1
|
Navaratnam M, Li EX, Chen S, Margetson T, Wolke O, Ma M, Ebel NH, Bonham CA, Ramamoorthy C. Perioperative Management of Pediatric Combined Heart and Liver Transplantation: A 17 year single center experience. Paediatr Anaesth 2024; 34:1130-1138. [PMID: 39115452 DOI: 10.1111/pan.14977] [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: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND An increasing number of centers are undertaking combined heart and liver transplantation in adult and pediatric patients with congenital heart disease. AIM The primary aim of this study was to describe the perioperative management of a single center cohort, identifying challenges and potential solutions. METHODS We conducted a retrospective review of all patients undergoing combined heart and liver transplantation at Stanford Children's Hospital from 2006 to 2022. Preoperative information included cardiac diagnosis, hemodynamics, and severity of liver disease. Intraoperative data included length of surgery, cardiopulmonary bypass time, and blood products transfused. Postoperative data included blood products transfused in the intensive care unit, time to extubation, length of intensive care unit stay, survival outcomes and 30-day adverse events. RESULTS Eighteen patients underwent en bloc combined heart and liver transplantation at Stanford Children's Hospital from 2006 to 2022, and the majority 15 (83%) were transplanted for failing Fontan circulation with Fontan Associated Liver Disease. Median surgical procedure time was 13.4 [11.5, 14.5] h with a cardiopulmonary bypass time of 4.3 [3.9, 5.8] h. Median total blood products transfused in the operating room post cardiopulmonary bypass was 89.4 [63.9, 127.0] mLs/kg. Nine patients (50%) had vasoplegia during cardiopulmonary bypass. Activated prothrombin complex concentrates were used post cardiopulmonary bypass in 15 (83%) patients with a 30-day thromboembolism rate of 22%. Median time to extubation was 4.0 [2.8, 6.5] days, median intensive care unit length of stay 20.0 [7.8, 48.3] days and median hospital length of stay 54.0 [30.5, 68.3] days. Incidence of renal replacement therapy was 11%; however, none required renal replacement therapy by the time of hospital discharge. Neurological events within 30 days were 17% and the 30 day and 1 year survival was 89%. CONCLUSIONS Perioperative challenges include major perioperative bleeding, unstable hemodynamics, and end organ injury including acute kidney injury and neurological events. Successful outcomes for en bloc combined heart and liver transplantation are possible with careful multidisciplinary planning, communication, patient selection, and integrated peri-operative management.
Collapse
Affiliation(s)
- Manchula Navaratnam
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| | - Emma Xi Li
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| | - Sharon Chen
- Department of Pediatrics, Division of Cardiology, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| | - Tristan Margetson
- Department of Cardiovascular Perfusion, Stanford Hospital and Clinic, Stanford Children's Hospital, Palo Alto, California, USA
| | - Olga Wolke
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| | - Michael Ma
- Department of Cardiothoracic Surgery, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| | - Noelle H Ebel
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| | - C Andrew Bonham
- Department of Abdominal Transplantation Surgery, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| | - Chandra Ramamoorthy
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Children's Hospital, Stanford University Medical Center, Palo Alto, California, USA
| |
Collapse
|
2
|
Pasten C, Lozano M, Osorio LA, Cisterna M, Jara V, Sepúlveda C, Ramírez-Balaguera D, Moreno-Hidalgo V, Arévalo-Gil D, Soto P, Hurtado V, Morales A, Méndez GP, Busso D, Leon P, Michea L, Corvalán D, Luarte A, Irarrazabal CE. The protective effect of 1400W against ischaemia and reperfusion injury is countered by transient medullary kidney endothelial dysregulation. J Physiol 2024. [PMID: 39057844 DOI: 10.1113/jp285944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
Renal ischaemia and reperfusion (I/R) is caused by a sudden temporary impairment of the blood flow. I/R is a prevalent cause of acute kidney injury. As nitric oxide generated by inducible nitric oxide synthase (iNOS) has detrimental effects during I/R, the pharmacological blockade of iNOS has been proposed as a potential strategy to prevent I/R injury. The aim of this study was to improve the understanding of 1400W (an iNOS inhibitor) on renal I/R as a pharmacological strategy against kidney disease. BALB/c mice received 30 min of bilateral ischaemia, followed by 48 h or 28 days of reperfusion. Vehicle or 1400W (10 mg/kg) was administered 30 min before inducing ischaemia. We found that after 48 h of reperfusion 1400W decreased the serum creatinine, blood urea nitrogen, neutrophil gelatinase-associated lipocalin and proliferating cell nuclear antigen 3 in the I/R animals. Unexpectedly, we observed mRNA upregulation of genes involved in kidney injury, cell-cycle arrest, inflammation, mesenchymal transition and endothelial activation in the renal medulla of sham animals treated with 1400W. We also explored if 1400W promoted chronic kidney dysfunction 28 days after I/R and did not find significant alterations in renal function, fibrosis, blood pressure or mortality. The results provide evidence that 1400W may have adverse effects in the renal medulla. Importantly, our data point to 1400W-induced endothelial dysfunction, establishing therapeutic limitations for its use. KEY POINTS: Acute kidney injury is a global health problem associated with high morbidity and mortality. The pharmacological blockade of inducible nitric oxide synthase (iNOS) has been proposed as a potential strategy to prevent AKI induced by ischaemia and reperfusion (I/R). Our main finding is that 1400W, a selective and irreversible iNOS inhibitor with low toxicity that is proposed as a therapeutic strategy to prevent kidney I/R injury, produces aberrant gene expression in the medulla associated to tissue injury, cell cycle arrest, inflammation, mesenchymal transition and endothelial activation. The negative effect of 1400W observed in the renal medulla at 48 h from drug administration, is transient as it did not translate into a chronic kidney disease condition.
Collapse
Affiliation(s)
- Consuelo Pasten
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
- Facultad de Medicina, Universidad de los Andes, Chile
| | - Mauricio Lozano
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Luis A Osorio
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Matías Cisterna
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Valeria Jara
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Catalina Sepúlveda
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Daniela Ramírez-Balaguera
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Viviana Moreno-Hidalgo
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Dayana Arévalo-Gil
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Paola Soto
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Valeria Hurtado
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | - Antonia Morales
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
| | | | - Dolores Busso
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Biología de la Reproducción, Universidad de los Andes, Chile
| | - Pablo Leon
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Michea
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Daniela Corvalán
- Neuroscience Program, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Universidad de los Andes, Chile
| | - Alejandro Luarte
- Neuroscience Program, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Universidad de los Andes, Chile
| | - Carlos E Irarrazabal
- Centro de Investigación e Innovación Biomédica (CiiB), Programa de Fisiología, Laboratorio de Fisiología Integrativa y Molecular, Universidad de los Andes, Chile
- Facultad de Medicina, Universidad de los Andes, Chile
| |
Collapse
|
3
|
Hulst AH, Ow CPC, May CN, Hood SH, Plummer MP, Hermanides J, van Raalte DH, Deane AM, Bellomo R, Lankadeva YR. Effects of sodium-glucose transporter-2 inhibition on systemic hemodynamics, renal function, and intra-renal oxygenation in sepsis-associated acute kidney injury. Intensive Care Med Exp 2024; 12:64. [PMID: 38977627 PMCID: PMC11231125 DOI: 10.1186/s40635-024-00647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND People with type 2 diabetes mellitus treated with sodium-glucose transporter-2 inhibitors (SGLT2i) have lower rates of acute kidney injury (AKI). Sepsis is responsible for the majority of AKI in critically ill patients. This study investigated whether SGLT2i is renoprotective in an ovine model of Gram-negative septic AKI. METHODS Sixteen healthy merino ewes were surgically instrumented to enable measurement of mean arterial pressure, cardiac output, renal blood flow, renal cortical and medullary perfusion, and oxygenation. After a 5-day recovery period, sepsis was induced via slow and continuous intravenous infusion of live Escherichia coli. Twenty-three hours later, sheep were randomized to receive an intravenous bolus of 0.2 mg/kg empagliflozin (n = 8) or a fluid-matched vehicle (n = 8). RESULTS Empagliflozin treatment did not significantly reduce renal medullary hypoperfusion or hypoxia, improve kidney function, or induce histological changes. Renal cortical oxygenation during the intervention period was 47.6 ± 5.9 mmHg in the empagliflozin group compared with 40.6 ± 8.2 mmHg in the placebo group (P = 0.16). Renal medullary oxygenation was 28.0 ± 18.5 mmHg in the empagliflozin compared with 25.7 ± 16.3 mmHg (P = 0.82). Empagliflozin treatment did not result in significant between-group differences in renal blood flow, kidney function, or renal histopathological changes. CONCLUSION In a large mammalian model of septic AKI, a single dose of empagliflozin did not improve renal microcirculatory perfusion, oxygenation, kidney function, or histopathology.
Collapse
Affiliation(s)
- Abraham H Hulst
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Connie P C Ow
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Clive N May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Critical Care, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
| | - Sally H Hood
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Mark P Plummer
- Department of Intensive Care, Royal Adelaide Hospital, Adelaide, Australia
| | - Jeroen Hermanides
- Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Adam M Deane
- Department of Intensive Care, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Rinaldo Bellomo
- Department of Intensive Care, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Critical Care, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, VIC, Australia
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
| | - Yugeesh R Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Critical Care, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
- Department of Anesthesia, Austin Hospital, Melbourne, VIC, Australia
| |
Collapse
|
4
|
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
|
5
|
Hillaert A, Sanmiguel Serpa LC, Xu Y, Hesta M, Bogaert S, Vanderperren K, Pullens P. Optimization of Fair Arterial Spin Labeling Magnetic Resonance Imaging (ASL-MRI) for Renal Perfusion Quantification in Dogs: Pilot Study. Animals (Basel) 2024; 14:1810. [PMID: 38929429 PMCID: PMC11201026 DOI: 10.3390/ani14121810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Arterial spin labeling (ASL) MRI allows non-invasive quantification of renal blood flow (RBF) and shows great potential for renal assessment. To our knowledge, renal ASL-MRI has not previously been performed in dogs. The aim of this pilot study was to determine parameters essential for ALS-MRI-based quantification of RBF in dogs: T1, blood (longitudinal relaxation time), λ (blood tissue partition coefficient) and TI (inversion time). A Beagle was scanned at 3T with a multi-TI ASL sequence, with TIs ranging from 250 to 2500 ms, to determine the optimal TI value. The T1 of blood for dogs was determined by scanning a blood sample with a 2D IR TSE sequence. The water content of the dog's kidney was determined by analyzing kidney samples from four dogs with a moisture analyzer and was subsequently used to calculate λ. The optimal TI and the measured values for T1,blood, and λ were 2000 ms, 1463 ms and 0.91 mL/g, respectively. These optimized parameters for dogs resulted in lower RBF values than those obtained from inline generated RBF maps. In conclusion, this study determined preliminary parameters essential for ALS-MRI-based RBF quantification in dogs. Further research is needed to confirm these values, but it may help guide future research.
Collapse
Affiliation(s)
- Amber Hillaert
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (A.H.)
| | - Luis Carlos Sanmiguel Serpa
- Department of Medical Imaging, Ghent University Hospital, 9000 Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, 9000 Ghent, Belgium
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Yangfeng Xu
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (A.H.)
| | - Myriam Hesta
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (A.H.)
| | - Stephanie Bogaert
- Department of Medical Imaging, Ghent University Hospital, 9000 Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, 9000 Ghent, Belgium
| | - Katrien Vanderperren
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; (A.H.)
| | - Pim Pullens
- Department of Medical Imaging, Ghent University Hospital, 9000 Ghent, Belgium
- Ghent Institute for Functional and Metabolic Imaging, Ghent University, 9000 Ghent, Belgium
- Institute of Biomedical Engineering and Technology (IBiTech)—MEDISP, Faculty of Engineering and Architecture, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
6
|
Iba T, Helms J, Maier CL, Levi M, Scarlatescu E, Levy JH. The role of thromboinflammation in acute kidney injury among patients with septic coagulopathy. J Thromb Haemost 2024; 22:1530-1540. [PMID: 38382739 DOI: 10.1016/j.jtha.2024.02.006] [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: 12/01/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
Inflammation and coagulation are critical self-defense mechanisms for mitigating infection that can nonetheless induce tissue injury and organ dysfunction. In severe cases, like sepsis, a dysregulated thromboinflammatory response may result in multiorgan dysfunction. Sepsis-associated acute kidney injury (AKI) is a significant contributor to patient morbidity and mortality. The connection between AKI and thromboinflammation is largely due to unique aspects of the renal vasculature. Specifically, the interaction between blood cells with the endothelial, glomerular, and peritubular capillary systems during thromboinflammation reduces oxygen supply to tubular epithelial cells. Previous studies have focused on tubular epithelial cell damage due to hypoxia, oxidative stress, and nephrotoxins. Although these factors are pivotal in acute tubular injury or necrosis, recent studies have demonstrated that AKI in sepsis encompasses a mixture of tubular and glomerular damage subtypes. In cases of sepsis-induced coagulopathy, thromboinflammation within the glomerulus and peritubular capillaries is an important pathogenic mechanism for AKI. Unfortunately, and despite the use of renal replacement therapy, the development of AKI in sepsis continues to be associated with high morbidity, mortality, and clinical challenges requiring alternative approaches. This review introduces the important role of thromboinflammation in AKI pathogenesis and details innovative vascular-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Julie Helms
- French National Institute of Health and Medical Research, United Medical Resources 1260, Regenerative Nanomedicine, Federation de Medicine Translationnelle de Strasbourg, Strasbourg University Hospital, Medical Intensive Care Unit - NHC, Strasbourg University, Strasbourg, France
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands; Department of Medicine, University College London Hospitals National Health Service Foundation Trust, Cardio-metabolic Programme-National Institute for Health and Care Research University College London Hospitals/University College London Biomedical Research Centre, London, United Kingdom
| | - Ecaterina Scarlatescu
- University of Medicine and Pharmacy "Carol Davila," Bucharest, Romania; Department of Anaesthesia and Intensive Care, Fundeni Clinical Institute, Bucharest, Romania
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
7
|
Fanous MS, de la Cruz JE, Michael OS, Afolabi JM, Kumar R, Adebiyi A. EARLY FLUID PLUS NOREPINEPHRINE RESUSCITATION DIMINISHES KIDNEY HYPOPERFUSION AND INFLAMMATION IN SEPTIC NEWBORN PIGS. Shock 2024; 61:885-893. [PMID: 38662580 PMCID: PMC11251746 DOI: 10.1097/shk.0000000000002343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
ABSTRACT Sepsis is the most frequent risk factor for acute kidney injury (AKI) in critically ill infants. Sepsis-induced dysregulation of kidney microcirculation in newborns is unresolved. The objective of this study was to use the translational swine model to evaluate changes in kidney function during the early phase of sepsis in newborns and the impact of fluid plus norepinephrine resuscitation. Newborn pigs (3-7-day-old) were allocated randomly to three groups: 1) sham, 2) sepsis (cecal ligation and puncture) without subsequent resuscitation, and 3) sepsis with lactated Ringer plus norepinephrine resuscitation. All animals underwent standard anesthesia and mechanical ventilation. Cardiac output and glomerular filtration rate were measured noninvasively. Mean arterial pressure, total renal blood flow, cortical perfusion, medullary perfusion, and medullary tissue oxygen tension (mtPO 2 ) were determined for 12 h. Cecal ligation and puncture decreased mean arterial pressure and cardiac output by more than 50%, with a proportional increase in renal vascular resistance and a 60-80% reduction in renal blood flow, cortical perfusion, medullary perfusion, and mtPO 2 compared to sham. Cecal ligation and puncture also decreased glomerular filtration rate by ~79% and increased AKI biomarkers. Isolated foci of tubular necrosis were observed in the septic piglets. Except for mtPO 2 , changes in all these parameters were ameliorated in resuscitated piglets. Resuscitation also attenuated sepsis-induced increases in the levels of plasma C-reactive protein, proinflammatory cytokines, lactate dehydrogenase, alanine transaminase, aspartate aminotransferase, and renal NLRP3 inflammasome. These data suggest that newborn pigs subjected to cecal ligation and puncture develop hypodynamic septic AKI. Early implementation of resuscitation lessens the degree of inflammation, AKI, and liver injury.
Collapse
Affiliation(s)
- Mina S. Fanous
- Stormont Vail Pediatric Critical Care, Topeka, Kansas
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
| | - Julia E. de la Cruz
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Olugbenga S. Michael
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jeremiah M. Afolabi
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
| | - Ravi Kumar
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Adebowale Adebiyi
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
8
|
Wang JY, Song QL, Wang YL, Jiang ZM. Urinary oxygen tension and its role in predicting acute kidney injury: A narrative review. J Clin Anesth 2024; 93:111359. [PMID: 38061226 DOI: 10.1016/j.jclinane.2023.111359] [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: 04/22/2023] [Revised: 11/12/2023] [Accepted: 12/01/2023] [Indexed: 01/14/2024]
Abstract
Acute kidney injury occurs frequently in the perioperative setting. The renal medulla often endures hypoxia or hypoperfusion and is susceptible to the imbalance between oxygen supply and demand due to the nature of renal blood flow distribution and metabolic rate in the kidney. The current available evidence demonstrated that the urine oxygen pressure is proportional to the variations of renal medullary tissue oxygen pressure. Thus, urine oxygenation can be a candidate for reflecting the change of oxygen in the renal medulla. In this review, we discuss the basic physiology of acute kidney injury, as well as techniques for monitoring urine oxygen tension, confounding factors affecting the reliable measurement of urine oxygen tension, and its clinical use, highlighting its potential role in early detection and prevention of acute kidney injury.
Collapse
Affiliation(s)
- Jing-Yan Wang
- Department of Anesthesia, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Qi-Liang Song
- Department of Anesthesia, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Yu-Long Wang
- Department of Anesthesia, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Zong-Ming Jiang
- Department of Anesthesia, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang Province, China.
| |
Collapse
|
9
|
Chen R, Gao B, Wang X, Zhao H, Wang X, Liu D. Ultrasonographic assessment of renal microcirculation is a new vision for the treatment of intensive care unit associated acute kidney injury. Eur J Med Res 2024; 29:115. [PMID: 38341556 PMCID: PMC10858548 DOI: 10.1186/s40001-024-01704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Affiliation(s)
- Rongping Chen
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Beijun Gao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Xinchen Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Hua Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| | - Dawei Liu
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
10
|
Chen R, Liu D, Zhao H, Wang X. Renal medullary perfusion differs from that in renal cortex in patients with sepsis associated acute kidney injury and correlates with renal function prognosis: A prospective cohort study. Clin Hemorheol Microcirc 2024; 88:181-198. [PMID: 39121113 PMCID: PMC11492038 DOI: 10.3233/ch-242296] [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: 08/11/2024]
Abstract
BACKGROUND Renal perfusion status remains poorly studied at the bedside during sepsis associated acute kidney injury (AKI). The aim of the study is to examine renal cortical and medullary perfusion using renal contrast enhanced ultrasound (CEUS) in septic patients. METHODS In this single-center, prospective longitudinal study, septic patients were enrolled. Renal ultrasonography was performed within 24 hours of ICU admission (D1), then repeated at D3, D5 and D7. Each measurement consisted of three destruction replenishment sequences that were recorded for delayed analysis with dedicated software (Vuebox). Renal cortex and medulla perfusion were quantified by measuring time to peak (TTP). Receiver operating characteristic (ROC) analysis was used to evaluate 28-day renal prognosis. RESULTS The study included 149 septic patients, including 70 non-AKI patients and 79 AKI patients. Both renal cortical and medullary TTP was longer in the AKI group than in the non-AKI group. The difference of TTP between renal cortex and medulla in AKI group was higher than that in the non-AKI group (p = 0.000). Medullary TTP on day 3 had the best performance in predicting the prognosis of 28-day renal function (AUC 0.673, 95% confidence interval 0.528-0.818, p = 0.024), and its cut-off value was 45 s with a sensitivity 52.2% and a specificity of 82.1%. Cortical TTP on day 3 also had the performance in predicting the prognosis of 28-day renal function (AUC 0.657, 95% confidence interval 0.514-0.800, p = 0.039), and its cut-off value was 33 s with a sensitivity 78.3% and a specificity of 55.0%. CONCLUSION Renal medullary perfusion alterations differ from those in cortex, with the medulla is worse. Simultaneous and dynamic assessment of cortical and medullary microcirculatory flow alterations necessary. TTP on day 3, especially medullary TTP, seems to be a relatively stable and useful indicator, which correlates with 28-day renal function prognosis in septic patients. Early correction of renal cortical and medullary perfusion alterations reduces the incidence of adverse renal events.
Collapse
Affiliation(s)
- Rongping Chen
- Peking Union Medical College Hospital, Beijing, China
| | - Dawei Liu
- Peking Union Medical College Hospital, Beijing, China
| | - Hua Zhao
- Peking Union Medical College Hospital, Beijing, China
| | - Xiaoting Wang
- Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
11
|
Zhao WT, Herrmann KH, Sibgatulin R, Nahardani A, Krämer M, Heitplatz B, van Marck V, Reuter S, Reichenbach JR, Hoerr V. Perfusion and T 2 Relaxation Time as Predictors of Severity and Outcome in Sepsis-Associated Acute Kidney Injury: A Preclinical MRI Study. J Magn Reson Imaging 2023; 58:1954-1963. [PMID: 37026419 DOI: 10.1002/jmri.28698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Preventing sepsis-associated acute kidney injury (S-AKI) can be challenging because it develops rapidly and is often asymptomatic. Probability assessment of disease progression for therapeutic follow-up and outcome are important to intervene and prevent further damage. PURPOSE To establish a noninvasive multiparametric MRI (mpMRI) tool, including T1 , T2 , and perfusion mapping, for probability assessment of the outcome of S-AKI. STUDY TYPE Preclinical randomized prospective study. ANIMAL MODEL One hundred and forty adult female SD rats (65 control and 75 sepsis). FIELD STRENGTH/SEQUENCE 9.4T; T1 and perfusion map (FAIR-EPI) and T2 map (multiecho RARE). ASSESSMENT Experiment 1: To identify renal injury in relation to sepsis severity, serum creatinine levels were determined (31 control and 35 sepsis). Experiment 2: Animals underwent mpMRI (T1 , T2 , perfusion) 18 hours postsepsis. A subgroup of animals was immediately sacrificed for histology examination (nine control and seven sepsis). Result of mpMRI in follow-up subgroup (25 control and 33 sepsis) was used to predict survival outcomes at 96 hours. STATISTICAL TESTS Mann-Whitney U test, Spearman/Pearson correlation (r), P < 0.05 was considered statistically significant. RESULTS Severely ill septic animals exhibited significantly increased serum creatinine levels compared to controls (70 ± 30 vs. 34 ± 9 μmol/L, P < 0.0001). Cortical perfusion (480 ± 80 vs. 330 ± 140 mL/100 g tissue/min, P < 0.005), and cortical and medullary T2 relaxation time constants were significantly reduced compared to controls (41 ± 4 vs. 37 ± 5 msec in cortex, P < 0.05, 52 ± 7 vs. 45 ± 6 msec in medulla, P < 0.05). The combination of cortical T2 relaxation time constants and perfusion results at 18 hours could predict survival outcomes at 96 hours with high sensitivity (80%) and specificity (73%) (area under curve of ROC = 0.8, Jmax = 0.52). DATA CONCLUSION This preclinical study suggests combined T2 relaxation time and perfusion mapping as first line diagnostic tool for treatment planning. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Wan-Ting Zhao
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Institute of Medical Microbiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Renat Sibgatulin
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Ali Nahardani
- Heart Center Bonn, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Martin Krämer
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Barbara Heitplatz
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Veerle van Marck
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Stefan Reuter
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital Münster, Münster, Germany
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Verena Hoerr
- Institute of Medical Microbiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Heart Center Bonn, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
- Translational Research Imaging Center (TRIC), Clinic of Radiology, University of Münster, Münster, Germany
| |
Collapse
|
12
|
Yanase F, Spano S, Maeda A, Chaba A, Naorungroj T, Ow CPC, Lankadeva YR, May CN, Betrie AH, Lane DJR, Eastwood GM, Plummer MP, Bellomo R. Mega-dose sodium ascorbate: a pilot, single-dose, physiological effect, double-blind, randomized, controlled trial. Crit Care 2023; 27:371. [PMID: 37828547 PMCID: PMC10571252 DOI: 10.1186/s13054-023-04644-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/09/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Mega-dose sodium ascorbate (NaAscorbate) appears beneficial in experimental sepsis. However, its physiological effects in patients with septic shock are unknown. METHODS We conducted a pilot, single-dose, double-blind, randomized controlled trial. We enrolled patients with septic shock within 24 h of diagnosis. We randomly assigned them to receive a single mega-dose of NaAscorbate (30 g over 1 h followed by 30 g over 5 h) or placebo (vehicle). The primary outcome was the total 24 h urine output (UO) from the beginning of the study treatment. Secondary outcomes included the time course of the progressive cumulative UO, vasopressor dose, and sequential organ failure assessment (SOFA) score. RESULTS We enrolled 30 patients (15 patients in each arm). The mean (95% confidence interval) total 24-h UO was 2056 (1520-2593) ml with placebo and 2948 (2181-3715) ml with NaAscorbate (mean difference 891.5, 95% confidence interval [- 2.1 to 1785.2], P = 0.051). Moreover, the progressive cumulative UO was greater over time on linear mixed modelling with NaAscorbate (P < 0.001). Vasopressor dose and SOFA score changes over time showed faster reductions with NaAscorbate (P < 0.001 and P = 0.042). The sodium level, however, increased more over time with NaAscorbate (P < 0.001). There was no statistical difference in other clinical outcomes. CONCLUSION In patients with septic shock, mega-dose NaAscorbate did not significantly increase cumulative 24-h UO. However, it induced a significantly greater increase in UO and a greater reduction in vasopressor dose and SOFA score over time. One episode of hypernatremia and one of hemolysis were observed in the NaAscorbate group. These findings support further cautious investigation of this novel intervention. Trial registration Australian New Zealand Clinical Trial Registry (ACTRN12620000651987), Date registered June/5/2020.
Collapse
Affiliation(s)
- Fumitaka Yanase
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
- Australian and New Zealand Intensive Care Research Centre (ANZIC-RC), School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Sofia Spano
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
| | - Akinori Maeda
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
| | - Anis Chaba
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
| | | | - Connie Pei Chen Ow
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Yugeesh R Lankadeva
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Critical Care, University of Melbourne, Melbourne, Australia
| | - Clive N May
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Critical Care, University of Melbourne, Melbourne, Australia
| | - Ashenafi H Betrie
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Darius J R Lane
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Glenn M Eastwood
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
- Australian and New Zealand Intensive Care Research Centre (ANZIC-RC), School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Mark P Plummer
- Department of Intensive Care, Royal Adelaide Hospital, Adelaide, Australia
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia.
- Australian and New Zealand Intensive Care Research Centre (ANZIC-RC), School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
- Department of Critical Care, University of Melbourne, Melbourne, Australia.
- Data Analytics Research and Evaluation Centre, Austin Hospital, Melbourne, Australia.
- Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Australia.
| |
Collapse
|
13
|
Betrie AH, Ma S, Ow CPC, Peiris RM, Evans RG, Ayton S, Lane DJR, Southon A, Bailey SR, Bellomo R, May CN, Lankadeva YR. Renal arterial infusion of tempol prevents medullary hypoperfusion, hypoxia, and acute kidney injury in ovine Gram-negative sepsis. Acta Physiol (Oxf) 2023; 239:e14025. [PMID: 37548350 PMCID: PMC10909540 DOI: 10.1111/apha.14025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023]
Abstract
AIM Renal medullary hypoperfusion and hypoxia precede acute kidney injury (AKI) in ovine sepsis. Oxidative/nitrosative stress, inflammation, and impaired nitric oxide generation may contribute to such pathophysiology. We tested whether the antioxidant and anti-inflammatory drug, tempol, may modify these responses. METHODS Following unilateral nephrectomy, we inserted renal arterial catheters and laser-Doppler/oxygen-sensing probes in the renal cortex and medulla. Noanesthetized sheep were administered intravenous (IV) Escherichia coli and, at sepsis onset, IV tempol (IVT; 30 mg kg-1 h-1 ), renal arterial tempol (RAT; 3 mg kg-1 h-1 ), or vehicle. RESULTS Septic sheep receiving vehicle developed renal medullary hypoperfusion (76 ± 16% decrease in perfusion), hypoxia (70 ± 13% decrease in oxygenation), and AKI (87 ± 8% decrease in creatinine clearance) with similar changes during IVT. However, RAT preserved medullary perfusion (1072 ± 307 to 1005 ± 271 units), oxygenation (46 ± 8 to 43 ± 6 mmHg), and creatinine clearance (61 ± 10 to 66 ± 20 mL min-1 ). Plasma, renal medullary, and cortical tissue malonaldehyde and medullary 3-nitrotyrosine decreased significantly with sepsis but were unaffected by IVT or RAT. Consistent with decreased oxidative/nitrosative stress markers, cortical and medullary nuclear factor-erythroid-related factor-2 increased significantly and were unaffected by IVT or RAT. However, RAT prevented sepsis-induced overexpression of cortical tissue tumor necrosis factor alpha (TNF-α; 51 ± 16% decrease; p = 0.003) and medullary Thr-495 phosphorylation of endothelial nitric oxide synthase (eNOS; 63 ± 18% decrease; p = 0.015). CONCLUSIONS In ovine Gram-negative sepsis, renal arterial infusion of tempol prevented renal medullary hypoperfusion and hypoxia and AKI and decreased TNF-α expression and uncoupling of eNOS. However, it did not affect markers of oxidative/nitrosative stress, which were significantly decreased by Gram-negative sepsis.
Collapse
Affiliation(s)
- Ashenafi H. Betrie
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Shuai Ma
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Division of Nephrology, Shanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Connie P. C. Ow
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Rachel M. Peiris
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Roger G. Evans
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Biomedicine Discovery Institute and Department of PhysiologyMonash UniversityMelbourneVictoriaAustralia
| | - Scott Ayton
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Darius J. R. Lane
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Adam Southon
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Simon R. Bailey
- Faculty of Veterinary and Agricultural SciencesThe University of MelbourneMelbourneVictoriaAustralia
| | - Rinaldo Bellomo
- Department of Critical Care, Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
- Australian and New Zealand Intensive Care Research CentreMonash UniversityMelbourneVictoriaAustralia
- Department of Intensive CareAustin HospitalMelbourneVictoriaAustralia
- Department of Intensive CareRoyal Melbourne HospitalMelbourneVictoriaAustralia
| | - Clive N. May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Department of Critical Care, Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
| | - Yugeesh R. Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Department of Critical Care, Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
14
|
Fan Z, Jiang J, Xiao C, Chen Y, Xia Q, Wang J, Fang M, Wu Z, Chen F. Construction and validation of prognostic models in critically Ill patients with sepsis-associated acute kidney injury: interpretable machine learning approach. J Transl Med 2023; 21:406. [PMID: 37349774 PMCID: PMC10286378 DOI: 10.1186/s12967-023-04205-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication in critically ill patients with sepsis and is often associated with a poor prognosis. We aimed to construct and validate an interpretable prognostic prediction model for patients with sepsis-associated AKI (S-AKI) using machine learning (ML) methods. METHODS Data on the training cohort were collected from the Medical Information Mart for Intensive Care IV database version 2.2 to build the model, and data of patients were extracted from Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine for external validation of model. Predictors of mortality were identified using Recursive Feature Elimination (RFE). Then, random forest, extreme gradient boosting (XGBoost), multilayer perceptron classifier, support vector classifier, and logistic regression were used to establish a prognosis prediction model for 7, 14, and 28 days after intensive care unit (ICU) admission, respectively. Prediction performance was assessed using the receiver operating characteristic (ROC) curve and decision curve analysis (DCA). SHapley Additive exPlanations (SHAP) were used to interpret the ML models. RESULTS In total, 2599 patients with S-AKI were included in the analysis. Forty variables were selected for the model development. According to the areas under the ROC curve (AUC) and DCA results for the training cohort, XGBoost model exhibited excellent performance with F1 Score of 0.847, 0.715, 0.765 and AUC (95% CI) of 0.91 (0.90, 0.92), 0.78 (0.76, 0.80), and 0.83 (0.81, 0.85) in 7 days, 14 days and 28 days group, respectively. It also demonstrated excellent discrimination in the external validation cohort. Its AUC (95% CI) was 0.81 (0.79, 0.83), 0.75 (0.73, 0.77), 0.79 (0.77, 0.81) in 7 days, 14 days and 28 days group, respectively. SHAP-based summary plot and force plot were used to interpret the XGBoost model globally and locally. CONCLUSIONS ML is a reliable tool for predicting the prognosis of patients with S-AKI. SHAP methods were used to explain intrinsic information of the XGBoost model, which may prove clinically useful and help clinicians tailor precise management.
Collapse
Affiliation(s)
- Zhiyan Fan
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Jiamei Jiang
- Department of Ultrasound, The First Affiliated Hospital Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
| | - Chen Xiao
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Youlei Chen
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Quan Xia
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Juan Wang
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Mengjuan Fang
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Zesheng Wu
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Fanghui Chen
- Department of Emergency, Hangzhou First People's Hospital Affiliated to Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Norepinephrine May Exacerbate Septic Acute Kidney Injury: A Narrative Review. J Clin Med 2023; 12:jcm12041373. [PMID: 36835909 PMCID: PMC9960985 DOI: 10.3390/jcm12041373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Sepsis, the most serious complication of infection, occurs when a cascade of potentially life-threatening inflammatory responses is triggered. Potentially life-threatening septic shock is a complication of sepsis that occurs when hemodynamic instability occurs. Septic shock may cause organ failure, most commonly involving the kidneys. The pathophysiology and hemodynamic mechanisms of acute kidney injury in the case of sepsis or septic shock remain to be elucidated, but previous studies have suggested multiple possible mechanisms or the interplay of multiple mechanisms. Norepinephrine is used as the first-line vasopressor in the management of septic shock. Studies have reported different hemodynamic effects of norepinephrine on renal circulation, with some suggesting that it could possibly exacerbate acute kidney injury caused by septic shock. This narrative review briefly covers the updates on sepsis and septic shock regarding definitions, statistics, diagnosis, and management, with an explanation of the putative pathophysiological mechanisms and hemodynamic changes, as well as updated evidence. Sepsis-associated acute kidney injury remains a major burden on the healthcare system. This review aims to improve the real-world clinical understanding of the possible adverse outcomes of norepinephrine use in sepsis-associated acute kidney injury.
Collapse
|
16
|
Hu RT, Lankadeva YR, Yanase F, Osawa EA, Evans RG, Bellomo R. Continuous bladder urinary oxygen tension as a new tool to monitor medullary oxygenation in the critically ill. Crit Care 2022; 26:389. [PMID: 36527088 PMCID: PMC9758873 DOI: 10.1186/s13054-022-04230-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury (AKI) is common in the critically ill. Inadequate renal medullary tissue oxygenation has been linked to its pathogenesis. Moreover, renal medullary tissue hypoxia can be detected before biochemical evidence of AKI in large mammalian models of critical illness. This justifies medullary hypoxia as a pathophysiological biomarker for early detection of impending AKI, thereby providing an opportunity to avert its evolution. Evidence from both animal and human studies supports the view that non-invasively measured bladder urinary oxygen tension (PuO2) can provide a reliable estimate of renal medullary tissue oxygen tension (tPO2), which can only be measured invasively. Furthermore, therapies that modify medullary tPO2 produce corresponding changes in bladder PuO2. Clinical studies have shown that bladder PuO2 correlates with cardiac output, and that it increases in response to elevated cardiopulmonary bypass (CPB) flow and mean arterial pressure. Clinical observational studies in patients undergoing cardiac surgery involving CPB have shown that bladder PuO2 has prognostic value for subsequent AKI. Thus, continuous bladder PuO2 holds promise as a new clinical tool for monitoring the adequacy of renal medullary oxygenation, with its implications for the recognition and prevention of medullary hypoxia and thus AKI.
Collapse
Affiliation(s)
- Raymond T. Hu
- grid.410678.c0000 0000 9374 3516Department of Anaesthesia, Austin Health, Heidelberg, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Critical Care, Melbourne Medical School, The University of Melbourne, Parkville, VIC Australia
| | - Yugeesh R. Lankadeva
- grid.1008.90000 0001 2179 088XDepartment of Critical Care, Melbourne Medical School, The University of Melbourne, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XPre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
| | - Fumitake Yanase
- grid.414094.c0000 0001 0162 7225Department of Intensive Care, Austin Hospital, Heidelberg, Australia
| | - Eduardo A. Osawa
- Cardiology Intensive Care Unit, DF Star Hospital, Brasília, Brazil ,grid.472984.4D’Or Institute for Research and Education (IDOR), DF Star Hospital, Brasília, Brazil
| | - Roger G. Evans
- grid.1008.90000 0001 2179 088XPre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia ,grid.1002.30000 0004 1936 7857Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC Australia
| | - Rinaldo Bellomo
- grid.1008.90000 0001 2179 088XDepartment of Critical Care, Melbourne Medical School, The University of Melbourne, Parkville, VIC Australia ,grid.414094.c0000 0001 0162 7225Department of Intensive Care, Austin Hospital, Heidelberg, Australia ,grid.1002.30000 0004 1936 7857Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia ,grid.416153.40000 0004 0624 1200Department of Intensive Care, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
17
|
Wang X, Chen L, Su T. Evaluating renal microcirculation in patients with acute kidney injury by contrast-enhanced ultrasonography: a protocol for an observational cohort study. BMC Nephrol 2022; 23:392. [PMID: 36482379 PMCID: PMC9733337 DOI: 10.1186/s12882-022-03021-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) in critically ill patients has poor renal outcome with high mortality. Changes in intra-renal microcirculation and tissue oxygenation are currently considered essential pathophysiological mechanisms to the development and progression of AKI. This study aims to investigate the characteristics of contrast-enhanced ultrasonography (CEUS) derived parameters in biopsy-proven AKI patients, and examine the predictive value of these markers for renal outcome. METHODS AND DESIGN This prospective observational study will enroll AKI patients who are diagnosed and staging following KDIGO (Kidney Disease: Improving Global Outcomes) criteria. All patients undergo a kidney biopsy and pathological tubulointerstitial nephropathy is confirmed. The CEUS examination will be performed at 0, 4 and 12 weeks after biopsy to monitor renal microcirculation. The percentage decrease of serum creatinine, 4-week and 12-week eGFR (estimated glomerular filtration rate) will also be reviewed as renal prognosis. The relationship of CEUS parameters with clinical and pathological markers will be analyzed. We perform a lassologit procedure to select potential affecting variables, including clinical, laboratory indexes and CEUS markers, to be included in the logistic regression model, and examine their predictive performance to AKI outcomes. DISCUSSION If we are able to show that CEUS derived parameters contribute to diagnosis and prognosis of AKI, the quality of life of patients will be improved while healthcare costs will be reduced. TRIAL REGISTRATION This study is retrospectively registered on the Chinese Medical Research Registration information System( https://61.49.19.26/login ) on December 31, 2021: MR-11-22-003,503. This study has been approved by the Ethics and Scientific Research Department of Peking University First Hospital.
Collapse
Affiliation(s)
- Xiangyu Wang
- grid.411472.50000 0004 1764 1621Department of Ultrasound, Peking University First Hospital, Beijing, China
| | - Luzeng Chen
- grid.411472.50000 0004 1764 1621Department of Ultrasound, Peking University First Hospital, Beijing, China
| | - Tao Su
- grid.411472.50000 0004 1764 1621Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Institute of Nephrology, Peking University, No 8, Xishiku Street, Xicheng District, Beijing, 100034 China
| |
Collapse
|
18
|
Kim J, Stolarski A, Zhang Q, Wee K, Remick D. HYDROCORTISONE, ASCORBIC ACID, AND THIAMINE THERAPY DECREASE RENAL OXIDATIVE STRESS AND ACUTE KIDNEY INJURY IN MURINE SEPSIS. Shock 2022; 58:426-433. [PMID: 36445231 PMCID: PMC9713586 DOI: 10.1097/shk.0000000000001995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
ABSTRACT Background: Acute kidney injury (AKI) occurs frequently in septic patients and correlates with increased mortality. Because clinical studies investigating hydrocortisone, ascorbic acid, and thiamine (HAT) have demonstrated discordant results, studies were performed using mortality stratification for therapy to identify candidates for therapy and determine mechanisms of organ injury. Methods: Sepsis was induced using the cecal ligation and puncture (CLP) model of sepsis with fluid and antibiotic support. Heart rate (HR) measurements obtained 6 hours after CLP stratified mice into live predicted (P-Live) or die predicted (P-Die). Stratified mice were then randomized for treatment with HAT or vehicle given 7 hours after CLP. Physiologic measurements were taken again at 24 hours, and mice were killed to collect blood and organs. Results: The following five groups were created: (1) P-Live vehicle, (2) P-Live HAT, (3) P-Die vehicle, (4) P-Die HAT, and (5) naive mice. Comparisons were made to test the hypotheses that (1) P-Die vehicle mice will have significant deterioration compared with P-Live mice targeting the kidney and (2) HAT will correct these deleterious changes in P-Die mice. Compared with P-Live, P-Die mice had a significant decline in all measured physiologic parameters (HR, cardiac output, breath rate, and temperature), which were corrected with HAT therapy (P < 0.05 for all parameters). The P-Die mice had declines in the ascorbic acid within the blood, peritoneal lavage, and kidney homogenate compared with P-Live mice indicating consumption, and the decline was corrected with HAT. Elevated IL-6, KC, Macrophage Inflammatory Protein-2, and IL-1RA were found in P-Die mice and decreased with HAT. Markers of endothelial cell injury (glypican 1 and glypican 4) were elevated in the P-Die mice, and these values were decreased with HAT therapy. Low oxygen levels with subsequent oxidative stress (OS) in the kidney were visualized in histologic sections using hypoxyprobe and also with carbonyl proteins and 8-iso-prostaglandin F2α in kidney homogenates. The P-Die mice had significant elevations of renal OSs, which was ameliorated with HAT. Kidney injury was evident in the P-Die mice compared with P-Live mice with elevations in blood urea nitrogen and cystatin C, which were significantly reduced with HAT. There was no evidence of global hypoxia or organ injury because hepatic parameters remained normal. Conclusions: Our data show that in CLP-induced sepsis, P-Die mice have increased inflammation, OS, and kidney injury. Hydrocortisone, ascorbic acid, and thiamine therapy decreased renal OS and injury in the P-Die group when given after the onset of sepsis-induced physiologic changes.
Collapse
Affiliation(s)
- John Kim
- Departments of Pathology and Laboratory Medicine
| | - Allan Stolarski
- Surgery, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | | | | | | |
Collapse
|
19
|
He FF, Wang YM, Chen YY, Huang W, Li ZQ, Zhang C. Sepsis-induced AKI: From pathogenesis to therapeutic approaches. Front Pharmacol 2022; 13:981578. [PMID: 36188562 PMCID: PMC9522319 DOI: 10.3389/fphar.2022.981578] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a heterogenous and highly complex clinical syndrome, which is caused by infectious or noninfectious factors. Acute kidney injury (AKI) is one of the most common and severe complication of sepsis, and it is associated with high mortality and poor outcomes. Recent evidence has identified that autophagy participates in the pathophysiology of sepsis-associated AKI. Despite the use of antibiotics, the mortality rate is still at an extremely high level in patients with sepsis. Besides traditional treatments, many natural products, including phytochemicals and their derivatives, are proved to exert protective effects through multiple mechanisms, such as regulation of autophagy, inhibition of inflammation, fibrosis, and apoptosis, etc. Accumulating evidence has also shown that many pharmacological inhibitors might have potential therapeutic effects in sepsis-induced AKI. Hence, understanding the pathophysiology of sepsis-induced AKI may help to develop novel therapeutics to attenuate the complications of sepsis and lower the mortality rate. This review updates the recent progress of underlying pathophysiological mechanisms of sepsis-associated AKI, focuses specifically on autophagy, and summarizes the potential therapeutic effects of phytochemicals and pharmacological inhibitors.
Collapse
|
20
|
Abstract
Sepsis-associated AKI is a life-threatening complication that is associated with high morbidity and mortality in patients who are critically ill. Although it is clear early supportive interventions in sepsis reduce mortality, it is less clear that they prevent or ameliorate sepsis-associated AKI. This is likely because specific mechanisms underlying AKI attributable to sepsis are not fully understood. Understanding these mechanisms will form the foundation for the development of strategies for early diagnosis and treatment of sepsis-associated AKI. Here, we summarize recent laboratory and clinical studies, focusing on critical factors in the pathophysiology of sepsis-associated AKI: microcirculatory dysfunction, inflammation, NOD-like receptor protein 3 inflammasome, microRNAs, extracellular vesicles, autophagy and efferocytosis, inflammatory reflex pathway, vitamin D, and metabolic reprogramming. Lastly, identifying these molecular targets and defining clinical subphenotypes will permit precision approaches in the prevention and treatment of sepsis-associated AKI.
Collapse
Affiliation(s)
- Shuhei Kuwabara
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Eibhlin Goggins
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
21
|
Fan H, Su BJ, Le JW, Zhu JH. Salidroside Protects Acute Kidney Injury in Septic Rats by Inhibiting Inflammation and Apoptosis. Drug Des Devel Ther 2022; 16:899-907. [PMID: 35386851 PMCID: PMC8978577 DOI: 10.2147/dddt.s361972] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/17/2022] [Indexed: 12/27/2022] Open
Abstract
Purpose To clarify the protective effect and mechanism of salidroside (SLDS) on acute kidney injury (AKI) in septic rats. Methods We pretreated rats with different doses of SLDS and analyzed the impact of SLDS on the survival of septic rats. We evaluated the levels of inflammatory factors in rats, the expression of NF-ƙB p65 in the kidney, and the apoptosis of kidney tubular epithelial cells (KTECs). Results SLDS significantly decreased the mortality of septic rats, and it reduced the levels of TNF-α, IL-1β, and IL-17A in plasma and kidneys and decreased the levels of serum creatinine, plasma renal injury molecule-1 and plasma neutrophil gelatin-associated lipocalin. Moreover, SLDS could significantly decrease the expression of NF-ƙB p65 in kidney tissues and the apoptotic number of KETCs, while reducing the mRNA levels of Caspase-3 and Bax mRNA, and increasing the level of Bcl-2 mRNA. Conclusion SLDS pretreatment protects against AKI in septic rats by inhibiting the inflammation of kidney and the apoptosis of KTECs.
Collapse
Affiliation(s)
- Heng Fan
- Department of Intensive Care Unit, Ningbo First Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Bin-Jie Su
- Department of Intensive Care Unit, Ningbo First Hospital Haishu Branch, Ningbo, Zhejiang Province, People's Republic of China
| | - Jian-Wei Le
- Department of Intensive Care Unit, Ningbo First Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Jian-Hua Zhu
- Department of Intensive Care Unit, Ningbo First Hospital, Ningbo, Zhejiang Province, People's Republic of China
| |
Collapse
|
22
|
Molema G, Zijlstra JG, van Meurs M, Kamps JAAM. Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat Rev Nephrol 2022; 18:95-112. [PMID: 34667283 DOI: 10.1038/s41581-021-00489-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 12/29/2022]
Abstract
Microvascular endothelial cells in the kidney have been a neglected cell type in sepsis-induced acute kidney injury (sepsis-AKI) research; yet, they offer tremendous potential as pharmacological targets. As endothelial cells in distinct cortical microvascular segments are highly heterogeneous, this Review focuses on endothelial cells in their anatomical niche. In animal models of sepsis-AKI, reduced glomerular blood flow has been attributed to inhibition of endothelial nitric oxide synthase activation in arterioles and glomeruli, whereas decreased cortex peritubular capillary perfusion is associated with epithelial redox stress. Elevated systemic levels of vascular endothelial growth factor, reduced levels of circulating sphingosine 1-phosphate and loss of components of the glycocalyx from glomerular endothelial cells lead to increased microvascular permeability. Although coagulation disbalance occurs in all microvascular segments, the molecules involved differ between segments. Induction of the expression of adhesion molecules and leukocyte recruitment also occurs in a heterogeneous manner. Evidence of similar endothelial cell responses has been found in kidney and blood samples from patients with sepsis. Comprehensive studies are needed to investigate the relationships between segment-specific changes in the microvasculature and kidney function loss in sepsis-AKI. The application of omics technologies to kidney tissues from animals and patients will be key in identifying these relationships and in developing novel therapeutics for sepsis.
Collapse
Affiliation(s)
- Grietje Molema
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - Jan G Zijlstra
- Dept. Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Matijs van Meurs
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Dept. Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan A A M Kamps
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
23
|
Piedrafita A, Balayssac S, Casemayou A, Saulnier-Blache JS, Lucas A, Iacovoni JS, Breuil B, Chauveau D, Decramer S, Malet-Martino M, Schanstra JP, Faguer S. Hepatocyte nuclear factor-1β shapes the energetic homeostasis of kidney tubule cells. FASEB J 2021; 35:e21931. [PMID: 34653285 DOI: 10.1096/fj.202100782rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/28/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022]
Abstract
Energetic metabolism controls key steps of kidney development, homeostasis, and epithelial repair following acute kidney injury (AKI). Hepatocyte nuclear factor-1β (HNF-1β) is a master transcription factor that controls mitochondrial function in proximal tubule (PT) cells. Patients with HNF1B pathogenic variant display a wide range of kidney developmental abnormalities and progressive kidney fibrosis. Characterizing the metabolic changes in PT cells with HNF-1β deficiency may help to identify new targetable molecular hubs involved in HNF1B-related kidney phenotypes and AKI. Here, we combined 1 H-NMR-based metabolomic analysis in a murine PT cell line with CrispR/Cas9-induced Hnf1b invalidation (Hnf1b-/- ), clustering analysis, targeted metabolic assays, and datamining of published RNA-seq and ChIP-seq dataset to identify the role of HNF-1β in metabolism. Hnf1b-/- cells grown in normoxic conditions display intracellular ATP depletion, increased cytosolic lactate concentration, increased lipid droplet content, failure to use pyruvate for energetic purposes, increased levels of tricarboxylic acid (TCA) cycle intermediates and oxidized glutathione, and a reduction of TCA cycle byproducts, all features consistent with mitochondrial dysfunction and an irreversible switch toward glycolysis. Unsupervised clustering analysis showed that Hnf1b-/- cells mimic a hypoxic signature and that they cannot furthermore increase glycolysis-dependent energetic supply during hypoxic challenge. Metabolome analysis also showed alteration of phospholipid biosynthesis in Hnf1b-/- cells leading to the identification of Chka, the gene coding for choline kinase α, as a new putative target of HNF-1β. HNF-1β shapes the energetic metabolism of PT cells and HNF1B deficiency in patients could lead to a hypoxia-like metabolic state precluding further adaptation to ATP depletion following AKI.
Collapse
Affiliation(s)
- Alexis Piedrafita
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Balayssac
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France.,Laboratoire des Interaction Moléculaires et Réactivité Chimique et Photochimique (IMRCP), UMR 5623, Toulouse, France
| | - Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jean-Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Alexandre Lucas
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Jason S Iacovoni
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Dominique Chauveau
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Service de Néphrologie, Médecine interne et Hypertension artérielle, Hôpital des Enfants, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Myriam Malet-Martino
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
24
|
Ow CPC, Trask-Marino A, Betrie AH, Evans RG, May CN, Lankadeva YR. Targeting Oxidative Stress in Septic Acute Kidney Injury: From Theory to Practice. J Clin Med 2021; 10:jcm10173798. [PMID: 34501245 PMCID: PMC8432047 DOI: 10.3390/jcm10173798] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Sepsis is the leading cause of acute kidney injury (AKI) and leads to increased morbidity and mortality in intensive care units. Current treatments for septic AKI are largely supportive and are not targeted towards its pathophysiology. Sepsis is commonly characterized by systemic inflammation and increased production of reactive oxygen species (ROS), particularly superoxide. Concomitantly released nitric oxide (NO) then reacts with superoxide, leading to the formation of reactive nitrogen species (RNS), predominantly peroxynitrite. Sepsis-induced ROS and RNS can reduce the bioavailability of NO, mediating renal microcirculatory abnormalities, localized tissue hypoxia and mitochondrial dysfunction, thereby initiating a propagating cycle of cellular injury culminating in AKI. In this review, we discuss the various sources of ROS during sepsis and their pathophysiological interactions with the immune system, microcirculation and mitochondria that can lead to the development of AKI. We also discuss the therapeutic utility of N-acetylcysteine and potential reasons for its efficacy in animal models of sepsis, and its inefficacy in ameliorating oxidative stress-induced organ dysfunction in human sepsis. Finally, we review the pre-clinical studies examining the antioxidant and pleiotropic actions of vitamin C that may be of benefit for mitigating septic AKI, including future implications for clinical sepsis.
Collapse
Affiliation(s)
- Connie P. C. Ow
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka 564-8565, Japan
| | - Anton Trask-Marino
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
| | - Ashenafi H. Betrie
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Roger G. Evans
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
| | - Clive N. May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Yugeesh R. Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
- Correspondence: ; Tel.: +61-3-8344-0417; Fax: +61-3-9035-3107
| |
Collapse
|
25
|
Ruegg G, Luethi N, Cioccari L. The Role of Dexmedetomidine for the Prevention of Acute Kidney Injury in Critical Care. EMJ NEPHROLOGY 2021:97-106. [DOI: 10.33590/emjnephrol/21-00087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
Acute kidney injury (AKI) occurs in up to 50% of patients admitted to the intensive care unit and is associated with increased mortality. Currently, there is no effective pharmacotherapy for prevention or treatment of AKI. In animal models of sepsis and ischaemia-reperfusion, α2-agonists like dexmedetomidine (DEX) exhibit anti-inflammatory properties and experimental data indicate a potential protective effect of DEX on renal function. However, clinical trials have yielded inconsistent results in critically ill patients. This review discusses the pathophysiological mechanisms involved in AKI, the renal effects of DEX in various intensive care unit-related conditions, and summarises the available literature addressing the use of DEX for the prevention of AKI.
Collapse
Affiliation(s)
- Gion Ruegg
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nora Luethi
- Department of Emergency Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Prahran, Australia
| | - Luca Cioccari
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Prahran, Australia
| |
Collapse
|
26
|
Lu HY, Wang GY, Zhao JW, Jiang HT. Knockdown of lncRNA MALAT1 ameliorates acute kidney injury by mediating the miR-204/APOL1 pathway. J Clin Lab Anal 2021; 35:e23881. [PMID: 34240756 PMCID: PMC8373329 DOI: 10.1002/jcla.23881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/09/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background Acute kidney injury (AKI) was characterized by loss of renal function, associated with chronic kidney disease, end‐stage renal disease, and length of hospital stay. Long non‐coding RNAs (lncRNAs) participated in AKI development and progression. Here, we aimed to investigate the roles and mechanisms of lncRNA MALAT1 in AKI. Methods AKI serum samples were obtained from 129 AKI patients. ROC analysis was conducted to confirm the diagnostic value of MALAT1 in differentiating AKI from healthy volunteers. After hypoxic treatment on HK‐2 cells, the expressions of inflammatory cytokines, MALAT1, miR‐204, APOL1, p65, and p‐p65, were measured by RT‐qPCR and Western blot assays. The targeted relationship between miR‐204 and MALAT1 or miR‐204 and APOL1 was determined by luciferase reporter assay and RNA pull‐down analysis. After transfection, CCK‐8, flow cytometry, and TUNEL staining assays were performed to evaluate the effects of MALAT1 and miR‐204 on AKI progression. Results From the results, lncRNA MALAT1 was strongly elevated in serum samples from AKI patients, with the high sensitivity and specificity concerning differentiating AKI patients from healthy controls. In vitro, we established the AKI cell model after hypoxic treatment. After experiencing hypoxia, we found significantly increased MALAT1, IL‐1β, IL‐6, and TNF‐α expressions along with decreased miR‐204 level. Moreover, the targeted relationship between MALAT1 and miR‐204 was confirmed. Silencing of MALAT1 could reverse hypoxia‐triggered promotion of HK‐2 cell apoptosis. Meanwhile, the increase of IL‐1β, IL‐6, and TNF‐α after hypoxia treatment could be repressed by MALAT1 knockdown as well. After co‐transfection with MALAT1 silencing and miR‐204 inhibition, we found that miR‐204 could counteract the effects of MALAT1 on HK‐2 cell progression and inflammation after under hypoxic conditions. Finally, NF‐κB signaling was inactivated while APOL1 expression was increased in HK‐2 cells after hypoxia treatment, and lncRNA MALAT1 inhibition reactivated NF‐κB signaling while suppressed APOL1 expression by sponging miR‐204. Conclusions Collectively, these results illustrated that knockdown of lncRNA MALAT1 could ameliorate AKI progression and inflammation by targeting miR‐204 through APOL1/NF‐κB signaling.
Collapse
Affiliation(s)
- Hai-Yuan Lu
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Guo-Yi Wang
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jin-Wen Zhao
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hai-Tao Jiang
- Department of Orthopedics, Huai'an First People's Hospital, Huai'an, China
| |
Collapse
|
27
|
May CN, Bellomo R, Lankadeva YR. Therapeutic potential of megadose vitamin C to reverse organ dysfunction in sepsis and COVID-19. Br J Pharmacol 2021; 178:3864-3868. [PMID: 34061355 PMCID: PMC8239596 DOI: 10.1111/bph.15579] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/13/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
Sepsis induced by bacteria or viruses can result in multiorgan dysfunction, which is a major cause of death in intensive care units. Current treatments are only supportive, and there are no treatments that reverse the pathophysiological effects of sepsis. Vitamin C has antioxidant, anti‐inflammatory, anticoagulant and immune modulatory actions, so it is a rational treatment for sepsis. Here, we summarise data that support the use of megadose vitamin C as a treatment for sepsis and COVID‐19. Megadose intravenous sodium ascorbate (150 g per 40 kg over 7 h) dramatically improved the clinical state and cardiovascular, pulmonary, hepatic and renal function and decreased body temperature, in a clinically relevant ovine model of Gram‐negative bacteria‐induced sepsis. In a critically ill COVID‐19 patient, intravenous sodium ascorbate (60 g) restored arterial pressure, improved renal function and increased arterial blood oxygen levels. These findings suggest that megadose vitamin C should be trialled as a treatment for sepsis and COVID‐19.
Collapse
Affiliation(s)
- Clive N May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Critical Care, Melbourne Medical School, University of Melbourne, Parkville, Victoria, Australia
| | - Rinaldo Bellomo
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Parkville, Victoria, Australia.,Department of Intensive Care, Austin Hospital, Heidelberg, Victoria, Australia
| | - Yugeesh R Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Critical Care, Melbourne Medical School, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
28
|
Beneficial Effects of Vasopressin Compared With Norepinephrine on Renal Perfusion, Oxygenation, and Function in Experimental Septic Acute Kidney Injury. Crit Care Med 2021; 48:e951-e958. [PMID: 32931198 DOI: 10.1097/ccm.0000000000004511] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVES To compare the effects of restoring mean arterial pressure with vasopressin or norepinephrine on systemic hemodynamics, renal blood flow, intrarenal perfusion and oxygenation, and renal function in ovine septic acute kidney injury. DESIGN Interventional Study. SETTING Research Institute. SUBJECTS Adult Merino ewes. INTERVENTIONS Flow probes were implanted on the pulmonary and renal arteries (and the mesenteric artery in sheep that received vasopressin). Fiber-optic probes were implanted in the renal cortex and medulla to measure tissue perfusion and oxygen tension (PO2). Conscious sheep were administered Escherichia coli to induce septic acute kidney injury. Vasopressin (0.03 IU/min [0.03-0.05 IU/min]; n = 7) or norepinephrine (0.60 μg/kg/min [0.30-0.70 μg/kg/min]; n = 7) was infused IV and titrated to restore baseline mean arterial pressure during 24-30 hours of sepsis. MEASUREMENTS AND MAIN RESULTS Ovine septic acute kidney injury was characterized by reduced mean arterial pressure (-16% ± 2%) and creatinine clearance (-65% ± 9%) and increased renal blood flow (+34% ± 7%) but reduced renal medullary perfusion (-44% ± 7%) and PO2 (-47% ± 10%). Vasopressin infusion did not significantly affect renal medullary perfusion or PO2 and induced a sustained (6 hr) ~2.5-fold increase in creatinine clearance. Vasopressin reduced sepsis-induced mesenteric hyperemia (+61 ± 13 to +9% ± 6%). Norepinephrine transiently (2 hr) improved creatinine clearance (by ~3.5-fold) but worsened renal medullary ischemia (to -64% ± 7%) and hypoxia (to -71% ± 6%). CONCLUSIONS In ovine septic acute kidney injury, restoration of mean arterial pressure with vasopressin induced a more sustained improvement in renal function than norepinephrine, without exacerbating renal medullary ischemia and hypoxia or reducing mesenteric blood flow below baseline values.
Collapse
|
29
|
Hirota K. HIF-α Prolyl Hydroxylase Inhibitors and Their Implications for Biomedicine: A Comprehensive Review. Biomedicines 2021; 9:biomedicines9050468. [PMID: 33923349 PMCID: PMC8146675 DOI: 10.3390/biomedicines9050468] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Oxygen is essential for the maintenance of the body. Living organisms have evolved systems to secure an oxygen environment to be proper. Hypoxia-inducible factor (HIF) plays an essential role in this process; it is a transcription factor that mediates erythropoietin (EPO) induction at the transcriptional level under hypoxic environment. After successful cDNA cloning in 1995, a line of studies were conducted for elucidating the molecular mechanism of HIF activation in response to hypoxia. In 2001, cDNA cloning of dioxygenases acting on prolines and asparagine residues, which play essential roles in this process, was reported. HIF-prolyl hydroxylases (PHs) are molecules that constitute the core molecular mechanism of detecting a decrease in the partial pressure of oxygen, or hypoxia, in the cells; they can be called oxygen sensors. In this review, I discuss the process of molecular cloning of HIF and HIF-PH, which explains hypoxia-induced EPO expression; the development of HIF-PH inhibitors that artificially or exogenously activate HIF by inhibiting HIF-PH; and the significance and implications of medical intervention using HIF-PH inhibitors.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
30
|
Lankadeva YR, Evans RG, Cochrane AD, Marino B, Hood SG, McCall PR, Iguchi N, Bellomo R, May CN. Reversal of renal tissue hypoxia during experimental cardiopulmonary bypass in sheep by increased pump flow and arterial pressure. Acta Physiol (Oxf) 2021; 231:e13596. [PMID: 34347356 DOI: 10.1111/apha.13596] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/17/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022]
Abstract
AIM Renal tissue hypoxia during cardiopulmonary bypass could contribute to the pathophysiology of acute kidney injury. We tested whether renal tissue hypoxia can be alleviated during cardiopulmonary bypass by the combined increase in target pump flow and mean arterial pressure. METHODS Cardiopulmonary bypass was established in eight instrumented sheep under isoflurane anaesthesia, at a target continuous pump flow of 80 mL·kg-1 min-1 and mean arterial pressure of 65 mmHg. We then tested the effects of simultaneously increasing target pump flow to 104 mL·kg-1 min-1 and mean arterial pressure to 80 mmHg with metaraminol (total dose 0.25-3.75 mg). We also tested the effects of transitioning from continuous flow to partially pulsatile flow (pulse pressure ~15 mmHg). RESULTS Compared with conscious sheep, at the lower target pump flow and mean arterial pressure, cardiopulmonary bypass was accompanied by reduced renal blood flow (6.8 ± 1.2 to 1.95 ± 0.76 mL·min-1 kg-1) and renal oxygen delivery (0.91 ± 0.18 to 0.24 ± 0.11 mL·O2 min-1 kg-1). There were profound reductions in cortical oxygen tension (PO2) (33 ± 13 to 6 ± 6 mmHg) and medullary PO2 (31 ± 12 to 8 ± 8 mmHg). Increasing target pump flow and mean arterial pressure increased renal blood flow (to 2.6 ± 1.0 mL·min-1 kg-1) and renal oxygen delivery (to 0.32 ± 0.13 mL·O2 min-1kg-1) and returned cortical PO2 to 58 ± 60 mmHg and medullary PO2 to 28 ± 16 mmHg; levels similar to those of conscious sheep. Partially pulsatile pump flow had no significant effects on renal perfusion or oxygenation. CONCLUSIONS Renal hypoxia during experimental CPB can be corrected by increasing target pump flow and mean arterial pressure within a clinically feasible range.
Collapse
Affiliation(s)
- Yugeesh R. Lankadeva
- Pre‐Clinical Critical Care Unit Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne Melbourne VIC Australia
| | - Roger G. Evans
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne VIC Australia
| | - Andrew D. Cochrane
- Department of Cardiothoracic Surgery Monash Health and Department of Surgery (School of Clinical Sciences at Monash Health) Monash University Melbourne VIC Australia
| | - Bruno Marino
- Cellsaving and Perfusion Resources Melbourne VIC Australia
| | - Sally G. Hood
- Pre‐Clinical Critical Care Unit Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne Melbourne VIC Australia
| | - Peter R. McCall
- Department of Anaesthesia Austin Health Heidelberg VIC Australia
| | - Naoya Iguchi
- Pre‐Clinical Critical Care Unit Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne Melbourne VIC Australia
| | - Rinaldo Bellomo
- Department of Intensive Care Austin Health Heidelberg VIC Australia
| | - Clive N. May
- Pre‐Clinical Critical Care Unit Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne Melbourne VIC Australia
| |
Collapse
|
31
|
Lankadeva YR, Shehabi Y, Deane AM, Plummer MP, Bellomo R, May CN. Emerging benefits and drawbacks of α 2 -adrenoceptor agonists in the management of sepsis and critical illness. Br J Pharmacol 2021; 178:1407-1425. [PMID: 33450087 DOI: 10.1111/bph.15363] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 12/29/2022] Open
Abstract
Agonists of α2 -adrenoceptors are increasingly being used for the provision of comfort, sedation and the management of delirium in critically ill patients, with and without sepsis. In this context, increased sympathetic and inflammatory activity are common pathophysiological features linked to multi-organ dysfunction, particularly in patients with sepsis or those undergoing cardiac surgery requiring cardiopulmonary bypass. Experimental and clinical studies support the notion that the α2 -adrenoceptor agonists, dexmedetomidine and clonidine, mitigate sympathetic and inflammatory overactivity in sepsis and cardiac surgery requiring cardiopulmonary bypass. These effects can protect vital organs, including the cardiovascular system, kidneys, heart and brain. We review the pharmacodynamic mechanisms by which α2 -adrenoceptor agonists might mitigate multi-organ dysfunction arising from pathophysiological conditions associated with excessive inflammatory and adrenergic stress in experimental studies. We also outline recent clinical trials that have examined the use of dexmedetomidine in critically ill patients with and without sepsis and in patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Yugeesh R Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.,Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Yahya Shehabi
- Department of Intensive Care Medicine, Monash Health, School of Clinical Sciences, Monash University, Melbourne, Prince of Wales Clinical School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Adam M Deane
- Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia.,Department of Intensive Care Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Mark P Plummer
- Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia.,Department of Intensive Care Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Rinaldo Bellomo
- Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Clive N May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.,Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Lankadeva YR, Peiris RM, Okazaki N, Birchall IE, Trask-Marino A, Dornom A, Vale TAM, Evans RG, Yanase F, Bellomo R, May CN. Reversal of the Pathophysiological Responses to Gram-Negative Sepsis by Megadose Vitamin C. Crit Care Med 2021; 49:e179-e190. [PMID: 33239507 PMCID: PMC7803449 DOI: 10.1097/ccm.0000000000004770] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Oxidative stress appears to initiate organ failure in sepsis, justifying treatment with antioxidants such as vitamin C at megadoses. We have therefore investigated the safety and efficacy of megadose sodium ascorbate in sepsis. DESIGN Interventional study. SETTING Research Institute. SUBJECTS Adult Merino ewes. INTERVENTIONS Sheep were instrumented with pulmonary and renal artery flow-probes, and laser-Doppler and oxygen-sensing probes in the kidney. Conscious sheep received an infusion of live Escherichia coli for 31 hours. At 23.5 hours of sepsis, sheep received fluid resuscitation (30 mL/kg, Hartmann solution) and were randomized to IV sodium ascorbate (0.5 g/kg over 0.5 hr + 0.5 g/kg/hr for 6.5 hr; n = 5) or vehicle (n = 5). Norepinephrine was titrated to restore mean arterial pressure to baseline values (~80 mm Hg). MEASUREMENTS AND MAIN RESULTS Sepsis-induced fever (41.4 ± 0.2°C; mean ± se), tachycardia (141 ± 2 beats/min), and a marked deterioration in clinical condition in all cases. Mean arterial pressure (86 ± 1 to 67 ± 2 mm Hg), arterial Po2 (102.1 ± 3.3 to 80.5 ± 3.4 mm Hg), and renal medullary tissue Po2 (41 ± 5 to 24 ± 2 mm Hg) decreased, and plasma creatinine doubled (71 ± 2 to 144 ± 15 µmol/L) (all p < 0.01). Direct observation indicated that in all animals, sodium ascorbate dramatically improved the clinical state, from malaise and lethargy to a responsive, alert state within 3 hours. Body temperature (39.3 ± 0.3°C), heart rate (99.7 ± 3 beats/min), and plasma creatinine (32.6 ± 5.8 µmol/L) all decreased. Arterial (96.5 ± 2.5 mm Hg) and renal medullary Po2 (48 ± 5 mm Hg) increased. The norepinephrine dose was decreased, to zero in four of five sheep, whereas mean arterial pressure increased (to 83 ± 2 mm Hg). We confirmed these physiologic findings in a coronavirus disease 2019 patient with shock by compassionate use of 60 g of sodium ascorbate over 7 hours. CONCLUSIONS IV megadose sodium ascorbate reversed the pathophysiological and behavioral responses to Gram-negative sepsis without adverse side effects. Clinical studies are required to determine if such a dose has similar benefits in septic patients.
Collapse
Affiliation(s)
- Yugeesh R Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
- Centre for Integrated Critical Care, Department of Medicine and Radiology, University of Melbourne, VIC, Australia
| | - Rachel M Peiris
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Nobuki Okazaki
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
- Department of Anesthesiology and Resuscitology, Okayama University, Okayama, Japan
| | - Ian E Birchall
- Neuropathology Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Anton Trask-Marino
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Anthony Dornom
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Tom A M Vale
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Roger G Evans
- Department of Physiology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, VIC, Australia
| | - Fumitaka Yanase
- School of Medicine, University of Melbourne, VIC, Australia
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
| | - Rinaldo Bellomo
- Centre for Integrated Critical Care, Department of Medicine and Radiology, University of Melbourne, VIC, Australia
- School of Medicine, University of Melbourne, VIC, Australia
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
| | - Clive N May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
- Centre for Integrated Critical Care, Department of Medicine and Radiology, University of Melbourne, VIC, Australia
| |
Collapse
|
33
|
Abosamak MF, Lippi G, Benoit SW, Henry BM, Shama AAA. Bladder urine oxygen partial pressure monitoring: Could it be a tool for early detection of acute kidney injury? EGYPTIAN JOURNAL OF ANAESTHESIA 2021. [DOI: 10.1080/11101849.2021.1878686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Mohammed Fawzi Abosamak
- Department of Anesthesia and Intensive Care, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Giuseppe Lippi
- Department of Neuroscience, Section of Clinical Biochemistry, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Stefanie W. Benoit
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Brandon Michael Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | | |
Collapse
|
34
|
Jufar AH, Lankadeva YR, May CN, Cochrane AD, Bellomo R, Evans RG. Renal functional reserve: from physiological phenomenon to clinical biomarker and beyond. Am J Physiol Regul Integr Comp Physiol 2020; 319:R690-R702. [PMID: 33074016 DOI: 10.1152/ajpregu.00237.2020] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glomerular filtration rate (GFR) is acutely increased following a high-protein meal or systemic infusion of amino acids. The mechanisms underlying this renal functional response remain to be fully elucidated. Nevertheless, they appear to culminate in preglomerular vasodilation. Inhibition of the tubuloglomerular feedback signal appears critical. However, nitric oxide, vasodilator prostaglandins, and glucagon also appear important. The increase in GFR during amino acid infusion reveals a "renal reserve," which can be utilized when the physiological demand for single nephron GFR increases. This has led to the concept that in subclinical renal disease, before basal GFR begins to reduce, renal functional reserve can be recruited in a manner that preserves renal function. The extension of this concept is that once a decline in basal GFR can be detected, renal disease is already well progressed. This concept likely applies both in the contexts of chronic kidney disease and acute kidney injury. Critically, its corollary is that deficits in renal functional reserve have the potential to provide early detection of renal dysfunction before basal GFR is reduced. There is growing evidence that the renal response to infusion of amino acids can be used to identify patients at risk of developing either chronic kidney disease or acute kidney injury and as a treatment target for acute kidney injury. However, large multicenter clinical trials are required to test these propositions. A renewed effort to understand the renal physiology underlying the response to amino acid infusion is also warranted.
Collapse
Affiliation(s)
- Alemayehu H Jufar
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Yugeesh R Lankadeva
- Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Clive N May
- Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew D Cochrane
- Department of Cardiothoracic Surgery, Monash Health and Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Health, Heidelberg, Victoria, Australia
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|