidence. Any cited references were reviewed to identify relevant literature. The purpose of this review is to discuss, explore, and summarize the relationship between AKI in SARS-CoV-2 patients, with a focus on its epidemiology, association with ACE2 receptors, and pathophysiology of AKI.
Collapse
Affiliation(s)
- Woon H Chong
- Department of Pulmonary and Critical Care Medicine; Albany Medical Center, Albany, New York, USA.
| | - Biplab K Saha
- Department of Pulmonary and Critical Care; Ozarks Medical Center, West Plains, Missouri, USA
| |
Collapse
|
23
|
Abstract
Physical trauma can affect any individual and is globally accountable for more than one in every ten deaths. Although direct severe kidney trauma is relatively infrequent, extrarenal tissue trauma frequently results in the development of acute kidney injury (AKI). Various causes, including haemorrhagic shock, rhabdomyolysis, use of nephrotoxic drugs and infectious complications, can trigger and exacerbate trauma-related AKI (TRAKI), particularly in the presence of pre-existing or trauma-specific risk factors. Injured, hypoxic and ischaemic tissues expose the organism to damage-associated and pathogen-associated molecular patterns, and oxidative stress, all of which initiate a complex immunopathophysiological response that results in macrocirculatory and microcirculatory disturbances in the kidney, and functional impairment. The simultaneous activation of components of innate immunity, including leukocytes, coagulation factors and complement proteins, drives kidney inflammation, glomerular and tubular damage, and breakdown of the blood-urine barrier. This immune response is also an integral part of the intense post-trauma crosstalk between the kidneys, the nervous system and other organs, which aggravates multi-organ dysfunction. Necessary lifesaving procedures used in trauma management might have ambivalent effects as they stabilize injured tissue and organs while simultaneously exacerbating kidney injury. Consequently, only a small number of pathophysiological and immunomodulatory therapeutic targets for TRAKI prevention have been proposed and evaluated.
Collapse
|
24
|
van Raalte DH, Bjornstad P. Role of sodium-glucose cotransporter 2 inhibition to mitigate diabetic kidney disease risk in type 1 diabetes. Nephrol Dial Transplant 2020; 35:i24-i32. [PMID: 32003832 PMCID: PMC6993198 DOI: 10.1093/ndt/gfz228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Indexed: 12/17/2022] Open
Abstract
Diabetic kidney disease (DKD) is a common complication of type 1 diabetes (T1D) and a major risk factor for premature death from cardiovascular disease (CVD). Current treatments, such as control of hyperglycaemia and hypertension, are beneficial, but only partially protect against DKD. Finding new, safe and effective therapies to halt nephropathy progression has proven to be challenging. Sodium-glucose cotransporter 2 (SGLT2) inhibitors have demonstrated, in addition to glycaemic lowering, impressive protection against DKD and CVD progression in people with type 2 diabetes. Although these beneficial cardiorenal effects may also apply to people with T1D, supporting data are lacking. Furthermore, the increased rates of euglycaemic diabetic ketoacidosis may limit the use of this class in people with T1D. In this review we highlight the pathophysiology of DKD in T1D and the unmet need that exists. We further detail the beneficial and adverse effects of SGLT2 inhibitors based on their mechanism of action. Finally, we balance the effects in people with T1D and indicate future lines of research.
Collapse
Affiliation(s)
- Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.,Division of Nephrology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
25
|
Legouis D, Ricksten SE, Faivre A, Verissimo T, Gariani K, Verney C, Galichon P, Berchtold L, Feraille E, Fernandez M, Placier S, Koppitch K, Hertig A, Martin PY, Naesens M, Pugin J, McMahon AP, Cippà PE, de Seigneux S. Altered proximal tubular cell glucose metabolism during acute kidney injury is associated with mortality. Nat Metab 2020; 2:732-743. [PMID: 32694833 DOI: 10.1038/s42255-020-0238-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 06/11/2020] [Indexed: 01/16/2023]
Abstract
Acute kidney injury (AKI) is strongly associated with mortality, independently of its cause. The kidney contributes to up to 40% of systemic glucose production by gluconeogenesis during fasting and under stress conditions. Whether kidney gluconeogenesis is impaired during AKI and how this might influence systemic metabolism remain unknown. Here we show that glucose production and lactate clearance are impaired during human and experimental AKI by using renal arteriovenous catheterization in patients, lactate tolerance testing in mice and glucose isotope labelling in rats. Single-cell transcriptomics reveal that gluconeogenesis is impaired in proximal tubule cells during AKI. In a retrospective cohort of critically ill patients, we demonstrate that altered glucose metabolism during AKI is a major determinant of systemic glucose and lactate levels and is strongly associated with mortality. Thiamine supplementation increases lactate clearance without modifying renal function in mice with AKI, enhances glucose production by renal tubular cells ex vivo and is associated with reduced mortality and improvement of the metabolic pattern in a retrospective cohort of critically ill patients with AKI. This study highlights an unappreciated systemic role of renal glucose and lactate metabolism under stress conditions, delineates general mechanisms of AKI-associated mortality and introduces a potential intervention targeting metabolism for a highly prevalent clinical condition with limited therapeutic options.
Collapse
Affiliation(s)
- David Legouis
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital and University of Geneva, Geneva, Switzerland.
- Division of Intensive Care, University Hospital of Geneva, Geneva, Switzerland.
| | - Sven-Erick Ricksten
- Department of Anaesthesiology, Intensive Care Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Faivre
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital and University of Geneva, Geneva, Switzerland
| | - Thomas Verissimo
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital and University of Geneva, Geneva, Switzerland
| | - Karim Gariani
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Charles Verney
- French National Institute of Health and Medical Research UMR_S1155, Rare and Common Kidney Diseases, Matrix Remodeling and Repair; AP-HP, Tenon Hospital, Renal Intensive Care Unit, Sorbonne Université, Paris, France
| | - Pierre Galichon
- French National Institute of Health and Medical Research UMR_S1155, Rare and Common Kidney Diseases, Matrix Remodeling and Repair; AP-HP, Tenon Hospital, Renal Intensive Care Unit, Sorbonne Université, Paris, France
| | - Lena Berchtold
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital and University of Geneva, Geneva, Switzerland
- Service of Nephrology, University Hospitals of Leuven, Leuven, Belgium
| | - Eric Feraille
- Department of Cell Biology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Marylise Fernandez
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital and University of Geneva, Geneva, Switzerland
| | - Sandrine Placier
- French National Institute of Health and Medical Research UMR_S1155, Rare and Common Kidney Diseases, Matrix Remodeling and Repair; AP-HP, Tenon Hospital, Renal Intensive Care Unit, Sorbonne Université, Paris, France
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Alexandre Hertig
- French National Institute of Health and Medical Research UMR_S1155, Rare and Common Kidney Diseases, Matrix Remodeling and Repair; AP-HP, Tenon Hospital, Renal Intensive Care Unit, Sorbonne Université, Paris, France
| | - Pierre-Yves Martin
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital and University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Maarten Naesens
- Service of Nephrology, University Hospitals of Leuven, Leuven, Belgium
| | - Jérôme Pugin
- Division of Intensive Care, University Hospital of Geneva, Geneva, Switzerland
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Pietro E Cippà
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Sophie de Seigneux
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital and University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
26
|
Abstract
Although kidney oxygen tensions are heterogenous, and mostly below renal vein level, the nephron is highly dependent on aerobic metabolism for active tubular transport. This renders the kidney particularly susceptible to hypoxia, which is considered a main characteristic and driver of acute and chronic kidney injury, albeit the evidence supporting this assumption is not entirely conclusive. Kidney transplants are exposed to several conditions that may interfere with the balance between oxygen supply and consumption, and enhance hypoxia and hypoxic injury. These include conditions leading to and resulting from brain death of kidney donors, ischemia and reperfusion during organ donation, storage and transplantation, postoperative vascular complications, vasoconstriction induced by immunosuppression, and impaired perfusion resulting from interstitial edema, inflammation, and fibrosis. Acute graft injury, the immediate consequence of hypoxia and reperfusion, results in delayed graft function and increased risk of chronic graft failure. Although current strategies to alleviate hypoxic/ischemic graft injury focus on limiting injury (eg, by reducing cold and warm ischemia times), experimental evidence suggests that preconditioning through local or remote ischemia, or activation of the hypoxia-inducible factor pathway, can decrease hypoxic injury. In combination with ex vivo machine perfusion such approaches hold significant promise for improving transplantation outcomes.
Collapse
Affiliation(s)
- Christian Rosenberger
- Department of Nephrology and Medical Intensive Care, Charité Universitaetsmedizin Berlin, Berlin, Germany.
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
27
|
Wolf JM, Stefanovski D, Silverstein DC. Retrospective evaluation of the influence of azotemia on plasma lactate concentrations in hypotensive dogs and cats (2008-2018): 337 cases. J Vet Emerg Crit Care (San Antonio) 2020; 30:449-454. [PMID: 32579283 DOI: 10.1111/vec.12982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To evaluate the relationship between azotemia and plasma lactate concentration in hypotensive dogs and cats presented to an emergency department. DESIGN Retrospective case-control study. SETTING University veterinary teaching hospital. ANIMALS The electronic medical record database was searched for dogs and cats presented to the emergency department that had severe azotemia (creatinine ≥ 443.1 μmol/L [5 mg/dL]), hypotension (systolic blood pressure ≤ 90 mm Hg), and a plasma lactate measurement within 2 h of each another. Non-azotemic, normotensive dogs and cats; non-azotemic, hypotensive dogs and cats; and azotemic, normotensive dogs and cats that presented to the emergency department were used as control populations. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Severely azotemic, hypotensive dogs (n = 10) and cats (n = 63) had a lower plasma lactate than non-azotemic, hypotensive dogs and cats (P = 0.031 and P < 0.001, respectively). Median plasma lactate concentrations in hypotensive dogs (1.75 mmol/L) and cats (1.90 mmol/L) with severe azotemia were within reference intervals. CONCLUSIONS Hypotensive dogs and cats with severe azotemia have decreased plasma lactate concentrations as compared to hypotensive, non-azotemic dogs and cats. The median plasma lactate in azotemic, hypotensive dogs and cats was within reference intervals. This may be due to either decreased cellular production of lactate or increased excretion of lactate. Further research is needed to determine which of these mechanisms is responsible and the clinical significance of plasma lactate concentrations in azotemic, hypotensive dogs and cats.
Collapse
Affiliation(s)
- Jacob M Wolf
- Department of Clinical Studies and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Darko Stefanovski
- Department of Clinical Studies and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Deborah C Silverstein
- Department of Clinical Studies and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Hesp AC, Schaub JA, Prasad PV, Vallon V, Laverman GD, Bjornstad P, van Raalte DH. The role of renal hypoxia in the pathogenesis of diabetic kidney disease: a promising target for newer renoprotective agents including SGLT2 inhibitors? Kidney Int 2020; 98:579-589. [PMID: 32739206 DOI: 10.1016/j.kint.2020.02.041] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/06/2020] [Accepted: 02/26/2020] [Indexed: 12/17/2022]
Abstract
Diabetic kidney disease is the most common cause of end-stage kidney disease and poses a major global health problem. Finding new, safe, and effective strategies to halt this disease has proven to be challenging. In part that is because the underlying mechanisms are complex and not fully understood. However, in recent years, evidence has accumulated suggesting that chronic hypoxia may be the primary pathophysiological pathway driving diabetic kidney disease and chronic kidney disease of other etiologies and was called the chronic hypoxia hypothesis. Hypoxia is the result of a mismatch between oxygen delivery and oxygen demand. The primary determinant of oxygen delivery is renal perfusion (blood flow per tissue mass), whereas the main driver of oxygen demand is active sodium reabsorption. Diabetes mellitus is thought to compromise the oxygen balance by impairing oxygen delivery owing to hyperglycemia-associated microvascular damage and exacerbate oxygen demand owing to increased sodium reabsorption as a result of sodium-glucose cotransporter upregulation and glomerular hyperfiltration. The resultant hypoxic injury creates a vicious cycle of capillary damage, inflammation, deposition of the extracellular matrix, and, ultimately, fibrosis and nephron loss. This review will frame the role of chronic hypoxia in the pathogenesis of diabetic kidney disease and its prospect as a promising therapeutic target. We will outline the cellular mechanisms of hypoxia and evidence for renal hypoxia in animal and human studies. In addition, we will highlight the promise of newer imaging modalities including blood oxygenation level-dependent magnetic resonance imaging and discuss salutary interventions such as sodium-glucose cotransporter 2 inhibition that (may) protect the kidney through amelioration of renal hypoxia.
Collapse
Affiliation(s)
- Anne C Hesp
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands.
| | - Jennifer A Schaub
- Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Pottumarthi V Prasad
- Department of Radiology, NorthShore University Health System, Evanston, Illinois, USA; Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Volker Vallon
- Department of Medicine, University of California San Diego and Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Gozewijn D Laverman
- Department of Internal Medicine, Ziekenhuis Groep Twente, Almelo, The Netherlands
| | - Petter Bjornstad
- Department of Medicine, Division of Nephrology, and Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Fogagnolo A, Taccone FS, Vincent JL, Benetto G, Cavalcante E, Marangoni E, Ragazzi R, Creteur J, Volta CA, Spadaro S. Using arterial-venous oxygen difference to guide red blood cell transfusion strategy. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:160. [PMID: 32312299 PMCID: PMC7171832 DOI: 10.1186/s13054-020-2827-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 03/06/2020] [Indexed: 01/28/2023]
Abstract
Background Guidelines recommend a restrictive red blood cell transfusion strategy based on hemoglobin (Hb) concentrations in critically ill patients. We hypothesized that the arterial-venous oxygen difference (A-V O2diff), a surrogate for the oxygen delivery to consumption ratio, could provide a more personalized approach to identify patients who may benefit from transfusion. Methods A prospective observational study including 177 non-bleeding adult patients with a Hb concentration of 7.0–10.0 g/dL within 72 h after ICU admission. The A-V O2diff, central venous oxygen saturation (ScvO2), and oxygen extraction ratio (O2ER) were noted when a patient’s Hb was first within this range. Transfusion decisions were made by the treating physician according to institutional policy. We used the median A-V O2diff value in the study cohort (3.7 mL) to classify the transfusion strategy in each patient as “appropriate” (patient transfused when the A-V O2diff > 3.7 mL or not transfused when the A-V O2diff ≤ 3.7 mL) or “inappropriate” (patient transfused when the A-V O2diff ≤ 3.7 mL or not transfused when the A-V O2diff > 3.7 mL). The primary outcome was 90-day mortality. Results Patients managed with an “appropriate” strategy had lower mortality rates (23/96 [24%] vs. 36/81 [44%]; p = 0.004), and an “appropriate” strategy was independently associated with reduced mortality (hazard ratio [HR] 0.51 [95% CI 0.30–0.89], p = 0.01). There was a trend to less acute kidney injury with the “appropriate” than with the “inappropriate” strategy (13% vs. 26%, p = 0.06), and the Sequential Organ Failure Assessment (SOFA) score decreased more rapidly (p = 0.01). The A-V O2diff, but not the ScvO2, predicted 90-day mortality in transfused (AUROC = 0.656) and non-transfused (AUROC = 0.630) patients with moderate accuracy. Using the ROC curve analysis, the best A-V O2diff cutoffs for predicting mortality were 3.6 mL in transfused and 3.5 mL in non-transfused patients. Conclusions In anemic, non-bleeding critically ill patients, transfusion may be associated with lower 90-day mortality and morbidity in patients with higher A-V O2diff. Trial registration ClinicalTrials.gov, NCT03767127. Retrospectively registered on 6 December 2018.
Collapse
Affiliation(s)
- Alberto Fogagnolo
- Department of Morphology, Surgery and Experimental Medicine, Section of Anaesthesia and Intensive Care, Azienda Ospedaliera-Universitaria Sant' Anna, University of Ferrara, 8, Aldo Moro, 44121, Ferrara, Italy
| | - Fabio Silvio Taccone
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean Louis Vincent
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Giulia Benetto
- Department of Morphology, Surgery and Experimental Medicine, Section of Anaesthesia and Intensive Care, Azienda Ospedaliera-Universitaria Sant' Anna, University of Ferrara, 8, Aldo Moro, 44121, Ferrara, Italy
| | - Elaine Cavalcante
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Elisabetta Marangoni
- Department of Morphology, Surgery and Experimental Medicine, Section of Anaesthesia and Intensive Care, Azienda Ospedaliera-Universitaria Sant' Anna, University of Ferrara, 8, Aldo Moro, 44121, Ferrara, Italy
| | - Riccardo Ragazzi
- Department of Morphology, Surgery and Experimental Medicine, Section of Anaesthesia and Intensive Care, Azienda Ospedaliera-Universitaria Sant' Anna, University of Ferrara, 8, Aldo Moro, 44121, Ferrara, Italy
| | - Jacques Creteur
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Carlo Alberto Volta
- Department of Morphology, Surgery and Experimental Medicine, Section of Anaesthesia and Intensive Care, Azienda Ospedaliera-Universitaria Sant' Anna, University of Ferrara, 8, Aldo Moro, 44121, Ferrara, Italy
| | - Savino Spadaro
- Department of Morphology, Surgery and Experimental Medicine, Section of Anaesthesia and Intensive Care, Azienda Ospedaliera-Universitaria Sant' Anna, University of Ferrara, 8, Aldo Moro, 44121, Ferrara, Italy.
| |
Collapse
|
30
|
Lytvyn Y, Bjornstad P, van Raalte DH, Heerspink HL, Cherney DZI. The New Biology of Diabetic Kidney Disease-Mechanisms and Therapeutic Implications. Endocr Rev 2020; 41:5601424. [PMID: 31633153 PMCID: PMC7156849 DOI: 10.1210/endrev/bnz010] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Diabetic kidney disease remains the most common cause of end-stage kidney disease in the world. Despite reductions in incidence rates of myocardial infarction and stroke in people with diabetes over the past 3 decades, the risk of diabetic kidney disease has remained unchanged, and may even be increasing in younger individuals afflicted with this disease. Accordingly, changes in public health policy have to be implemented to address the root causes of diabetic kidney disease, including the rise of obesity and diabetes, in addition to the use of safe and effective pharmacological agents to prevent cardiorenal complications in people with diabetes. The aim of this article is to review the mechanisms of pathogenesis and therapies that are either in clinical practice or that are emerging in clinical development programs for potential use to treat diabetic kidney disease.
Collapse
Affiliation(s)
- Yuliya Lytvyn
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Petter Bjornstad
- Department of Medicine, Division of Nephrology, Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel H van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Netherlands
| | - Hiddo L Heerspink
- The George Institute for Global Health, Sydney, Australia.,Department of Clinical Pharmacology, University of Groningen, Groningen, Netherlands
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Parsons M, Greenberg J, Parikh C, Brown J, Parker D, Zhu J, Vricella L, Everett AD. Post-operative acute kidney injury is associated with a biomarker of acute brain injury after paediatric cardiac surgery. Cardiol Young 2020; 30:505-510. [PMID: 32223775 DOI: 10.1017/s1047951120000451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Children with CHD who undergo cardiopulmonary bypass are at an increased risk of acute kidney injury. This study evaluated the association of end-organ specific injury plasma biomarkers for brain: glial fibrillary acidic protein and heart: Galectin 3, soluble suppression of tumorgenicity 2, and N-terminal pro b-type natriuretic peptide with acute kidney injury in children undergoing cardiopulmonary bypass. MATERIALS AND METHODS We enrolled consecutive children undergoing cardiac surgery with cardiopulmonary bypass. Blood samples were collected pre-bypass in the operating room and in the immediate post-operative period. Acute kidney injury was defined as a rise of serum creatinine ≥50% from pre-operative baseline within 7 days after surgery. RESULTS Overall, 162 children (mean age 4.05 years, sd 5.28 years) were enrolled. Post-operative acute kidney injury developed in 55 (34%) children. Post-operative plasma glial fibrillary acidic protein levels were significantly higher in patients with acute kidney injury (median 0.154 (inter-quartile range 0.059-0.31) ng/ml) compared to those without acute kidney injury (median 0.056 (inter-quartile range 0.001-0.125) ng/ml) (p = 0.043). After adjustment for age, weight, and The Society of Thoracic Surgeons-European Association for Cardio-Thoracic Surgery category, each natural log increase in post-operative glial fibrillary acidic protein was significantly associated with a higher risk for subsequent acute kidney injury (adjusted odds ratio glial fibrillary acidic protein 1.25; 95% confidence interval 1.01-1.59). Pre/post-operative levels of galectin 3, soluble suppression of tumorgenicity 2, and N-terminal pro b-type natriuretic peptide did not significantly differ between patients with and without acute kidney injury. CONCLUSIONS Higher plasma glial fibrillary acidic protein levels measured in the immediate post-operative period were independently associated with subsequent acute kidney injury in children after cardiopulmonary bypass. Elevated glial fibrillary acidic protein likely reflects intraoperative brain injury which may occur in the context of acute kidney injury-associated end-organ dysfunction.
Collapse
Affiliation(s)
- Michael Parsons
- Department of Pediatrics, The Helen B. Taussig Congenital Heart Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jason Greenberg
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Chirag Parikh
- Department of Pediatrics, The Helen B. Taussig Congenital Heart Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeremiah Brown
- Department of Epidemiology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Devin Parker
- Department of Epidemiology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Jie Zhu
- Department of Pediatrics, The Helen B. Taussig Congenital Heart Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luca Vricella
- Department of Pediatric Cardiac Surgery, University of Chicago School of Medicine, Chicago, IL, USA
| | - Allen D Everett
- Department of Pediatrics, The Helen B. Taussig Congenital Heart Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
32
|
Abstract
The endothelial glycocalyx (EG) is the most luminal layer of the blood vessel, growing on and within the vascular wall. Shedding of the EG plays a central role in many critical illnesses. Degradation of the EG is associated with increased morbidity and mortality. Certain illnesses and iatrogenic interventions can cause degradation of the EG. It is not known whether restitution of the EG promotes the survival of the patient. First trials that focus on the reorganization and/or restitution of the EG seem promising. Nevertheless, the step "from bench to bedside" is still a big one.
Collapse
Affiliation(s)
- Jan Jedlicka
- Department of Anaesthesiology, University Hospital of Munich (LMU), Nussbaumstr. 20, Munich 80336, Germany
| | - Bernhard F Becker
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-University, Marchioninistr. 27, Munich 81377, Germany
| | - Daniel Chappell
- Department of Anaesthesiology, University Hospital of Munich (LMU), Marchioninistr. 15, Munich 81377, Germany.
| |
Collapse
|
33
|
Wiromrat P, Bjornstad P, Vinovskis C, Chung LT, Roncal C, Pyle L, Lanaspa MA, Johnson RJ, Cherney DZ, Reznick-Lipina TK, Bishop F, Maahs DM, Wadwa RP. Elevated copeptin, arterial stiffness, and elevated albumin excretion in adolescents with type 1 diabetes. Pediatr Diabetes 2019; 20:1110-1117. [PMID: 31433534 PMCID: PMC7151746 DOI: 10.1111/pedi.12909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/18/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE We sought to evaluate copeptin concentrations in adolescents with and without type 1 diabetes (T1D) and examine the associations between copeptin and measures of arterial stiffness and kidney dysfunction. RESEARCH DESIGN AND METHODS This analysis included 169 adolescents with T1D (12-19 years of age, 59% girls, mean HbA1c 9.0 ± 1.5% and diabetes duration of 8.6 ± 2.9 years), in addition to 61 controls without T1D. Arterial stiffness including carotid-femoral pulse wave velocity (CF-PWV), carotid-radial PWV (CR-PWV), augmentation index normalized to heart rate of 75 bpm (AIx@HR75), and brachial artery distensibility (BAD). Serum copeptin, urinary albumin-to-creatinine ratio (UACR), and estimated glomerular filtration rate (eGFR) by serum creatinine and cystatin C were also assessed. RESULTS Compared to controls, adolescents with T1D had higher median (Q1-Q3) copeptin (7.5 [5.2-11.3] vs 6.4 [4.8-8.3] pmol/L, P = .01), mean ± SD eGFR (121 ± 23 vs 112 ± 16 mL/min/1.73m2 , P = .002) and lower BAD (7.1 ± 1.3 vs 7.2 ± 1.2%, P = .02). Adolescents with T1D in the in high tertile copeptin group (>9.1 pmol/L) had higher AIx@HR75 (10.7 ± 1.2 vs 5 ± 1.2, P = .001), CR-PWV (5.30 ± 1.0 vs 5.18 ± 1.0 m/s, P = .04), and UACR (12 ± 1 vs 8 ± 1 mg/g, P = .025) compared to those in low tertile (<5.8 pmol/L) after adjusting for age, sex, and eGFR. Copeptin inversely associated with CF-PWV independent of age, sex, eGFR, SBP, and HbA1c in T1D adolescents. CONCLUSIONS Our data demonstrate that elevated copeptin was associated with worse arterial stiffness in adolescents with T1D. These findings suggest that copeptin could improve CVD risk stratification in adolescents with T1D.
Collapse
Affiliation(s)
- Pattara Wiromrat
- Section of Endocrinology, Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States,Section of Nephrology, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - Carissa Vinovskis
- Section of Endocrinology, Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - Linh T. Chung
- Section of Endocrinology, Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - Carlos Roncal
- Section of Nephrology, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - Laura Pyle
- Section of Endocrinology, Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States,Department of Biostatistics and Informatics, University of Colorado Denver School of Public Health, Aurora, Colorado, United States
| | - Miguel A. Lanaspa
- Section of Nephrology, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - Richard J. Johnson
- Section of Nephrology, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - David Z. Cherney
- Department of Nephrology, University of Toronto School of Medicine, Ontario, Canada
| | - Tyler K. Reznick-Lipina
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - Franziska Bishop
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| | - David M. Maahs
- Section of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California,Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA
| | - R. Paul Wadwa
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver School of Medicine, Aurora, Colorado, United States
| |
Collapse
|
34
|
Mansour SG, Martin TG, Obeid W, Pata RW, Myrick KM, Kukova L, Jia Y, Bjornstad P, El-Khoury JM, Parikh CR. The Role of Volume Regulation and Thermoregulation in AKI during Marathon Running. Clin J Am Soc Nephrol 2019; 14:1297-1305. [PMID: 31413064 PMCID: PMC6730516 DOI: 10.2215/cjn.01400219] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/08/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND OBJECTIVES Marathon runners develop transient AKI with urine sediments and injury biomarkers suggesting nephron damage. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS To investigate the etiology, we examined volume and thermoregulatory responses as possible mechanisms in runners' AKI using a prospective cohort of runners in the 2017 Hartford Marathon. Vitals, blood, and urine samples were collected in 23 runners 1 day premarathon and immediately and 1 day postmarathon. We measured copeptin at each time point. Continuous core body temperature, sweat sodium, and volume were assessed during the race. The primary outcome of interest was AKI, defined by AKIN criteria. RESULTS Runners ranged from 22 to 63 years old; 43% were men. Runners lost a median (range) of 2.34 (0.50-7.21) g of sodium and 2.47 (0.36-6.81) L of volume via sweat. After accounting for intake, they had a net negative sodium and volume balance at the end of the race. The majority of runners had increases in core body temperature to 38.4 (35.8-41)°C during the race from their baseline. Fifty-five percent of runners developed AKI, yet 74% had positive urine microscopy for acute tubular injury. Runners with more running experience and increased participation in prior marathons developed a rise in creatinine as compared with those with lesser experience. Sweat sodium losses were higher in runners with AKI versus non-AKI (median, 3.41 [interquartile range (IQR), 1.7-4.8] versus median, 1.4 [IQR, 0.97-2.8] g; P=0.06, respectively). Sweat volume losses were higher in runners with AKI versus non-AKI (median, 3.89 [IQR, 1.49-5.09] versus median, 1.66 [IQR, 0.72-2.84] L; P=0.03, respectively). Copeptin was significantly higher in runners with AKI versus those without (median, 79.9 [IQR, 25.2-104.4] versus median, 11.3 [IQR, 6.6-43.7]; P=0.02, respectively). Estimated temperature was not significantly different. CONCLUSIONS All runners experienced a substantial rise in copeptin and body temperature along with salt and water loss due to sweating. Sodium and volume loss via sweat as well as plasma copeptin concentrations were associated with AKI in runners. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2019_08_13_CJASNPodcast_19_09_.mp3.
Collapse
Affiliation(s)
- Sherry G Mansour
- Program of Applied Translational Research, Department of Medicine and.,Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | | | - Wassim Obeid
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Rachel W Pata
- Department of Physical Therapy, Quinnipiac University, North Haven, Connecticut; and
| | - Karen M Myrick
- University of Saint Joseph, School of Interdisciplinary Health and Science, West Hartford, Connecticut
| | - Lidiya Kukova
- Program of Applied Translational Research, Department of Medicine and
| | - Yaqi Jia
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Petter Bjornstad
- Division of Endocrinology, Department of Pediatrics and.,Division of Nephrology, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Joe M El-Khoury
- Program of Applied Translational Research, Department of Medicine and
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland;
| |
Collapse
|
35
|
Ortega-Loubon C, Fierro I, Tamayo E. Should kidney oxygen saturation be monitored to prevent acute kidney injury after adult cardiac surgery? J Thorac Cardiovasc Surg 2019; 158:e105-e106. [PMID: 31208801 DOI: 10.1016/j.jtcvs.2019.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 02/01/2023]
Affiliation(s)
| | - Inmaculada Fierro
- Department of Health Science, Universidad Europea Miguel de Cervantes, Valladolid, Spain
| | - Eduardo Tamayo
- Anesthesiology and Critical Care, Clinic University Hospital of Valladolid, Valladolid, Spain
| |
Collapse
|
36
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Mesenchymal stem cell therapy targeting mitochondrial dysfunction in acute kidney injury. J Transl Med 2019; 17:142. [PMID: 31046805 PMCID: PMC6498508 DOI: 10.1186/s12967-019-1893-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/25/2019] [Indexed: 12/13/2022] Open
Abstract
Mitochondria take part in a network of cellular processes that regulate cell homeostasis. Defects in mitochondrial function are key pathophysiological changes during acute kidney injury (AKI). Mesenchymal stem cells (MSCs) have shown promising regenerative effects in experimental AKI models, but the specific mechanism is still unclear. Some studies have demonstrated that MSCs are able to target mitochondrial dysfunction during AKI. In this review, we summarize these articles, providing an integral and updated view of MSC therapy targeting mitochondrial dysfunction during AKI, which is aimed at promoting the therapeutic effect of MSCs in AKI patients.
Collapse
Affiliation(s)
- Lingfei Zhao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ping Zhang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hua Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China. .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
37
|
Xiang L, Thompson MS, Clemmer JS, Mittwede PN, Khan T, Hester RL. Early treatment with GLP-1 after severe trauma preserves renal function in obese Zucker rats. Am J Physiol Regul Integr Comp Physiol 2019; 316:R621-R627. [PMID: 30811247 DOI: 10.1152/ajpregu.00312.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Early posttrauma hyperglycemia (EPTH) is correlated with later adverse outcomes, including acute kidney injury (AKI). Controlling EPTH in the prehospital setting is difficult because of the variability in the ideal insulin dosage and the potential risk of hypoglycemia, especially in those with confounding medical comorbidities of obesity and insulin resistance. Glucagon-like peptide-1 (GLP-1) controls glucose levels in a glucose-dependent manner and is a current target in antidiabetic therapy. We have shown that after orthopedic trauma, obese Zucker rats exhibit EPTH and a later development of AKI (within 24 h). We hypothesized that GLP-1 treatment after trauma decreases EPTH and protects renal function in obese Zucker rats. Obese Zucker rats (~12 wk old) were fasted for 4 h before trauma. Soft tissue injury, fibula fracture, and homogenized bone component injection were then performed in both hind limbs to induce severe extremity trauma. Plasma glucose levels were measured before and 15, 30, 60, 120, 180, 240, and 300 min after trauma. GLP-1 (3 μg·kg-1·h-1, 1.5 ml/kg total) or saline was continuously infused from 30 min to 5 h after trauma. Afterwards, rats were placed in metabolic cages overnight for urine collection. The following day, plasma interleukin (IL)-6 levels, renal blood flow (RBF), glomerular filtration rate (GFR), and renal oxygen delivery (Do2) and consumption (V̇o2) were measured. EPTH was evident within 15 min after trauma but was significantly ameliorated during the 5 h of GLP-1 infusion. One day after trauma, plasma IL-6 was markedly increased in the trauma group and decreased in GLP-1-treated animals. RBF, GFR, and Do2 all significantly decreased with trauma, but renal V̇o2 was unchanged. GLP-1 treatment normalized RBF, GFR, and Do2 without affecting V̇o2. These results suggest that GLP-1 decreases EPTH and protects against a later development of AKI. Early treatment with GLP-1 (or its analogs) to rapidly, effectively, and safely control EPTH may be beneficial in the prehospital care of obese patients after trauma.
Collapse
Affiliation(s)
- Lusha Xiang
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,United States Army Institute of Surgical Research , San Antonio, Texas
| | - Michael S Thompson
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - John S Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Peter N Mittwede
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| | - Tazim Khan
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Robert L Hester
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
38
|
Iguchi N, Kosaka J, Booth LC, Iguchi Y, Evans RG, Bellomo R, May CN, Lankadeva YR. Renal perfusion, oxygenation, and sympathetic nerve activity during volatile or intravenous general anaesthesia in sheep. Br J Anaesth 2019; 122:342-349. [DOI: 10.1016/j.bja.2018.11.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/22/2022] Open
|
39
|
Keeley TP, Mann GE. Defining Physiological Normoxia for Improved Translation of Cell Physiology to Animal Models and Humans. Physiol Rev 2019; 99:161-234. [PMID: 30354965 DOI: 10.1152/physrev.00041.2017] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extensive oxygen gradient between the air we breathe (Po2 ~21 kPa) and its ultimate distribution within mitochondria (as low as ~0.5-1 kPa) is testament to the efforts expended in limiting its inherent toxicity. It has long been recognized that cell culture undertaken under room air conditions falls short of replicating this protection in vitro. Despite this, difficulty in accurately determining the appropriate O2 levels in which to culture cells, coupled with a lack of the technology to replicate and maintain a physiological O2 environment in vitro, has hindered addressing this issue thus far. In this review, we aim to address the current understanding of tissue Po2 distribution in vivo and summarize the attempts made to replicate these conditions in vitro. The state-of-the-art techniques employed to accurately determine O2 levels, as well as the issues associated with reproducing physiological O2 levels in vitro, are also critically reviewed. We aim to provide the framework for researchers to undertake cell culture under O2 levels relevant to specific tissues and organs. We envisage that this review will facilitate a paradigm shift, enabling translation of findings under physiological conditions in vitro to disease pathology and the design of novel therapeutics.
Collapse
Affiliation(s)
- Thomas P Keeley
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| |
Collapse
|
40
|
O'Neill J, Jasionek G, Drummond SE, Brett O, Lucking EF, Abdulla MA, O'Halloran KD. Renal cortical oxygen tension is decreased following exposure to long-term but not short-term intermittent hypoxia in the rat. Am J Physiol Renal Physiol 2019; 316:F635-F645. [PMID: 30648908 DOI: 10.1152/ajprenal.00254.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic kidney disease (CKD) occurs in more than 50% of patients with obstructive sleep apnea (OSA). However, the impact of intermittent hypoxia (IH) on renal function and oxygen homeostasis is unclear. Male Sprague-Dawley rats were exposed to IH (270 s at 21% O2; 90 s hypoxia, 6.5% O2 at nadir) for 4 h [acute IH (AIH)] or to chronic IH (CIH) for 8 h/day for 2 wk. Animals were anesthetized and surgically prepared for the measurement of mean arterial pressure (MAP), and left renal excretory function, renal blood flow (RBF), and renal oxygen tension (Po2). AIH had no effect on MAP (123 ± 14 vs. 129 ± 14 mmHg, means ± SE, sham vs. IH). The CIH group was hypertensive (122 ± 9 vs. 144 ± 15 mmHg, P < 0.05). Glomerular filtration rate (GFR) (0.92 ± 0.27 vs. 1.33 ± 0.33 ml/min), RBF (3.8 ± 1.5 vs. 7.2 ± 2.4 ml/min), and transported sodium (TNa) (132 ± 39 vs. 201 ± 47 μmol/min) were increased in the AIH group (all P < 0.05). In the CIH group, GFR (1.25 ± 0.28 vs. 0.86 ± 0.28 ml/min, P < 0.05) and TNa (160 ± 39 vs. 120 ± 40 μmol/min, P < 0.05) were decreased, while RBF (4.13 ± 1.5 vs. 3.08 ± 1.5 ml/min) was not significantly different. Oxygen consumption (QO2) was increased in the AIH group (6.76 ± 2.60 vs. 13.60 ± 7.77 μmol/min, P < 0.05), but it was not significantly altered in the CIH group (3.97 ± 2.63 vs. 6.82 ± 3.29 μmol/min). Cortical Po2 was not significantly different in the AIH group (46 ± 4 vs. 46 ± 3 mmHg), but it was decreased in the CIH group (44 ± 5 mmHg vs. 38 ± 2 mmHg, P < 0.05). For AIH, renal oxygen homeostasis was preserved through a maintained balance between O2 supply (RBF) and consumption (GFR). For CIH, mismatched TNa and QO2 reflect inefficient O2 utilization and, thereby, sustained decrease in cortical Po2.
Collapse
Affiliation(s)
- Julie O'Neill
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Greg Jasionek
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Sarah E Drummond
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Orla Brett
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Mohammed A Abdulla
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork , Cork , Ireland
| |
Collapse
|
41
|
Lannemyr L, Ricksten S, Rundqvist B, Andersson B, Bartfay S, Ljungman C, Dahlberg P, Bergh N, Hjalmarsson C, Gilljam T, Bollano E, Karason K. Differential Effects of Levosimendan and Dobutamine on Glomerular Filtration Rate in Patients With Heart Failure and Renal Impairment:A Randomized Double-Blind Controlled Trial. J Am Heart Assoc 2018; 7:e008455. [PMID: 30369310 PMCID: PMC6201411 DOI: 10.1161/jaha.117.008455] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/02/2018] [Indexed: 01/07/2023]
Abstract
Background The management of the cardiorenal syndrome in advanced heart failure is challenging, and the role of inotropic drugs has not been fully defined. Our aim was to compare the renal effects of levosimendan versus dobutamine in patients with heart failure and renal impairment. Methods and Results In a randomized double-blind study, we assigned patients with chronic heart failure (left ventricular ejection fraction <40%) and impaired renal function (glomerular filtration rate <80 mL/min per 1.73 m2) to receive either levosimendan (loading dose 12 μg/kg+0.1 μg/kg per minute) or dobutamine (7.5 μg/kg per minute) for 75 minutes. A pulmonary artery catheter was used for measurements of systemic hemodynamics, and a renal vein catheter was used to measure renal plasma flow by the infusion clearance technique for PAH (para-aminohippurate) corrected by renal extraction of PAH . Filtration fraction was measured by renal extraction of chromium ethylenediamine tetraacetic acid. A total of 32 patients completed the study. Following treatment, the levosimendan and dobutamine groups displayed similar increases in renal blood flow (22% and 26%, respectively) with no significant differences between groups. Glomerular filtration rate increased by 22% in the levosimendan group but remained unchanged in the dobutamine group ( P=0.012). Filtration fraction was not affected by levosimendan but decreased by 17% with dobutamine ( P=0.045). Conclusions In patients with chronic heart failure and renal impairment, levosimendan increases glomerular filtration rate to a greater extent than dobutamine and thus may be the preferred inotropic agent for treating patients with the cardiorenal syndrome. Clinical Trial Registration URL: https://www.clinicaltrials.gov . Unique identifier: NCT 02133105.
Collapse
Affiliation(s)
- Lukas Lannemyr
- Department of Anesthesiology and Intensive Care MedicineUniversity of GothenburgSahlgrenska UniversityGothenburgSweden
| | - Sven‐Erik Ricksten
- Department of Anesthesiology and Intensive Care MedicineUniversity of GothenburgSahlgrenska UniversityGothenburgSweden
| | - Bengt Rundqvist
- Department of TransplantationSahlgrenska University HospitalGothenburgSweden
| | - Bert Andersson
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Sven‐Erik Bartfay
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | | | - Pia Dahlberg
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Niklas Bergh
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Clara Hjalmarsson
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Thomas Gilljam
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Entela Bollano
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Kristjan Karason
- Department of TransplantationSahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
42
|
Leow EH, Chan YH, Ng YH, Lim JKB, Nakao M, Lee JH. Prevention of Acute Kidney Injury in Children Undergoing Cardiac Surgery: A Narrative Review. World J Pediatr Congenit Heart Surg 2018; 9:79-90. [PMID: 29310552 DOI: 10.1177/2150135117743211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Children undergoing cardiac surgery are at risk of developing acute kidney injury (AKI). Preventing cardiac surgery-associated AKI (CS-AKI) is important as it is associated with increased early- and long-term mortality and morbidity. Targeting modifiable risk factors (eg, avoiding poor renal perfusion, nephrotoxic drugs, and fluid overload) reduces the risk of CS-AKI. There is currently no strong evidence for the routine use of pharmacological approaches (eg, aminophylline, dexmedetomidine, fenoldopam, and steroids) to prevent CS-AKI. There is robust evidence to support the role of early peritoneal dialysis as a nonpharmacologic approach to prevent CS-AKI.
Collapse
Affiliation(s)
- Esther Huimin Leow
- 1 Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yoke Hwee Chan
- 2 Children's Intensive Care Unit, KK Women's and Children's Hospital, Singapore, Singapore.,3 Duke-NUS School of Medicine, Singapore, Singapore
| | - Yong Hong Ng
- 1 Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Joel Kian Boon Lim
- 1 Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Masakazu Nakao
- 4 Department of Paediatric Cardiothoracic Surgery, KK Women's and Children's Hospital, Singapore, Singapore
| | - Jan Hau Lee
- 2 Children's Intensive Care Unit, KK Women's and Children's Hospital, Singapore, Singapore.,3 Duke-NUS School of Medicine, Singapore, Singapore
| |
Collapse
|
43
|
Olesen ND, Jørgensen TB, Eiberg J, Helgstrand UJV, Sillesen HH, Cedergreen P, Secher NH, Nielsen HB. Elevated Renal Oxygen Extraction During Open Abdominal Aortic Aneurysm Repair Is Related to Postoperative Renal Dysfunction. Semin Cardiothorac Vasc Anesth 2018; 22:369-375. [PMID: 30047299 DOI: 10.1177/1089253218790270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Open abdominal aortic aneurysm repair is often followed by elevated plasma creatinine, likely due to impaired renal blood flow. We evaluated whether postoperative elevation in creatinine relates to renal oxygen extraction during surgery as an index of renal blood flow and also monitored frontal lobe oxygenation. METHODS For 19 patients (66 ± 10 years; mean ± SD) undergoing open infrarenal abdominal aortic aneurysm repair, renal oxygen extraction was determined by arterial and renal vein catheterization. Near-infrared spectroscopy determined frontal lobe oxygenation. RESULTS During surgery mean arterial pressure (from 102 ± 14 to 65 ± 11 mm Hg; P < .0001), arterial hemoglobin (from 7.7 ± 0.7 to 6.6 ± 0.8 mmol/L; P < 0.0001), and frontal lobe oxygenation (from 74 ± 6% to 70 ± 6%; P = .0414) decreased, while renal oxygen extraction increased (from 5.3% [4.3-8.1]; median [interquartile range] to 10.8% [5.8-17.5]; P = .0405). Plasma creatinine became significantly elevated on the second day after the operation (from 83 [73-101] to 105 µmol/L [79-143]; P = .0062) with a peak increase observed after 2 days (1-2). The peak increase in creatinine correlated to intraoperative renal oxygen extraction ( r = 0.51; P = .026). CONCLUSION Kidney function was affected after open abdominal aortic aneurysm repair likely related to limited renal blood flow. We take the increase in renal oxygen extraction and reduction in frontal lobe oxygenation to suggest that mean arterial pressure and hemoglobin were too low to maintain renal and cerebral circulation in vascular surgical patients.
Collapse
|
44
|
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: 68] [Impact Index Per Article: 9.7] [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
|
45
|
Renal Oxygen Flux during Cardiopulmonary Bypass; Tubular Damage to Preserve Glomerular Filtration-What's a Kidney to Do? Anesthesiology 2017; 126:199-201. [PMID: 27906704 DOI: 10.1097/aln.0000000000001462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Cao W, Cui S, Yang L, Wu C, Liu J, Yang F, Liu Y, Bin J, Hou FF. Contrast-Enhanced Ultrasound for Assessing Renal Perfusion Impairment and Predicting Acute Kidney Injury to Chronic Kidney Disease Progression. Antioxid Redox Signal 2017; 27:1397-1411. [PMID: 28715949 DOI: 10.1089/ars.2017.7006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AIMS Acute kidney injury (AKI) is increasingly recognized as a major risk factor leading to progression to chronic kidney disease (CKD). However, the diagnostic tools for predicting AKI to CKD progression are particularly lacking. Here, we tested the utility of contrast-enhanced ultrasound (CEUS) for predicting progression to CKD after AKI by using both mild (20-min) and severe (45-min) bilateral renal ischemia-reperfusion injury mice. RESULTS Renal perfusion measured by CEUS reduced to 25% ± 7% and 14% ± 6% of the pre-ischemic levels in mild and severe AKI 1 h after ischemia (p < 0.05). Renal perfusion returned to pre-ischemic levels 1 day after mild AKI followed by restoration of kidney function. However, severe AKI caused persistent renal perfusion impairment (60% ± 9% of baseline levels) accompanied by progressive renal fibrosis and sustained decrease in renal function. Renal perfusion at days 1-21 significantly correlated with tubulointerstitial fibrosis 42 days after AKI. For predicting renal fibrosis at day 42, the area under the receiver operating characteristics curve of renal perfusion impairment at day 1 was 0.84. Similar changes in the renal image of CEUS were observed in patients with AKI-CKD progression. INNOVATION This study demonstrates that CEUS enables dynamic and noninvasive detection of renal perfusion impairment after ischemic AKI and the perfusion abnormalities shown by CEUS can early predict the progression to CKD after AKI. CONCLUSIONS These results indicate that CEUS enables the evaluation of renal perfusion impairment associated with CKD after ischemic AKI and may serve as a noninvasive technique for assessing AKI-CKD progression. Antioxid. Redox Signal. 27, 1397-1411.
Collapse
Affiliation(s)
- Wei Cao
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University , Guangzhou, P.R. China
| | - Shuang Cui
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University , Guangzhou, P.R. China
| | - Li Yang
- 2 Division of Pharmacology, Nanfang Hospital , Southern Medical University, Guangzhou, P.R. China
| | - Chunyi Wu
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University , Guangzhou, P.R. China
| | - Jian Liu
- 3 Division of Cardiology, Nanfang Hospital , Southern Medical University, Guangzhou, P.R. China
| | - Fang Yang
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University , Guangzhou, P.R. China
| | - Youhua Liu
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University , Guangzhou, P.R. China
| | - Jianping Bin
- 3 Division of Cardiology, Nanfang Hospital , Southern Medical University, Guangzhou, P.R. China
| | - Fan Fan Hou
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University , Guangzhou, P.R. China
| |
Collapse
|
47
|
Pavlakou P, Liakopoulos V, Eleftheriadis T, Mitsis M, Dounousi E. Oxidative Stress and Acute Kidney Injury in Critical Illness: Pathophysiologic Mechanisms-Biomarkers-Interventions, and Future Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6193694. [PMID: 29104728 PMCID: PMC5637835 DOI: 10.1155/2017/6193694] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/09/2017] [Accepted: 08/20/2017] [Indexed: 01/22/2023]
Abstract
Acute kidney injury (AKI) is a multifactorial entity that occurs in a variety of clinical settings. Although AKI is not a usual reason for intensive care unit (ICU) admission, it often complicates critically ill patients' clinical course requiring renal replacement therapy progressing sometimes to end-stage renal disease and increasing mortality. The causes of AKI in the group of ICU patients are further complicated from damaged metabolic state, systemic inflammation, sepsis, and hemodynamic dysregulations, leading to an imbalance that generates oxidative stress response. Abundant experimental and to a less extent clinical data support the important role of oxidative stress-related mechanisms in the injury phase of AKI. The purpose of this article is to present the main pathophysiologic mechanisms of AKI in ICU patients focusing on the different aspects of oxidative stress generation, the available evidence of interventional measures for AKI prevention, biomarkers used in a clinical setting, and future perspectives in oxidative stress regulation.
Collapse
Affiliation(s)
- Paraskevi Pavlakou
- Department of Nephrology, Medical School University of Ioannina, Ioannina, Greece
| | - Vassilios Liakopoulos
- Division of nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Michael Mitsis
- Department of Surgery, Medical School University of Ioannina, Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, Medical School University of Ioannina, Ioannina, Greece
| |
Collapse
|
48
|
Cardiac Surgery-Associated Acute Kidney Injury. CURRENT ANESTHESIOLOGY REPORTS 2017. [DOI: 10.1007/s40140-017-0224-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
49
|
Hertzberg D, Rydén L, Pickering JW, Sartipy U, Holzmann MJ. Acute kidney injury-an overview of diagnostic methods and clinical management. Clin Kidney J 2017; 10:323-331. [PMID: 28616210 PMCID: PMC5466115 DOI: 10.1093/ckj/sfx003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 01/13/2017] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is a common condition in multiple clinical settings. Patients with AKI are at an increased risk of death, over both the short and long term, and of accelerated renal impairment. As the condition has become more recognized and definitions more unified, there has been a rapid increase in studies examining AKI across many different clinical settings. This review focuses on the classification, diagnostic methods and clinical management that are available, or promising, for patients with AKI. Furthermore, preventive measures with fluids, acetylcysteine, statins and remote ischemic preconditioning, as well as when dialysis should be initiated in AKI patients are discussed. The classification of AKI includes both changes in serum creatinine concentrations and urine output. Currently, no kidney injury biomarkers are included in the classification of AKI, but proposals have been made to include them as independent diagnostic markers. Treatment of AKI is aimed at addressing the underlying causes of AKI, and at limiting damage and preventing progression. The key principles are: to treat the underlying disease, to optimize fluid balance and optimize hemodynamics, to treat electrolyte disturbances, to discontinue or dose-adjust nephrotoxic drugs and to dose-adjust drugs with renal elimination.
Collapse
Affiliation(s)
- Daniel Hertzberg
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Linda Rydén
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Anesthesiology, Surgical Services and Intensive Care Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - John W. Pickering
- Deparment of Medicine, University of Otago Christchurch and Emergency Department, Christchurch Hospital, Christchurch, New Zealand
| | - Ulrik Sartipy
- Section of Cardiothoracic Surgery, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Martin J. Holzmann
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Emergency Medicine, Huddinge, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
50
|
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
|