101
|
Hu Q, Zhang Y, Xu H, Zhu L, Chen L, Hao C. Association between admission serum procalcitonin and the occurrence of acute kidney injury in patients with septic shock: A retrospective cohort study. Sci Prog 2021; 104:368504211043768. [PMID: 34558992 PMCID: PMC10361585 DOI: 10.1177/00368504211043768] [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: 11/17/2022]
Abstract
BACKGROUND Procalcitonin (PCT) is an effective and sensitive diagnostic biomarker that can facilitate the early detection of infection and septicemia, but whether it can similarly be utilized to predict the development of acute kidney injury (AKI) in patients suffering from septic shock remains to be established. Herein, the relationship between serum PCT at admission and the onset of AKI in septic shock patients was thus evaluated following adjustment for other potential covariates. METHODS This was a retrospective cohort study of 303 septic shock patients treated in a Chinese hospital between May 2015 and May 2019. All patients in whom PCT levels were measured on admission and who did not exhibit AKI or chronic kidney disease at the time of admission were assessed for AKI development within one week following intensive care unit (ICU) admission as per the KDIGO criteria. The relationship between serum PCT at admission and AKI incidence was then assessed for these patients. RESULTS These 303 patients were an average of 64 years old, and were 59.7% male. Of these patients, 50.5% developed AKI within the first 7 days following ICU admission. A dully-adjusted binary logistic regression analysis revealed PCT levels at admission to be associated with AKI following adjustment for potential confounding factors (odds ratio (OR) = 1.01, 95%CI (1.01,1.02), p = 0.0007). Receiver operating characteristic curve analysis further indicated that a PCT cutoff level of 52.59 ng/ml at admission was able to predict the incidence of AKI with respective sensitivity and specificity values of 50% and 84%. Interaction analysis revealed no significant interactive relationship between PCT and AKI, suggesting that serum PCT levels represent an early predictor of AKI incidence in septic shock patients. CONCLUSIONS Serum PCT at the time of admission can be used as a predictor of AKI in patients suffering from septic shock.
Collapse
Affiliation(s)
- Qinghe Hu
- Division of ICU, Affiliated Hospital of Jining Medical
University, Jining, Shandong, China
| | - Yaqing Zhang
- Division of ICU, Affiliated Hospital of Jining Medical
University, Jining, Shandong, China
| | - Hongying Xu
- Division of ICU, Affiliated Hospital of Jining Medical
University, Jining, Shandong, China
| | - Lina Zhu
- Division of ICU, Affiliated Hospital of Jining Medical
University, Jining, Shandong, China
| | - Lingzhi Chen
- Division of Nutrition, Affiliated Hospital of Jining Medical
University, Jining, Shandong, China
| | - Cuiping Hao
- Division of ICU, Affiliated Hospital of Jining Medical
University, Jining, Shandong, China
| |
Collapse
|
102
|
Affiliation(s)
- Paul M Palevsky
- Kidney Medicine Section, Medical Service, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania and Renal-Electrolye Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
103
|
Le S, Allen A, Calvert J, Palevsky PM, Braden G, Patel S, Pellegrini E, Green-Saxena A, Hoffman J, Das R. Convolutional Neural Network Model for Intensive Care Unit Acute Kidney Injury Prediction. Kidney Int Rep 2021; 6:1289-1298. [PMID: 34013107 PMCID: PMC8116756 DOI: 10.1016/j.ekir.2021.02.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/04/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) is common among hospitalized patients and has a significant impact on morbidity and mortality. Although early prediction of AKI has the potential to reduce adverse patient outcomes, it remains a difficult condition to predict and diagnose. The purpose of this study was to evaluate the ability of a machine learning algorithm to predict for AKI as defined by Kidney Disease: Improving Global Outcomes (KDIGO) stage 2 or 3 up to 48 hours in advance of onset using convolutional neural networks (CNNs) and patient electronic health record (EHR) data. METHODS A CNN prediction system was developed to use EHR data gathered during patients' stays to predict AKI up to 48 hours before onset. A total of 12,347 patient encounters were retrospectively analyzed from the Medical Information Mart for Intensive Care III (MIMIC-III) database. An XGBoost AKI prediction model and the sequential organ failure assessment (SOFA) scoring system were used as comparators. The outcome was AKI onset. The model was trained on routinely collected patient EHR data. Measurements included area under the receiver operating characteristic (AUROC) curve, positive predictive value (PPV), and a battery of additional performance metrics for advance prediction of AKI onset. RESULTS On a hold-out test set, the algorithm attained an AUROC of 0.86 and PPV of 0.24, relative to a cohort AKI prevalence of 7.62%, for long-horizon AKI prediction at a 48-hour window before onset. CONCLUSION A CNN machine learning-based AKI prediction model outperforms XGBoost and the SOFA scoring system, revealing superior performance in predicting AKI 48 hours before onset, without reliance on serum creatinine (SCr) measurements.
Collapse
Affiliation(s)
| | | | | | - Paul M. Palevsky
- VA Pittsburgh Healthcare System and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gregory Braden
- Baystate Medical Center, Springfield, Massachusetts, USA
| | - Sharad Patel
- Department of Critical Care Medicine, Cooper University Health Care, Camden, New Jersey, USA
| | | | | | | | | |
Collapse
|
104
|
The application of omic technologies to research in sepsis-associated acute kidney injury. Pediatr Nephrol 2021; 36:1075-1086. [PMID: 32356189 PMCID: PMC7606209 DOI: 10.1007/s00467-020-04557-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/25/2022]
Abstract
Acute kidney injury (AKI) is common in critically ill children and adults, and sepsis-associated AKI (SA-AKI) is the most frequent cause of AKI in the ICU. To date, no mechanistically targeted therapeutic interventions have been identified. High-throughput "omic" technologies (e.g., genomics, proteomics, metabolomics, etc.) offer a new angle of approach to achieve this end. In this review, we provide an update on the current understanding of SA-AKI pathophysiology. Omic technologies themselves are briefly discussed to facilitate interpretation of studies using them. We next summarize the body of SA-AKI research to date that has employed omic technologies. Importantly, omic studies are helping to elucidate a pathophysiology of SA-AKI centered around cellular stress responses, metabolic changes, and dysregulation of energy production that underlie its clinical features. Finally, we propose opportunities for future research using clinically relevant animal models, integrating multiple omic technologies and ultimately progressing to translational human studies focusing therapeutic strategies on targeted disease mechanisms.
Collapse
|
105
|
van Wijk XMR, Yun C, Lynch KL. Evaluation of Biomarkers in Sepsis: High Dimethylarginine (ADMA and SDMA) Concentrations Are Associated with Mortality. J Appl Lab Med 2021; 6:592-605. [PMID: 33382901 DOI: 10.1093/jalm/jfaa156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND As modulators of nitric oxide generation, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) may play important roles in sepsis. Current data on dimethylarginines are conflicting, and direct comparison data with other biomarkers are limited. METHODS Fifty-five patients were included in the final analysis and were divided into 4 groups: infection without sepsis, sepsis, severe sepsis, and septic shock. The first available samples on hospital admission were analyzed for ADMA, SDMA, procalcitonin (PCT), C-reactive protein, heparin binding protein (HBP), zonulin, soluble CD25 (sCD25), and soluble CD163 (sCD163). White blood cell (WBC) counts and lactate results were obtained from the medical record. RESULTS There were no statistically significant differences in ADMA and SDMA concentrations among the 4 groups; however, PCT, WBC, HBP, and sCD25 showed statistically significant differences. Lactate only trended toward statistical significance, likely because of limited availability in the medical record. Differences between survivors of sepsis and nonsurvivors at 30 days were highly statistically significant for ADMA and SDMA. Areas under the curve (AUCs) for ROC analysis were 0.88 and 0.95, respectively. There was also a statistically significant difference between survivors of sepsis and nonsurvivors for HBP, lactate, sCD25, and sCD163; however, AUCs for ROC curves were not statistically significantly different from 0.5. CONCLUSIONS Analysis of biomarkers other than dimethylarginines were in general agreement with expectations from the literature. ADMA and SDMA may not be specific markers for diagnosis of sepsis; however, they may be useful in short-term mortality risk assessment.
Collapse
Affiliation(s)
- Xander M R van Wijk
- Laboratory Medicine, University of California, San Francisco and Zuckerberg San Francisco General, Chicago, IL
| | - Cassandra Yun
- Laboratory Medicine, University of California, San Francisco and Zuckerberg San Francisco General, Chicago, IL
| | - Kara L Lynch
- Laboratory Medicine, University of California, San Francisco and Zuckerberg San Francisco General, Chicago, IL
| |
Collapse
|
106
|
Long-term Renal Outcomes in Adults With Sepsis-Induced Acute Kidney Injury: A Systematic Review. Dimens Crit Care Nurs 2021; 39:259-268. [PMID: 32740196 DOI: 10.1097/dcc.0000000000000432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Despite advances in medical technologies and intervention occurrences, acute kidney injury (AKI) incidence continues to rise. Early interventions after sepsis are essential to prevent AKI and its long-term consequences. Acute kidney injury is the leading cause of organ failure in sepsis; therefore, more research is needed on its long-term consequences and progression to kidney injury. OBJECTIVES The aim of this study was to review the state of the science on long-term renal outcomes after sepsis-induced AKI and long-term renal consequences. METHODS We identified research articles from PubMed and CINAHL databases using relevant key words for sepsis-induced AKI within 5 years delimited to full-text articles in English. RESULTS Among 1280 abstracts identified, we ultimately analyzed 12 full-text articles, identifying four common themes in the literature: (1) AKI determination criteria, (2) severity/prognosis-related factors, (3) time frame for long-term outcome measures, and (4) chronic kidney disease (CKD) and renal related exclusions. Researchers primarily used KDIGO (Kidney Disease: Improving Global Outcomes) guidelines to define AKI. All of these studies excluded patients with CKD. The range of time for long-term renal outcomes was 28 days to 3 years, with the majority being 1 year. Renal outcomes ranged from recovery to renal replacement therapy to death. CONCLUSIONS To better understand the long-term renal outcomes after sepsis-induced AKI, more consistent measures are needed across all studies regarding the time frame and specific renal outcomes. Because all of these articles excluded patients with CKD, a gap exists on long-term renal outcome in acute on CKD.
Collapse
|
107
|
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a common and life-threatening complication in hospitalized and critically ill patients. It is characterized by rapid deterioration of renal function associated with sepsis. The pathophysiology of S-AKI remains incompletely understood, so most therapies remain reactive and nonspecific. Possible pathogenic mechanisms to explain S-AKI include microcirculatory dysfunction, a dysregulated inflammatory response, and cellular metabolic reprogramming. In addition, several biomarkers have been developed in an attempt to improve diagnostic sensitivity and specificity of S-AKI. This article discusses the current understanding of S-AKI, recent advances in pathophysiology and biomarker development, and current preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Carlos L Manrique-Caballero
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA
| | - Gaspar Del Rio-Pertuz
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Hernando Gomez
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA.
| |
Collapse
|
108
|
Kellum JA, Artigas A, Gunnerson KJ, Honore PM, Kampf JP, Kwan T, McPherson P, Nguyen HB, Rimmelé T, Shapiro NI, Shi J, Vincent JL, Chawla LS. Use of Biomarkers to Identify Acute Kidney Injury to Help Detect Sepsis in Patients With Infection. Crit Care Med 2021; 49:e360-e368. [PMID: 33566467 PMCID: PMC7963439 DOI: 10.1097/ccm.0000000000004845] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Although early recognition of sepsis is vital to improving outcomes, the diagnosis may be missed or delayed in many patients. Acute kidney injury is one of the most common organ failures in patients with sepsis but may not be apparent on presentation. Novel biomarkers for acute kidney injury might improve organ failure recognition and facilitate earlier sepsis care. DESIGN Retrospective, international, Sapphire study. SETTING Academic Medical Center. PATIENTS Adults admitted to the ICU without evidence of acute kidney injury at time of enrollment. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We stratified patients enrolled in the Sapphire study into three groups-those with a clinical diagnosis of sepsis (n = 216), those with infection without sepsis (n = 120), and those without infection (n = 387) at enrollment. We then examined 30-day mortality stratified by acute kidney injury within each group. Finally, we determined the operating characteristics for kidney stress markers (tissue inhibitor of metalloproteinases-2) × (insulin-like growth factor binding protein 7) for prediction of acute kidney injury as a sepsis-defining organ failure in patients with infection without a clinical diagnosis of sepsis at enrollment. Combining all groups, 30-day mortality was 23% for patients who developed stage 2-3 acute kidney injury within the first 3 days compared with 14% without stage 2-3 acute kidney injury. However, this difference was greatest in the infection without sepsis group (34% vs 11%; odds ratio, 4.09; 95% CI, 1.53-11.12; p = 0.005). Using a (tissue inhibitor of metalloproteinases-2) × (insulin-like growth factor binding protein 7) cutoff of 2.0 units, 14 patients (11.7%), in the infection/no sepsis group, tested positive of which 10 (71.4%) developed stage 2-3 acute kidney injury. The positive test result occurred a median of 19 hours (interquartile range, 0.8-34.0 hr) before acute kidney injury manifested by serum creatinine or urine output. Similar results were obtained using a cutoff of 1.0 for any stage of acute kidney injury. CONCLUSIONS Use of the urinary (tissue inhibitor of metalloproteinases-2) × (insulin-like growth factor binding protein 7) test could identify acute kidney injury in patients with infection, possibly helping to detect sepsis, nearly a day before acute kidney injury is apparent by clinical criteria.
Collapse
Affiliation(s)
- John A Kellum
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Antonio Artigas
- Critical Care Center, Corporacion Sanitaria Parc Tauli, CIBER Enfermedades Respiratorias, Autonomous University of Barcelona, Sabadell, Spain
| | - Kyle J Gunnerson
- Departments of Emergency Medicine/Critical Care, Anesthesiology, Internal Medicine, Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
| | - Patrick M Honore
- Department of Intensive Care Medicine, Brugmann University Hospital, Brussels, Belgium
| | | | - Thomas Kwan
- Astute Medical, Inc. (a bioMérieux company), San Diego, CA
| | - Paul McPherson
- Astute Medical, Inc. (a bioMérieux company), San Diego, CA
| | - H Bryant Nguyen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA
| | - Thomas Rimmelé
- Department of Anesthesiology and Critical Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Nathan I Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Jing Shi
- Walker BioSciences, Carlsbad, CA
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Lakhmir S Chawla
- Department of Medicine, Veterans Affairs Medical Center, San Diego, CA
| |
Collapse
|
109
|
Abstract
Emerging evidence from observational studies suggests that both slower and faster net ultrafiltration rates during kidney replacement therapy are associated with increased mortality in critically ill patients with acute kidney injury and fluid overload. Faster rates are associated with ischemic organ injury. The net ultrafiltration rate should be prescribed based on patient body weight in milliliters per kilogram per hour, with close monitoring of patient hemodynamics and fluid balance. Randomized trials are required to examine whether moderate net ultrafiltration rates compared with slower and faster rates are associated with reduced risk of hemodynamic instability, organ injury, and improved outcomes.
Collapse
Affiliation(s)
- Vikram Balakumar
- Department of Critical Care Medicine, Mercy Hospitals, Springfield, MO, USA; Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. https://twitter.com/vikrambalakumar
| | - Raghavan Murugan
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Critical Care Medicine, The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) Center, University of Pittsburgh School of Medicine, University of Pittsburgh, 3347 Forbes Avenue, Suite 220, Room 206, Pittsburgh, PA 15261, USA.
| |
Collapse
|
110
|
An H, Hu Z, Chen Y, Cheng L, Shi J, Han L. Angiotensin II-mediated improvement of renal mitochondrial function via the AMPK/PGC-1α/NRF-2 pathway is superior to norepinephrine in a rat model of septic shock associated with acute renal injury. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:481. [PMID: 33850878 PMCID: PMC8039700 DOI: 10.21037/atm-21-621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background This study sought to compare the therapeutic effects of angiotensin II (ANG II) and norepinephrine (NE) on cecal ligation and puncture (CLP)-induced septic acute kidney injury (AKI) in rats. Methods Sepsis shock was induced in anesthesia Sprague-Dawley male rats by CLP model for 24 hours. A total of 40 rats were divided into five groups, including control group, sham group, CLP group, CLP + ANG II group, and CLP + NE group. CLP + ANG II and CLP + NE group were administration of ANG II or NE after sepsis shock respectively, maintaining the MAP at 75–85 mmHg. CLP group was administration of saline for contrast. At 0, 18, 24 hours measured the renal blood grades and resistant index (RI) by ultrasound equipment. At 6, 12, 18 and 24 hours collected 0.5 mL blood sample for creatinine and lactic acid examination. Rats were observed for 24 hours after CLP procedure and then sacrificed for subsequent examination, rat serum were used to determine the levels of inflammatory response factors, kidney tissues were used to examine the oxidative stress factors and mitochondrial related proteins.” We added the sentence as following: “The AMPK, PGC-1α and NRF-2 expression in renal cortex was significantly increased in the CLP + ANG II group. Results Compared to the vehicle treatment, both ANG II and NE administration restored the decrease in the mean arterial pressure (MAP) and alleviated mitochondrial impairments in CLP rats. However, only ANG II alleviated CLP-induced abnormalities in serum creatinine and lactic acid concentrations, renal blood flow, the renal resistant index, renal histopathology, the production of proinflammatory cytokines, and oxidative stress markers in rats. ANG II was also found to be superior to NE in reversing the CLP-induced suppression of mitochondrial biogenesis-related protein expression in the kidneys of rats. Conclusions ANG II was better than NE in alleviating CLP-induced septic AKI in rats.
Collapse
Affiliation(s)
- Hui An
- Department of Intensive Care Unit, Hebei Medical University, Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China.,Department of Intensive Care Unit, Baoding First Central Hospital, Baoding, China
| | - Zhenjie Hu
- Department of Intensive Care Unit, Hebei Medical University, Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Yuhong Chen
- Department of Intensive Care Unit, Hebei Medical University, Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Lianfang Cheng
- Department of Intensive Care Unit, Baoding First Central Hospital, Baoding, China
| | - Jian Shi
- Cardiovascular Surgery Department, Baoding First Central Hospital, Baoding, China
| | - Linan Han
- Department of Intensive Care Unit, Hebei Medical University, Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| |
Collapse
|
111
|
Funke BE, Jackson KE, Self WH, Collins SP, Saunders CT, Wang L, Blume JD, Wickersham N, Brown RM, Casey JD, Bernard GR, Rice TW, Siew ED, Semler MW. Effect of balanced crystalloids versus saline on urinary biomarkers of acute kidney injury in critically ill adults. BMC Nephrol 2021; 22:54. [PMID: 33546622 PMCID: PMC7863046 DOI: 10.1186/s12882-021-02236-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Recent trials have suggested use of balanced crystalloids may decrease the incidence of major adverse kidney events compared to saline in critically ill adults. The effect of crystalloid composition on biomarkers of early acute kidney injury remains unknown. METHODS From February 15 to July 15, 2016, we conducted an ancillary study to the Isotonic Solutions and Major Adverse Renal Events Trial (SMART) comparing the effect of balanced crystalloids versus saline on urinary levels of neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) among 261 consecutively-enrolled critically ill adults admitted from the emergency department to the medical ICU. After informed consent, we collected urine 36 ± 12 h after hospital admission and measured NGAL and KIM-1 levels using commercially available ELISAs. Levels of NGAL and KIM-1 at 36 ± 12 h were compared between patients assigned to balanced crystalloids versus saline using a Mann-Whitney U test. RESULTS The 131 patients (50.2%) assigned to the balanced crystalloid group and the 130 patients (49.8%) assigned to the saline group were similar at baseline. Urinary NGAL levels were significantly lower in the balanced crystalloid group (median, 39.4 ng/mg [IQR 9.9 to 133.2]) compared with the saline group (median, 64.4 ng/mg [IQR 27.6 to 339.9]) (P < 0.001). Urinary KIM-1 levels did not significantly differ between the balanced crystalloid group (median, 2.7 ng/mg [IQR 1.5 to 4.9]) and the saline group (median, 2.4 ng/mg [IQR 1.3 to 5.0]) (P = 0.36). CONCLUSIONS In this ancillary analysis of a clinical trial comparing balanced crystalloids to saline among critically ill adults, balanced crystalloids were associated with lower urinary concentrations of NGAL and similar urinary concentrations of KIM-1, compared with saline. These results suggest only a modest reduction in early biomarkers of acute kidney injury with use of balanced crystalloids compared with saline. TRIAL REGISTRATION ClinicalTrials.gov number: NCT02444988 . Date registered: May 15, 2015.
Collapse
Affiliation(s)
- Blake E Funke
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Karen E Jackson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, C-1216 MCN, 1161 21st Ave South, Nashville, TN, 37232, USA
| | - Wesley H Self
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sean P Collins
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christina T Saunders
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Li Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey D Blume
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nancy Wickersham
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, C-1216 MCN, 1161 21st Ave South, Nashville, TN, 37232, USA
| | - Ryan M Brown
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, C-1216 MCN, 1161 21st Ave South, Nashville, TN, 37232, USA
| | - Jonathan D Casey
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, C-1216 MCN, 1161 21st Ave South, Nashville, TN, 37232, USA
| | - Gordon R Bernard
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, C-1216 MCN, 1161 21st Ave South, Nashville, TN, 37232, USA
| | - Todd W Rice
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, C-1216 MCN, 1161 21st Ave South, Nashville, TN, 37232, USA
| | - Edward D Siew
- Division of Nephrology and Hypertension, Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for AKI (VIP-AKI), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew W Semler
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, C-1216 MCN, 1161 21st Ave South, Nashville, TN, 37232, USA.
| |
Collapse
|
112
|
Li X, Li J, Lu P, Li M. LINC00261 relieves the progression of sepsis-induced acute kidney injury by inhibiting NF-κB activation through targeting the miR-654-5p/SOCS3 axis. J Bioenerg Biomembr 2021; 53:129-137. [PMID: 33481135 DOI: 10.1007/s10863-021-09874-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
Sepsis is a life-threatening disease, which can cause the dysfunction of multiple organs, including kidney. Recently, a number of studies found that the long non-coding RNA (lncRNA) is closely associated with the development and progression of sepsis; however, the role of long intergenic non-protein coding RNA 261 (LINC00261) in sepsis-induced acute kidney injury is poorly understood. In this study, we found the expression of LINC00261 was significantly decreased in the serum of patients with sepsis than healthy controls. A similar result was also observed in the mouse model of sepsis induced by lipopolysaccharide (LPS). Further investigations revealed that overexpression of LINC00261 improved the viability, suppressed the apoptosis and reduced the generation of inflammatory cytokines in LPS-treated HK-2 cells. Mechanistically, we confirmed that LINC00261 could function as a sponge to combine with microRNA-654-5p (miR-654-5p) which inhibits nuclear factor-κB (NF-κB) activity by targeting suppressor of cytokine signaling 3 (SOCS3). In conclusion, our results demonstrate that LINC00261 may regulate the progression of sepsis-induced acute kidney injury via the miR-654-5p/SOCS3/NF-κB pathway and therefore provides a new insight into the treatment of this disease.
Collapse
Affiliation(s)
- Xinying Li
- Department of Emergency, Shandong Otolaryngological Hospital Affiliated to Shandong University, No.4 DuanXing West Road, Huaiyin District, Jinan, 250022, Shandong Province, China
| | - Jinying Li
- Department of Emergency, Shandong Otolaryngological Hospital Affiliated to Shandong University, No.4 DuanXing West Road, Huaiyin District, Jinan, 250022, Shandong Province, China
| | - Ping Lu
- Department of Emergency, The Fourth People's Hospital of Jinan, Jinan, 250031, Shandong Province, China
| | - Mingzhe Li
- Department of Emergency, Shandong Otolaryngological Hospital Affiliated to Shandong University, No.4 DuanXing West Road, Huaiyin District, Jinan, 250022, Shandong Province, China.
| |
Collapse
|
113
|
Abstract
PURPOSE OF REVIEW Acute kidney injury (AKI) is a common complication in critically ill patients. Understanding the pathophysiology of AKI is essential to guide patient management. Imaging techniques that inform the pathogenesis of AKI in critically ill patients are urgently needed, in both research and ultimately clinical settings. Renal contrast-enhanced ultrasonography (CEUS) and multiparametric MRI appear to be the most promising imaging techniques for exploring the pathophysiological mechanisms involved in AKI. RECENT FINDINGS CEUS and MRI can be used to noninvasively and safely evaluate renal macrocirculation and microcirculation and oxygenation in critical ill patients. These techniques show that a decrease in renal blood flow, particularly cortical blood flow, may be observed in septic AKI and may contribute to its development. MRI may be a valuable method to quantify long-term renal damage after AKI that cannot currently be detected using standard clinical approaches. SUMMARY CEUS and multiparametric renal MRI are promising imaging techniques but more evidence is needed to show how they can first be more widely used in a research setting to test key hypotheses about the pathophysiology and recovery of AKI, and then ultimately be adopted in clinical practice to guide patient management.
Collapse
Affiliation(s)
- Nicholas M Selby
- Centre for Kidney Research and Innovation, Division of Medical Sciences and Graduate Entry Medicine, University of Nottingham, UK
| | - Jacques Duranteau
- Department of Anesthesiology and Intensive Care, Paris-Saclay University, Bicêtre Hospital, Assistance Publique Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France
| |
Collapse
|
114
|
Gomez H, Priyanka P, Bataineh A, Keener CM, Clermont G, Kellum JA. Effects of 5% Albumin Plus Saline Versus Saline Alone on Outcomes From Large-Volume Resuscitation in Critically Ill Patients. Crit Care Med 2021; 49:79-90. [PMID: 33165027 PMCID: PMC7746571 DOI: 10.1097/ccm.0000000000004706] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To compare 5% albumin with 0.9% saline for large-volume resuscitation (> 60 mL/Kg within 24 hr), on mortality and development of acute kidney injury. DESIGN Retrospective cohort study. SETTING Patients admitted to ICUs in 13 hospitals across Western Pennsylvania. We analyzed two independent cohorts, the High-Density Intensive Care databases: High-Density Intensive Care-08 (July 2000 to October 2008, H08) and High-Density Intensive Care-15 (October 2008 to December 2014, H15). PATIENTS Total of 18,629 critically ill patients requiring large-volume resuscitation. INTERVENTIONS Five percent of albumin in addition to saline versus 0.9% saline. MEASUREMENTS AND MAIN RESULTS After excluding patients with acute kidney injury prior to large-volume resuscitation, 673 of 2,428 patients (27.7%) and 1,814 of 16,201 patients (11.2%) received 5% albumin in H08 and H15, respectively. Use of 5% albumin was associated with decreased 30-day mortality by multivariate regression in H08 (odds ratio 0.65; 95% CI 0.49-0.85; p = 0.002) and in H15 (0.52; 95% CI 0.44-0.62; p < 0.0001) but was associated with increased acute kidney injury in H08 (odds ratio 1.98; 95% CI 1.56-2.51; p < 0.001) and in H15 (odds ratio 1.75; 95% CI 1.58-1.95; p < 0.001). However, 5% albumin was not associated with persistent acute kidney injury and resulted in decreased major adverse kidney event at 30, 90, and 365 days. Propensity matched analysis confirmed similar associations with mortality and acute kidney injury. CONCLUSIONS During large-volume resuscitation, 5% albumin was associated with reduced mortality and major adverse kidney event at 30, 90, and 365 days. However, a higher rate of acute kidney injury of any stage was observed that did not translate into persistent renal dysfunction.
Collapse
Affiliation(s)
- Hernando Gomez
- The Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA
- CRISMA (Clinical Research Investigation and Systems Modeling of Acute illness) Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Priyanka Priyanka
- The Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA
| | - Ayham Bataineh
- The Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA
- Division of Nephrology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | | | - Gilles Clermont
- CRISMA (Clinical Research Investigation and Systems Modeling of Acute illness) Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
| | - John A. Kellum
- The Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA
- CRISMA (Clinical Research Investigation and Systems Modeling of Acute illness) Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
- Division of Nephrology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
115
|
Zhao H, Liang L, Pan S, Liu Z, Liang Y, Qiao Y, Liu D, Liu Z. Diabetes Mellitus as a Risk Factor for Progression from Acute Kidney Injury to Acute Kidney Disease: A Specific Prediction Model. Diabetes Metab Syndr Obes 2021; 14:2367-2379. [PMID: 34079315 PMCID: PMC8164678 DOI: 10.2147/dmso.s307776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Acute kidney injury is very common in hospitalized patients and carries a significant risk of mortality. Although timely intervention may improve patient prognosis, studies on the development of acute kidney disease in patients with acute kidney injury remain scarce. Thus, we constructed a prediction model to identify patients likely to develop acute kidney disease. PATIENTS AND METHODS Among 474 patients screened for eligibility, 261 were enrolled and randomly divided into training (185 patients) and independent validation cohorts (76 patients). Least absolute shrinkage and selection operator regression and multivariate logistic regression analyses were used to select features and build a nomogram incorporating the selected predictors: diabetes, anemia, oliguria, and peak creatinine. Calibration, discrimination, and the clinical usefulness of the model were assessed using calibration plots, the C-index, receiver operating characteristic curves, and decision curve analysis. RESULTS Diabetes was significantly associated with the presence of AKD. Peak creatinine, oliguria, and anemia also contributed to the progression of acute kidney injury. The model displayed good predictive power with a C-index of 0.834 and an AUC of 0.834 (95% confidence interval (CI): 0.773-0.895) in the training cohort and a C-index of 0.851 and an AUC of 0.851 (95% CI: 0.753-0.949) in the validation cohort. The calibration curves also showed that the model had a medium ability to predict acute kidney disease risk. Decision curve analysis showed that the nomogram was clinically useful when interventions were decided at the possibility threshold of 22%. CONCLUSION This novel prediction nomogram may allow for convenient prediction of acute kidney disease in patients with acute kidney injury, which may help to improve outcomes.
Collapse
Affiliation(s)
- Huanhuan Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Center for Kidney Disease, Zhengzhou, 450052, Henan Province, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People’s Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People’s Republic of China
| | - Lulu Liang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Center for Kidney Disease, Zhengzhou, 450052, Henan Province, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People’s Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People’s Republic of China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Zhenjie Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Yan Liang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Yingjin Qiao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Center for Kidney Disease, Zhengzhou, 450052, Henan Province, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People’s Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People’s Republic of China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Research Center for Kidney Disease, Zhengzhou, 450052, Henan Province, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People’s Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People’s Republic of China
- Correspondence: Zhangsuo Liu; Dongwei Liu Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People’s Republic of ChinaTel +86-0371-66295921 Email ;
| |
Collapse
|
116
|
Liu Z, Yang D, Gao J, Xiang X, Hu X, Li S, Wu W, Cai J, Tang C, Zhang D, Dong Z. Discovery and validation of miR-452 as an effective biomarker for acute kidney injury in sepsis. Theranostics 2020; 10:11963-11975. [PMID: 33204323 PMCID: PMC7667674 DOI: 10.7150/thno.50093] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/12/2020] [Indexed: 12/28/2022] Open
Abstract
Rationale: Sepsis is the cause of nearly half of acute kidney injury (AKI) and, unfortunately, AKI in sepsis is associated with unacceptably high rates of mortality. Early detection of AKI would guide the timely intervention and care of sepsis patients. Currently, NephroCheck, based on urinary [TIMP2]*[IGFBP7], is the only FDA approved test for early detection of AKI, which has a relatively low sensitivity for sepsis patients. Methods:In vitro, BUMPT (Boston University mouse proximal tubular cell line) cells were treated with lipopolysaccharides (LPS). In vivo, sepsis was induced in mice by LPS injection or cecal ligation and puncture (CLP). To validate the biomarker potential of miR-452, serum and urinary samples were collected from 47 sepsis patients with AKI, 50 patients without AKI, and 10 healthy subjects. Results: miR-452 was induced in renal tubular cells in septic AKI, and the induction was shown to be mediated by NF-κB. Notably, serum and urinary miR-452 increased early in septic mice following LPS or CLP treatment, prior to detectable renal dysfunction or tissue damage. Sepsis patients with AKI had significantly higher levels of serum and urinary miR-452 than the patients without AKI. Spearman's test demonstrated a remarkable positive correlation between urinary miR-452 and serum creatinine in sepsis patients (r=0.8269). The area under the receiver operating characteristic curve (AUC) was 0.8985 for urinary miR-452. Logistic regression analysis showed a striking 72.48-fold increase of AKI risk for every 1-fold increase of urinary miR-452 in sepsis patients. The sensitivity of urinary miR-452 for AKI detection in sepsis patients reached 87.23%, which was notably higher than the 61.54% achieved by urinary [TIMP2]*[IGFBP7], while the specificity of urinary miR-452 (78.00%) was slightly lower than that of [TIMP2]*[IGFBP7] (87.18%). Conclusions: miR-452 is induced via NF-κB in renal tubular cells in septic AKI. The increase of miR-452, especially that in urine, may be an effective biomarker for early detection of AKI in sepsis patients.
Collapse
|
117
|
Mertens C, Kuchler L, Sola A, Guiteras R, Grein S, Brüne B, von Knethen A, Jung M. Macrophage-Derived Iron-Bound Lipocalin-2 Correlates with Renal Recovery Markers Following Sepsis-Induced Kidney Damage. Int J Mol Sci 2020; 21:ijms21207527. [PMID: 33065981 PMCID: PMC7589935 DOI: 10.3390/ijms21207527] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 12/22/2022] Open
Abstract
During the course of sepsis in critically ill patients, kidney dysfunction and damage are among the first events of a complex scenario toward multi-organ failure and patient death. Acute kidney injury triggers the release of lipocalin-2 (Lcn-2), which is involved in both renal injury and recovery. Taking into account that Lcn-2 binds and transports iron with high affinity, we aimed at clarifying if Lcn-2 fulfills different biological functions according to its iron-loading status and its cellular source during sepsis-induced kidney failure. We assessed Lcn-2 levels both in serum and in the supernatant of short-term cultured renal macrophages (MΦ) as well as renal tubular epithelial cells (TEC) isolated from either Sham-operated or cecal ligation and puncture (CLP)-treated septic mice. Total kidney iron content was analyzed by Perls’ staining, while Lcn-2-bound iron in the supernatants of short-term cultured cells was determined by atomic absorption spectroscopy. Lcn-2 protein in serum was rapidly up-regulated at 6 h after sepsis induction and subsequently increased up to 48 h. Lcn-2-levels in the supernatant of TEC peaked at 24 h and were low at 48 h with no change in its iron-loading. In contrast, in renal MΦ Lcn-2 was low at 24 h, but increased at 48 h, where it mainly appeared in its iron-bound form. Whereas TEC-secreted, iron-free Lcn-2 was associated with renal injury, increased MΦ-released iron-bound Lcn-2 was linked to renal recovery. Therefore, we hypothesized that both the cellular source of Lcn-2 as well as its iron-load crucially adds to its biological function during sepsis-induced renal injury.
Collapse
Affiliation(s)
- Christina Mertens
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
| | - Laura Kuchler
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
| | - Anna Sola
- Department of Experimental Nephrology, IDIBELL, 08908 L’Hospitalet del Llobregat, Barcelona, Spain; (A.S.); (R.G.)
| | - Roser Guiteras
- Department of Experimental Nephrology, IDIBELL, 08908 L’Hospitalet del Llobregat, Barcelona, Spain; (A.S.); (R.G.)
| | - Stephan Grein
- Department of Mathematics, Temple University, Philadelphia, PA 19122, USA;
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
- Project Group Translational Medicine & Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 60590 Frankfurt am Main, Germany
| | - Andreas von Knethen
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
- Project Group Translational Medicine & Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 60590 Frankfurt am Main, Germany
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
- Correspondence:
| |
Collapse
|
118
|
Kellum JA, van Till JWO, Mulligan G. Targeting acute kidney injury in COVID-19. Nephrol Dial Transplant 2020; 35:1652-1662. [PMID: 33022712 PMCID: PMC7665651 DOI: 10.1093/ndt/gfaa231] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
As of 15 August 2020, Coronavirus disease 2019 (COVID-19) has been reported in >21 million people world-wide and is responsible for more than 750,000 deaths. The occurrence of acute kidney injury (AKI) in patients hospitalized with COVID-19 has been reported to be as high as 43%. This is comparable to AKI in other forms of pneumonia requiring hospitalization, as well as in non-infectious conditions like cardiac surgery. The impact of AKI on COVID-19 outcomes is difficult to assess at present but, similar to other forms of sepsis, AKI is strongly associated with hospital mortality. Indeed, mortality is reported to be very low in COVID-19 patients without AKI. Given that AKI contributes to fluid and acid-base imbalances, compromises immune response and may impair resolution of inflammation, it seems likely that AKI contributes to mortality in these patients. The pathophysiologic mechanisms of AKI in COVID-19 are thought to be multifactorial including systemic immune and inflammatory responses induced by viral infection, systemic tissue hypoxia, reduced renal perfusion, endothelial damage and direct epithelial infection with Severe Acute Respiratory Syndrome Coronavirus 2. Mitochondria play a central role in the metabolic deregulation in the adaptive response to the systemic inflammation and are also found to be vital in response to both direct viral damage and tissue reperfusion. These stress conditions are associated with increased glycolysis and reduced fatty acid oxidation. Thus, there is a strong rationale to target AKI for therapy in COVID-19. Furthermore, many approaches that have been developed for other etiologies of AKI such as sepsis, inflammation and ischemia-reperfusion, have relevance in the treatment of COVID-19 AKI and could be rapidly pivoted to this new disease.
Collapse
Affiliation(s)
- John A Kellum
- Department of Critical Care Medicine, The Center for Critical Care Nephology, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | |
Collapse
|
119
|
Yang B, Xie Y, Garzotto F, Ankawi G, Passannante A, Brendolan A, Bonato R, Carta M, Giavarina D, Vidal E, Gregori D, Ronco C. Influence of patients’ clinical features at intensive care unit admission on performance of cell cycle arrest biomarkers in predicting acute kidney injury. ACTA ACUST UNITED AC 2020; 59:333-342. [DOI: 10.1515/cclm-2020-0670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/11/2020] [Indexed: 12/23/2022]
Abstract
Abstract
Objectives
Identification of acute kidney injury (AKI) can be challenging in patients with a variety of clinical features at intensive care unit (ICU) admission, and the capacity of biomarkers in this subpopulation has been poorly studied. In our study we examined the influence that patients’ clinical features at ICU admission have over the predicting ability of the combination of urinary tissue inhibitor of metalloproteinase-2 (TIMP2) and insulin-like growth factor binding protein 7 (IGFBP7).
Methods
Urinary [TIMP2]•[IGFBP7] were measured for all patients upon admission to ICU. We calculated the receiver operating characteristics (ROC) curves for AKI prediction in the overall cohort and for subgroups of patients according to etiology of ICU admission, which included: sepsis, trauma, neurological conditions, cardiovascular diseases, respiratory diseases, and non-classifiable causes.
Results
In the overall cohort of 719 patients, 239 (33.2%) developed AKI in the first seven days. [TIMP2]•[IGFBP7] at ICU admission were significantly higher in AKI patients than in non-AKI patients. This is true not only for the overall cohort but also in the other subgroups. The area under the ROC curve (AUC) for [TIMP2]•[IGFBP7] in predicting AKI in the first seven days was 0.633 (95% CI 0.588–0.678), for the overall cohort, with sensitivity and specificity of 66.1 and 51.9% respectively. When we considered patients with combined sepsis, trauma, and respiratory disease we found a higher AUC than patients without these conditions (0.711 vs. 0.575; p=0.002).
Conclusions
The accuracy of [TIMP2]•[IGFBP7] in predicting the risk of AKI in the first seven days after ICU admission has significant variability when the reason for ICU admission is considered.
Collapse
Affiliation(s)
- Bo Yang
- Department of Nephrology , First Teaching Hospital of Tianjin University of Traditional Chinese Medicine , Tianjin , PR China
- International Renal Research Institute of Vicenza , San Bortolo Hospital , Vicenza , Italy
| | - Yun Xie
- International Renal Research Institute of Vicenza , San Bortolo Hospital , Vicenza , Italy
- Department of Nephrology , Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine , Shanghai , P.R. China
| | - Francesco Garzotto
- International Renal Research Institute of Vicenza , San Bortolo Hospital , Vicenza , Italy
- Biostatistics, Epidemiology and Public Health Unit, Department of Cardiac, Thoracic and Vascular Sciences , University of Padova , Padova , Italy
| | - Ghada Ankawi
- International Renal Research Institute of Vicenza , San Bortolo Hospital , Vicenza , Italy
- Department of Internal Medicine and Nephrology , King Abdulaziz University , Jeddah , Saudi Arabia
| | - Alberto Passannante
- International Renal Research Institute of Vicenza , San Bortolo Hospital , Vicenza , Italy
- Department of Anaesthesia and Intensive Care , University of Trieste , Trieste , Italy
| | - Alessandra Brendolan
- International Renal Research Institute of Vicenza , San Bortolo Hospital , Vicenza , Italy
- Department of Nephrology, Dialysis and Transplantation , San Bortolo Hospital , Vicenza , Italy
| | - Raffaele Bonato
- Department of Intensive Care , San Bortolo Hospital , Vicenza , Italy
| | - Mariarosa Carta
- Department of Laboratory Medicine , San Bortolo Hospital , Vicenza , Italy
| | - Davide Giavarina
- Department of Laboratory Medicine , San Bortolo Hospital , Vicenza , Italy
| | - Enrico Vidal
- Nephrology, Dialysis and Transplant Unit, Department of Woman’s and Child’s Health , University-Hospital of Padova , Padova , Italy
| | - Dario Gregori
- Biostatistics, Epidemiology and Public Health Unit, Department of Cardiac, Thoracic and Vascular Sciences , University of Padova , Padova , Italy
| | - Claudio Ronco
- International Renal Research Institute of Vicenza , San Bortolo Hospital , Vicenza , Italy
- Department of Nephrology, Dialysis and Transplantation , San Bortolo Hospital , Vicenza , Italy
- Department of Medicine , University of Padova , Padova , Italy
| |
Collapse
|
120
|
Xu L, Hu G, Xing P, Zhou M, Wang D. Paclitaxel alleviates the sepsis-induced acute kidney injury via lnc-MALAT1/miR-370-3p/HMGB1 axis. Life Sci 2020; 262:118505. [PMID: 32998017 DOI: 10.1016/j.lfs.2020.118505] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022]
Abstract
AIMS To investigate the effects of paclitaxel on lipopolysaccharide (LPS)-induced acute kidney injury (AKI) and its related mechanisms. MAIN METHODS The sepsis-associated AKI was induced by LPS using HK-2 cells. Then the mRNA and protein expression levels of relevant genes in the serum of sepsis patients and HK-2 cells with LPS-induced AKI were detected by qRT-PCR and western blot analyses before and after paclitaxel treatment, respectively. Subsequently, the cell counting kit-8 (CCK-8) and flow cytometry assays were performed to estimate the effects of paclitaxel, lnc-MALAT1, miR-370-3p and HMGB1 on the proliferation and apoptosis of HK-2 cells injured by LPS. KEY FINDINGS Lnc-MALAT1 was increased both in the serum of sepsis patients and cells injured by LPS, which could inhibit the cell proliferation, promote the cell apoptosis and increase the expression of TNF-α, IL-6 and IL-1β caused by paclitaxel. Moreover, lnc-MALAT1 was sponged with miR-370-3p which had the inverse effects with lnc-MALAT1 in LPS induced HK-2 cells. What's more, miR-370-3p targeted HMGB1 which was induced in serum and cells of sepsis. Knockdown of miR-370-3p inhibited the expression of HMGB1 and suppressed the proliferation but promoted the apoptosis of HK-2 cells injured by LPS as well as the expression of TNF-α, IL-6 and IL-1β. Besides, paclitaxel restrained the expression of HMGB1 via regulating lnc-MALAT1/miR-370-3p axis. SIGNIFICANCE Paclitaxel could protect against LPS-induced AKI via the regulation of lnc-MALAT1/miR-370-3p/HMGB1 axis and the expression of TNF-α, IL-6 and IL-1β, revealing that paclitaxel might act as a therapy drug in reducing sepsis-associated AKI.
Collapse
Affiliation(s)
- Lina Xu
- Department of Infectious Diseases, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| | - Guyong Hu
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| | - Pengcheng Xing
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China.
| | - Minjie Zhou
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| | - Donglian Wang
- Department of Emergency, Shanghai University of Medicine & Health Sciences Affiliated Shanghai Sixth People's Hospital East, No. 222, West Three Road Around Lake, Pudong District, Shanghai 201306, PR China
| |
Collapse
|
121
|
Karnuta J, Featherall J, Lawrenz J, Gordon J, Golubovsky J, Thomas J, Ramanathan D, Simpfendorfer C, Nystrom LM, Babic M, Mesko NW. What Demographic and Clinical Factors Are Associated with In-hospital Mortality in Patients with Necrotizing Fasciitis? Clin Orthop Relat Res 2020; 478:1770-1779. [PMID: 32168071 PMCID: PMC7371088 DOI: 10.1097/corr.0000000000001187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/05/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Necrotizing fasciitis is a rare infection with rapid deterioration and a high mortality rate. Factors associated with in-hospital mortality have not been thoroughly evaluated. Although predictive models identifying the diagnosis of necrotizing fasciitis have been described (such as the Laboratory Risk Indicator for Necrotizing Fasciitis [LRINEC]), their use in predicting mortality is limited. QUESTIONS/PURPOSES (1) What demographic factors are associated with in-hospital mortality in patients with necrotizing fasciitis? (2) What clinical factors are associated with in-hospital mortality? (3) What laboratory values are associated with in-hospital mortality? (4) Is the LRINEC score useful in predicting mortality? METHODS We retrospectively studied all patients with necrotizing fasciitis at our tertiary care institution during a 10-year period. In all, 134 patients were identified; after filtering out patients with missing data (seven) and those without histologically confirmed necrotizing fasciitis (12), 115 patients remained. These patients were treated with early-initiation antibiotic therapy and aggressive surgical intervention once the diagnosis was suspected. Demographic data, clinical features, laboratory results, and treatment variables were identified. The median age was 56 years and 42% of patients were female. Of the 115 patients analyzed, 15% (17) died in the hospital. Univariate and receiver operating characteristic analyses were performed due to the low number of mortality events seen in this cohort. RESULTS The demographic factors associated with in-hospital mortality were older age (median: 64 years for nonsurvivors [interquartile range (IQR) 57-79] versus 55 years for survivors [IQR 45-63]; p = 0.002), coronary artery disease (odds ratio 4.56 [95% confidence interval (CI) 1.51 to 14]; p = 0.008), chronic kidney disease (OR 4.92 [95% CI 1.62 to 15]; p = 0.006), and transfer from an outside hospital (OR 3.47 [95% CI 1.19 to 10]; p = 0.02). The presenting clinical characteristics associated with in-hospital mortality were positive initial blood culture results (OR 4.76 [95% CI 1.59 to 15]; p = 0.01), lactic acidosis (OR 4.33 [95% CI 1.42 to 16]; p = 0.02), and multiple organ dysfunction syndrome (OR 6.37 [95% CI 2.05 to 20]; p = 0.002). Laboratory values at initial presentation that were associated with in-hospital mortality were platelet count (difference of medians -136 [95% CI -203 to -70]; p < 0.001), serum pH (difference of medians -0.13 [95% CI -0.21 to -0.03]; p = 0.02), serum lactate (difference of medians 0.90 [95% CI 0.40 to 4.80]; p < 0.001), serum creatinine (difference of medians 1.93 [95% CI 0.65 to 3.44]; p < 0.001), partial thromboplastin time (difference of medians 8.30 [95% CI 1.85 to 13]; p = 0.03), and international normalized ratio (difference of medians 0.1 [95% CI 0.0 to 0.5]; p = 0.004). The LRINEC score was a poor predictor of mortality with an area under the receiver operating characteristics curve of 0.56 [95% CI 0.45-0.67]. CONCLUSIONS Factors aiding clinical recognition of necrotizing fasciitis are not consistently helpful in predicting mortality of this infection. Identifying patients with potentially compromised organ function should lead to aggressive and expedited measures for diagnosis and treatment. Future multicenter studies with larger populations and a standardized algorithm of treatment triggered by high clinical suspicion can be used to validate these findings to better help prognosticate this potentially fatal diagnosis.Level of Evidence Level III, therapeutic study.
Collapse
Affiliation(s)
- Jaret Karnuta
- J. Karnuta, J. Golubovsky, J. Thomas, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Joseph Featherall
- J. Featherall, J. Lawrenz, J. Gordon, D. Ramanathan, L. M. Nystrom, N. W. Mesko, Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joshua Lawrenz
- J. Featherall, J. Lawrenz, J. Gordon, D. Ramanathan, L. M. Nystrom, N. W. Mesko, Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jaymeson Gordon
- J. Featherall, J. Lawrenz, J. Gordon, D. Ramanathan, L. M. Nystrom, N. W. Mesko, Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joshua Golubovsky
- J. Karnuta, J. Golubovsky, J. Thomas, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Jonah Thomas
- J. Karnuta, J. Golubovsky, J. Thomas, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Deepak Ramanathan
- J. Featherall, J. Lawrenz, J. Gordon, D. Ramanathan, L. M. Nystrom, N. W. Mesko, Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Claus Simpfendorfer
- C. Simpfendorfer, Department of Diagnostic Radiology, Cleveland Clinic, Cleveland, OH, USA
| | - Lukas M Nystrom
- J. Featherall, J. Lawrenz, J. Gordon, D. Ramanathan, L. M. Nystrom, N. W. Mesko, Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Maja Babic
- M. Babic, Department of Infectious Disease, Cleveland Clinic, Cleveland, OH, USA
| | - Nathan W Mesko
- J. Featherall, J. Lawrenz, J. Gordon, D. Ramanathan, L. M. Nystrom, N. W. Mesko, Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
122
|
Ostermann M, Bellomo R, Burdmann EA, Doi K, Endre ZH, Goldstein SL, Kane-Gill SL, Liu KD, Prowle JR, Shaw AD, Srisawat N, Cheung M, Jadoul M, Winkelmayer WC, Kellum JA. Controversies in acute kidney injury: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Conference. Kidney Int 2020; 98:294-309. [PMID: 32709292 PMCID: PMC8481001 DOI: 10.1016/j.kint.2020.04.020] [Citation(s) in RCA: 305] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
In 2012, Kidney Disease: Improving Global Outcomes (KDIGO) published a guideline on the classification and management of acute kidney injury (AKI). The guideline was derived from evidence available through February 2011. Since then, new evidence has emerged that has important implications for clinical practice in diagnosing and managing AKI. In April of 2019, KDIGO held a controversies conference entitled Acute Kidney Injury with the following goals: determine best practices and areas of uncertainty in treating AKI; review key relevant literature published since the 2012 KDIGO AKI guideline; address ongoing controversial issues; identify new topics or issues to be revisited for the next iteration of the KDIGO AKI guideline; and outline research needed to improve AKI management. Here, we present the findings of this conference and describe key areas that future guidelines may address.
Collapse
Affiliation(s)
- Marlies Ostermann
- Department of Critical Care, King's College London, Guy's & St. Thomas' Hospital, King's College London, London, UK.
| | - Rinaldo Bellomo
- Centre for Integrated Critical Care, The University of Melbourne, Melbourne, Victoria, Australia
| | - Emmanuel A Burdmann
- Laboratório de Investigação Médica 12, Division of Nephrology, University of Sao Paulo Medical School, Sao Paulo, Sao Paulo, Brazil
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, The University of Tokyo, Tokyo, Japan
| | - Zoltan H Endre
- Prince of Wales Hospital and Clinical School, University of New South Wales, Randwick, NSW, Australia
| | - Stuart L Goldstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Sandra L Kane-Gill
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Kathleen D Liu
- Department of Medicine, Division of Nephrology, University of California, San Francisco, San Francisco, California, USA; Department of Anesthesia, Division of Critical Care Medicine, University of California, San Francisco, San Francisco, California, USA
| | - John R Prowle
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Andrew D Shaw
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Nattachai Srisawat
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Critical Care Nephrology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Tropical Medicine Cluster, Chulalongkorn University, Bangkok, Thailand; Excellence Center for Critical Care Nephrology, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Academy of Science, Royal Society of Thailand, Bangkok, Thailand
| | - Michael Cheung
- Kidney Disease: Improving Global Outcomes (KDIGO), Brussels, Belgium
| | - Michel Jadoul
- Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Wolfgang C Winkelmayer
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - John A Kellum
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
123
|
Fiorentino M, Xu Z, Smith A, Singbartl K, Palevsky PM, Chawla LS, Huang DT, Yealy DM, Angus DC, Kellum JA. Serial Measurement of Cell-cycle Arrest Biomarkers [TIMP-2]•[IGFBP7] and Risk for Progression to Death, Dialysis or Severe Acute Kidney Injury in Patients with Septic Shock. Am J Respir Crit Care Med 2020; 202:1262-1270. [PMID: 32584598 DOI: 10.1164/rccm.201906-1197oc] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
RATIONALE Urinary tissue inhibitor of metalloproteinases-2 (TIMP-2) and insulin-like growth factor-binding protein 7 (IGFBP7) can predict AKI in patients with sepsis. OBJECTIVES Since most sepsis patients present with AKI, critical questions are whether biomarkers can inform on the response to treatment and whether they might be used to guide therapy. METHODS We measured [TIMP-2]•[IGFBP7] before and after a 6-hour resuscitation in 688 patients with septic shock enrolled in the ProCESS trial. Our primary endpoint was stage 3 AKI, renal replacement therapy or death within 7 days. MEASUREMENTS AND MAIN RESULTS The endpoint was reached in 113 patients (16.4%). In patients with negative [TIMP-2]•[IGFBP7] at baseline, those who became positive (>0.3 units) after resuscitation had 3-times higher risk compared to those who remained negative (21.8% vs 8.5%, p=0.01; OR 3.0, 95%CI 1.31-6.87). Conversely, compared to patients with a positive biomarker at baseline that were still positive at hour 6, risk was reduced for patients who became negative (23.8% vs 9.8%, p=0.01; OR 2.15, 95%CI 1.17-3.95). A positive [TIMP-2]•[IGFBP7] following resuscitation was associated with worse outcomes in both patients with and without AKI at that time point. Clinical response to resuscitation, as judged by APACHE II score, was weakly predictive of the endpoint (AUC 0.68, 95%CI 0.62-0.73) and improved with addition of [TIMP-2]•[IGFBP7] (0.72, 95%CI 0.66-0.77 p=0.03). Different resuscitation protocols did not alter biomarker trajectories, nor outcomes in biomarker positive or negative patients. However, biomarker trajectories were associated with outcome. CONCLUSIONS Changes in urinary [TIMP-2]•[IGFBP7] following initial fluid resuscitation identify sepsis patients with differing risk for progression of AKI. Clinical trial registration available at www.clinicaltrials.gov, ID: NCT00510835.
Collapse
Affiliation(s)
- Marco Fiorentino
- University of Pittsburgh, Center for Critical Care Nephrology; Department of Critical Care Medicine, Pittsburgh, Pennsylvania, United States.,University of Bari, Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, Bari, Italy
| | - Zhongying Xu
- University of Pittsburgh Graduate School of Public Health, 51303, Pittsburgh, Pennsylvania, United States
| | - Ali Smith
- University of Pittsburgh, Center for Critical Care Nephrology; Department of Critical Care Medicine, Pittsburgh, Pennsylvania, United States
| | - Kai Singbartl
- University of Pittsburgh, Center for Critical Care Nephrology; Department of Critical Care Medicine, Pittsburgh, Pennsylvania, United States.,Mayo Clinic, Critical Care Medicine, Phoenix, Arizona, United States
| | - Paul M Palevsky
- University of Pittsburgh, 6614, Center for Critical Care Nephrology; Department of Critical Care Medicine, Pittsburgh, Pennsylvania, United States.,Pittsburgh VA Medical Center, Renal Section, Medical Service, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Renal-Electrolyte Division, Pittsburgh, Pennsylvania, United States
| | | | - David T Huang
- University of Pittsburgh, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Critical Care Medicine, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, 6614, Emergency Medicine, Pittsburgh, Pennsylvania, United States
| | - Donald M Yealy
- University of Pittsburgh, Emergency Medicine, Pittsburgh, Pennsylvania, United States
| | - Derek C Angus
- University of Pittsburgh, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Critical Care Medicine, Pittsburgh, Pennsylvania, United States
| | - John A Kellum
- University of Pittsburgh, Center for Critical Care Nephrology; Department of Critical Care Medicine, Pittsburgh, Pennsylvania, United States;
| | | |
Collapse
|
124
|
Uhel F, Peters-Sengers H, Falahi F, Scicluna BP, van Vught LA, Bonten MJ, Cremer OL, Schultz MJ, van der Poll T. Mortality and host response aberrations associated with transient and persistent acute kidney injury in critically ill patients with sepsis: a prospective cohort study. Intensive Care Med 2020; 46:1576-1589. [PMID: 32514599 PMCID: PMC7381452 DOI: 10.1007/s00134-020-06119-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/14/2020] [Indexed: 12/26/2022]
Abstract
Purpose Sepsis is the most frequent cause of acute kidney injury (AKI). The “Acute Disease Quality Initiative Workgroup” recently proposed new definitions for AKI, classifying it as transient or persistent. We investigated the incidence, mortality, and host response aberrations associated with transient and persistent AKI in sepsis patients. Methods A total of 1545 patients admitted with sepsis to 2 intensive care units in the Netherlands were stratified according to the presence (defined by any urine or creatinine RIFLE criterion within the first 48 h) and evolution of AKI (with persistent defined as remaining > 48 h). We determined 30-day mortality by logistic regression adjusting for confounding variables and analyzed 16 plasma biomarkers reflecting pathways involved in sepsis pathogenesis (n = 866) and blood leukocyte transcriptomes (n = 392). Results AKI occurred in 37.7% of patients, of which 18.4% was transient and 81.6% persistent. On admission, patients with persistent AKI had higher disease severity scores and more frequently had severe (injury or failure) RIFLE AKI stages than transient AKI patients. Persistent AKI, but not transient AKI, was associated with increased mortality by day 30 and up to 1 year. Persistent AKI was associated with enhanced and sustained inflammatory and procoagulant responses during the first 4 days, and a more severe loss of vascular integrity compared with transient AKI. Baseline blood gene expression showed minimal differences with respect to the presence or evolution of AKI. Conclusion Persistent AKI is independently associated with sepsis mortality, as well as with sustained inflammatory and procoagulant responses, and loss of vascular integrity as compared with transient AKI. Electronic supplementary material The online version of this article (10.1007/s00134-020-06119-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fabrice Uhel
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Room G2-130; Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Hessel Peters-Sengers
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Room G2-130; Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Fahimeh Falahi
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Room G2-130; Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Brendon P Scicluna
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Room G2-130; Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lonneke A van Vught
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Room G2-130; Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Marc J Bonten
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcus J Schultz
- Department of Intensive Care Medicine, and Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand
- Nuffield Department of medicine, University of Oxford, Oxford, UK
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Room G2-130; Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
125
|
Shen R, Zhang W, Ming S, Li L, Peng Y, Gao X. Gender-related differences in the performance of sequential organ failure assessment (SOFA) to predict septic shock after percutaneous nephrolithotomy. Urolithiasis 2020; 49:65-72. [PMID: 32372319 DOI: 10.1007/s00240-020-01190-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
The study aims to identify whether gender differences exist in the sequential organ failure assessment (SOFA) score to the extent of affecting its predictive accuracy for septic shock after percutaneous nephrolithotomy (PCNL). A retrospective study of 612 patients undergoing PCNL was performed. The SOFA scores of male and female groups were compared to identify any gender differences. The ROC curve was used to find differences between the original and adjusted SOFA scores. Postoperative septic shock developed in 21 (3.43%) cases. A marginally significant discrepancy in median SOFA scores between genders was discovered in a subgroup of patients < 40 years old (p = 0.048). A gender difference existed in the SOFA score after PCNL, with greater proportion of high scores in female patients (p = 0.011). Male patients had a higher proportion of ≥ 2 sub-score in hepatic and renal systems than female patients, caused by their higher preoperative bilirubin and creatinine (p < 0.05). An adjusted SOFA score was created to replace the original postoperative SOFA score with the perioperative changed values of bilirubin and creatinine. Performance of the adjusted SOFA score for predicting septic shock was comparable with the original SOFA score (AUC 0.987 vs. 0.985, p = 0.932). Under the premise of ensuring 100% sensitivity, the adjusted SOFA score reduced the 43.7% (31/71) false-positive rate for predicting septic shock compared with the original SOFA score. In conclusion, the gender should not be neglected when applying SOFA score for patients after PCNL. The adjusted SOFA score eliminates negative effects caused by gender differences in predicting septic shock.
Collapse
Affiliation(s)
- Rong Shen
- Department of Urology, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Wei Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Shaoxiong Ming
- Department of Urology, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Ling Li
- Department of Urology, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Yonghan Peng
- Department of Urology, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China.
| | - Xiaofeng Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China.
| |
Collapse
|
126
|
Jin K, Ma Y, Manrique-Caballero CL, Li H, Emlet DR, Li S, Baty CJ, Wen X, Kim-Campbell N, Frank A, Menchikova EV, Pastor-Soler NM, Hallows KR, Jackson EK, Shiva S, Pinsky MR, Zuckerbraun BS, Kellum JA, Gómez H. Activation of AMP-activated protein kinase during sepsis/inflammation improves survival by preserving cellular metabolic fitness. FASEB J 2020; 34:7036-7057. [PMID: 32246808 PMCID: PMC11956121 DOI: 10.1096/fj.201901900r] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/24/2020] [Accepted: 03/18/2020] [Indexed: 01/08/2023]
Abstract
The purpose was to determine the role of AMPK activation in the renal metabolic response to sepsis, the development of sepsis-induced acute kidney injury (AKI) and on survival. In a prospective experimental study, 167 10- to 12-week-old C57BL/6 mice underwent cecal ligation and puncture (CLP) and human proximal tubule epithelial cells (TEC; HK2) were exposed to inflammatory mix (IM), a combination of lipopolysaccharide (LPS) and high mobility group box 1 (HMGB1). Renal/TEC metabolic fitness was assessed by monitoring the expression of drivers of oxidative phosphorylation (OXPHOS), the rates of utilization of OXPHOS/glycolysis in response to metabolic stress, and mitochondrial function by measuring O2 consumption rates (OCR) and the membrane potential (Δψm ). Sepsis/IM resulted in AKI, increased mortality, and in renal AMPK activation 6-24 hours after CLP/IM. Pharmacologic activation of AMPK with 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) or metformin during sepsis improved the survival, while AMPK inhibition with Compound C increased mortality, impaired mitochondrial respiration, decreased OCR, and disrupted TEC metabolic fitness. AMPK-driven protection was associated with increased Sirt 3 expression and restoration of metabolic fitness. Renal AMPK activation in response to sepsis/IM is an adaptive mechanism that protects TEC, organs, and the host by preserving mitochondrial function and metabolic fitness likely through Sirt3 signaling.
Collapse
Affiliation(s)
- Kui Jin
- Department of Critical Care, Anhui Provincial Hospital, He Fei, China
| | - Yujie Ma
- Department of Critical Care Medicine, Air Force Medical Center, Beijing, China
| | - Carlos L Manrique-Caballero
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hui Li
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - David R Emlet
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shengnan Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Catherine J Baty
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoyan Wen
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nahmah Kim-Campbell
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alicia Frank
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Elizabeth V Menchikova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nuria M Pastor-Soler
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Kenneth R Hallows
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael R Pinsky
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian S Zuckerbraun
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hernando Gómez
- Center for Critical Care Nephrology, Department of Critical Care Medicine, The CRISMA Center, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
127
|
Chacko B. Kidney Injury in Sepsis: Fuel to the Fire. Indian J Crit Care Med 2020; 24:216-217. [PMID: 32565629 PMCID: PMC7297247 DOI: 10.5005/jp-journals-10071-23414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
How to cite this article: Chacko B. Kidney Injury in Sepsis: Fuel to the Fire. Indian J Crit Care Med 2020;24(4):216-217.
Collapse
Affiliation(s)
- Binila Chacko
- Medical Intensive Care Unit, Division of Critical Care, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
128
|
Peerapornratana S, Priyanka P, Wang S, Smith A, Singbartl K, Palevsky PM, Chawla LS, Yealy DM, Angus DC, Kellum JA. Sepsis-Associated Acute Kidney Disease. Kidney Int Rep 2020; 5:839-850. [PMID: 32518866 PMCID: PMC7270721 DOI: 10.1016/j.ekir.2020.03.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 02/10/2020] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction About one-third of critically ill patients with acute kidney injury (AKI) develop persistently decreased kidney function, known as acute kidney disease (AKD), which may progress to chronic kidney disease (CKD). Although sepsis is the most common cause of AKI, little is known about sepsis-associated AKD. Methods Using data from a large randomized trial including 1341 patients with septic shock, we studied patients with stage 2 or 3 AKI on day 1 of hospitalization. We defined AKD as a persistently reduced glomerular filtration rate for >7 days. In addition to clinical data, we measured several urinary biomarkers (tissue inhibitor of metalloproteinases-2 and insulin-like growth factor-binding protein 7 [TIMP-2∗IGFBP7], neutrophil gelatinase-associated lipocalin [NGAL], kidney injury molecule-1 [KIM-1], liver-type fatty acid binding protein, and type 4 collagen) at 0, 6, and 24 hours, to predict AKD. Results Of 598 patients, 119 (19.9%) died within 7 days, 318 (53.2%) had early reversal of AKI within the first 7 days, whereas 161 (26.9%) developed AKD. In patients with early reversal, 45 (14.2%) had relapsed AKI after early reversal, and only about one-third of these recovered. Among patients developing AKD, only 15 (9.3%) recovered renal function prior to discharge. Male sex, African American race, and underlying CKD were more predominant in patients developing AKD. None of the biomarkers tested performed well for prediction of AKD, although NGAL modestly increased the performance of a clinical model. Conclusions AKD is common in patients with septic shock, especially among African American males and those with underlying CKD. Existing AKI biomarkers have limited utility for predicting AKD but might be useful together with clinical variables. Novel predictive biomarkers for renal recovery are needed.
Collapse
Affiliation(s)
- Sadudee Peerapornratana
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Excellence Center for Critical Care Nephrology, Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Critical Care Nephrology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Priyanka Priyanka
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shu Wang
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,University of Florida Health Cancer Center, Gainesville, Florida, USA.,Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Ali Smith
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kai Singbartl
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, Mayo Clinic Hospital, Phoenix, Arizona, USA
| | - Paul M Palevsky
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Renal Section, Medical Service, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA.,Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lakhmir S Chawla
- Department of Medicine, Veterans Affairs Medical Center, San Diego, California, USA
| | - Donald M Yealy
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Derek C Angus
- The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
129
|
|
130
|
John S. Lessons learned from kidney dysfunction : Preventing organ failure. Med Klin Intensivmed Notfmed 2020; 115:21-27. [PMID: 32052096 DOI: 10.1007/s00063-020-00659-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/14/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) is a common and severe complication in patients in the intensive care unit with a significant impact on patient's mortality and morbidity. Therefore renal protective therapy is very important in these severely ill patients. AIM Several renal protective strategies have been postulated during recent decades, which came from pathophysiologic concepts and have been contradicted or changed during the last few years. So lessons had to be learned in AKI, leading to new, in many cases completely reversed preventive and therapeutic concepts which may also be important for protection in other organs. RECENT FINDINGS Most important for renal protection is the early identification of patients at risk for AKI or with acute kidney damage before renal function further deteriorates. A stage-based management of AKI comprises more general measures like discontinuation of the nephrotoxic agent but most importantly early hemodynamic stabilization. Recent research has contradicted that AKI is renal ischemia caused by vasoconstriction with consecutive tubular necrosis. In septic AKI, renal blood flow is even increased. Intrarenal vasodilation together with microcirculatory changes and redistribution of blood flow lead to a drop in glomerular filtration by functional changes. Accordingly it had to be learned that not vasodilators but vasoconstrictors are beneficial in AKI. A mean arterial blood pressure target of >65 mm Hg is often recommended but exact targets are not known, and patients with pre-existing hypertension even need higher perfusion pressure. Also the concept that fluid therapy is always beneficial for the kidney in shock states had to be revised. A volume restrictive therapy with only balanced crystalloids is also becoming more important in AKI. Still no specific pharmacological therapy for renal protection is available. Inflammation and mitochondrial dysfunction appear to play a significant role in AKI. Anti-inflammatory strategies are under investigation and may become more important for AKI prevention and therapy in the future. (This article is freely available.).
Collapse
Affiliation(s)
- Stefan John
- Medical Intensive Care, Medical Department 8, Klinikum Nuernberg-Sued, Paracelsus Medical University and University of Erlangen-Nuernberg, Breslauerstr. 201, 90471, Nuernberg, Germany.
| |
Collapse
|
131
|
Wen X, Li S, Frank A, Chen X, Emlet D, Hukriede NA, Kellum JA. Time-dependent effects of histone deacetylase inhibition in sepsis-associated acute kidney injury. Intensive Care Med Exp 2020; 8:9. [PMID: 32034542 PMCID: PMC7007462 DOI: 10.1186/s40635-020-0297-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/29/2020] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Sepsis, a dysregulated host response to infection with results in organ dysfunction, has been a major challenge to the development of effective therapeutics. Sepsis-associated acute kidney injury (S-AKI) results in a 3-5-fold increase in the risk of hospital mortality compared to sepsis alone. The development of therapies to reverse S-AKI could therefore significantly affect sepsis outcomes. However, the translation of therapies from preclinical studies into humans requires model systems that recapitulate clinical scenarios and the development of renal fibrosis indicative of the transition from acute to chronic kidney disease. RESULTS Here we characterized a murine model of S-AKI induced by abdominal sepsis developing into a chronic phenotype. We applied a small molecule histone deacetylase-8 inhibitor, UPHD186, and found that early treatment, beginning at 48 h post-sepsis, worsened renal outcome accompanied by decreasing mononuclear cell infiltration in the kidney, skewing cells into a pro-inflammatory phenotype, and increased pro-fibrotic gene expression, while delayed treatment, beginning at 96 h post-sepsis, after the acute inflammation in the kidney had subsided, resulted in improved survival and kidney histology presumably through promoting proliferation and inhibiting fibrosis. CONCLUSIONS These findings not only present a clinically relevant S-AKI model, but also introduce a timing dimension into S-AKI therapeutic interventions that delayed treatment with UPHD186 may enhance renal histologic repair. Our results provide novel insights into successful repair of kidney injury and sepsis therapy.
Collapse
Affiliation(s)
- Xiaoyan Wen
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3347 Forbes Ave, Suite 220, Pittsburgh, PA, 15213, USA
| | - Shengnan Li
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3347 Forbes Ave, Suite 220, Pittsburgh, PA, 15213, USA
| | - Alicia Frank
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3347 Forbes Ave, Suite 220, Pittsburgh, PA, 15213, USA
| | - Xiukai Chen
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3347 Forbes Ave, Suite 220, Pittsburgh, PA, 15213, USA
| | - David Emlet
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3347 Forbes Ave, Suite 220, Pittsburgh, PA, 15213, USA
| | - Neil A Hukriede
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3347 Forbes Ave, Suite 220, Pittsburgh, PA, 15213, USA
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3347 Forbes Ave, Suite 220, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
132
|
Abstract
Acute kidney injury (AKI) is defined by a rapid increase in serum creatinine, decrease in urine output, or both. AKI occurs in approximately 10-15% of patients admitted to hospital, while its incidence in intensive care has been reported in more than 50% of patients. Kidney dysfunction or damage can occur over a longer period or follow AKI in a continuum with acute and chronic kidney disease. Biomarkers of kidney injury or stress are new tools for risk assessment and could possibly guide therapy. AKI is not a single disease but rather a loose collection of syndromes as diverse as sepsis, cardiorenal syndrome, and urinary tract obstruction. The approach to a patient with AKI depends on the clinical context and can also vary by resource availability. Although the effectiveness of several widely applied treatments is still controversial, evidence for several interventions, especially when used together, has increased over the past decade.
Collapse
Affiliation(s)
- Claudio Ronco
- Department of Medicine, University of Padova, Padova, Italy; International Renal Research Institute of Vicenza, Vicenza, Italy; Department of Nephrology, San Bortolo Hospital, Vicenza, Italy.
| | - Rinaldo Bellomo
- Critical Care Department, Austin Hospital, Melbourne, VIC, Australia
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
133
|
Lankadeva YR, Okazaki N, Evans RG, Bellomo R, May CN. Renal Medullary Hypoxia: A New Therapeutic Target for Septic Acute Kidney Injury? Semin Nephrol 2019; 39:543-553. [DOI: 10.1016/j.semnephrol.2019.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
134
|
Renal Blood Flow Measurement in Early Clinical Sepsis-Can You Catch a Shadow? Crit Care Med 2019; 46:1028-1030. [PMID: 29762410 DOI: 10.1097/ccm.0000000000003108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
135
|
Hoste EAJ, Kellum JA, Selby NM, Zarbock A, Palevsky PM, Bagshaw SM, Goldstein SL, Cerdá J, Chawla LS. Global epidemiology and outcomes of acute kidney injury. Nat Rev Nephrol 2019; 14:607-625. [PMID: 30135570 DOI: 10.1038/s41581-018-0052-0] [Citation(s) in RCA: 826] [Impact Index Per Article: 137.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute kidney injury (AKI) is a commonly encountered syndrome associated with various aetiologies and pathophysiological processes leading to decreased kidney function. In addition to retention of waste products, impaired electrolyte homeostasis and altered drug concentrations, AKI induces a generalized inflammatory response that affects distant organs. Full recovery of kidney function is uncommon, which leaves these patients at risk of long-term morbidity and death. Estimates of AKI prevalence range from <1% to 66%. These variations can be explained by not only population differences but also inconsistent use of standardized AKI classification criteria. The aetiology and incidence of AKI also differ between high-income and low-to-middle-income countries. High-income countries show a lower incidence of AKI than do low-to-middle-income countries, where contaminated water and endemic diseases such as malaria contribute to a high burden of AKI. Outcomes of AKI are similar to or more severe than those of patients in high-income countries. In all resource settings, suboptimal early recognition and care of patients with AKI impede their recovery and lead to high mortality, which highlights unmet needs for improved detection and diagnosis of AKI and for efforts to improve care for these patients.
Collapse
Affiliation(s)
- Eric A J Hoste
- Intensive Care Unit, Ghent University Hospital, Ghent University, Ghent, Belgium.
| | - John A Kellum
- Center for Critical Care Nephrology, Pittsburgh, PA, USA
| | - Nicholas M Selby
- Centre for Kidney Research and Innovation, Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Royal Derby Hospital Campus, Nottingham, UK
| | - Alexander Zarbock
- University of Münster, Department of Anesthesiology, Intensive Care and Pain Medicine, Münster, Germany
| | - Paul M Palevsky
- VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sean M Bagshaw
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jorge Cerdá
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY, USA
| | | |
Collapse
|
136
|
Pena-Hernandez C, Nugent K. One approach to circulation and blood flow in the critical care unit. World J Crit Care Med 2019; 8:36-48. [PMID: 31667132 PMCID: PMC6817932 DOI: 10.5492/wjccm.v8.i4.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/24/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
Evaluating and managing circulatory failure is one of the most challenging tasks for medical practitioners involved in critical care medicine. Understanding the applicability of some of the basic but, at the same time, complex physiological processes occurring during a state of illness is sometimes neglected and/or presented to the practitioners as point-of-care protocols to follow. Furthermore, managing hemodynamic shock has shown us that the human body is designed to fight to sustain life and that the compensatory mechanisms within organ systems are extraordinary. In this review article, we have created a minimalistic guide to the clinical information relevant when assessing critically ill patients with failing circulation. Measures such as organ blood flow, circulating volume, and hemodynamic biomarkers of shock are described. In addition, we will describe historical scientific events that led to some of our current medical practices and its validation for clinical decision making, and we present clinical advice for patient care and medical training.
Collapse
Affiliation(s)
- Camilo Pena-Hernandez
- Department of Internal Medicine, Division of Nephrology and Hypertension, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States
| | - Kenneth Nugent
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States
| |
Collapse
|
137
|
Panitchote A, Mehkri O, Hastings A, Hanane T, Demirjian S, Torbic H, Mireles-Cabodevila E, Krishnan S, Duggal A. Clinical predictors of renal non-recovery in acute respiratory distress syndrome. BMC Nephrol 2019; 20:255. [PMID: 31291909 PMCID: PMC6617675 DOI: 10.1186/s12882-019-1439-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/24/2019] [Indexed: 12/26/2022] Open
Abstract
Background Acute kidney injury (AKI) is the most common extra-pulmonary organ failure in acute respiratory distress syndrome (ARDS). Renal recovery after AKI is determined by several factors. The objective of this study was to determine the predictors of renal non-recovery in ARDS patients. Methods A single center retrospective cohort study of patients with AKI after onset of ARDS. Patients with preexisting chronic kidney disease or intensive care unit stay < 24 h were excluded. AKI staging was defined according to the Kidney Disease Improving Global Outcomes (KDIGO) 2012 guidelines. Renal non-recovery was defined as death, dialysis dependence, serum creatinine ≥1.5 times the baseline, or urine output < 0.5 mL/kg/h more than 6 h. Results Of the 244 patients that met study criteria, 60 (24.6%) had stage I AKI, 66 (27%) had stage II AKI, and 118 (48.4%) had stage III AKI. Of those, 148 (60.7%) patients had renal non-recovery. On multivariable analysis, factors associated with renal non-recovery were a higher stage of AKI (odds ratio [OR] stage II 5.71, 95% confidence interval [CI] 2.17–14.98; OR stage III 45.85, 95% CI 16.27–129.2), delay in the onset of AKI (OR 1.12, 95% CI 1.03–1.21), history of malignancy (OR 4.02, 95% CI 1.59–10.15), septic shock (OR 3.2, 95% CI 1.52–6.76), and a higher tidal volume on day 1–3 of ARDS (OR 1.41, 95% CI 1.05–1.90). Subgroup analysis of survival at day 28 of ARDS also found that higher severity of AKI (OR stage II 8.17, 95% CI 0.84–79.91; OR stage III 111.67, 95% CI 12.69–982.91), delayed onset of AKI (OR 1.12, 95% CI 1.02–1.23), and active malignancy (OR 6.55, 95% CI 1.34–32.04) were significant predictors of renal non-recovery. Conclusions A higher stage of AKI, delayed onset of AKI, a history of malignancy, septic shock, and a higher tidal volume on day 1–3 of ARDS predicted renal non-recovery in ARDS patients. Among survivors, a higher stage of AKI, delayed onset of AKI, and a history of malignancy were associated with renal non-recovery. Electronic supplementary material The online version of this article (10.1186/s12882-019-1439-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anupol Panitchote
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA.,Division of Critical Care Medicine, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Omar Mehkri
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrei Hastings
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tarik Hanane
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sevag Demirjian
- Department of Nephrology, Cleveland Clinic, Cleveland, OH, USA
| | - Heather Torbic
- Department of Pharmacology, Cleveland Clinic, Cleveland, OH, USA
| | | | - Sudhir Krishnan
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Abhijit Duggal
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
138
|
Guzzi LM, Bergler T, Binnall B, Engelman DT, Forni L, Germain MJ, Gluck E, Göcze I, Joannidis M, Koyner JL, Reddy VS, Rimmelé T, Ronco C, Textoris J, Zarbock A, Kellum JA. Clinical use of [TIMP-2]•[IGFBP7] biomarker testing to assess risk of acute kidney injury in critical care: guidance from an expert panel. Crit Care 2019; 23:225. [PMID: 31221200 PMCID: PMC6585126 DOI: 10.1186/s13054-019-2504-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/04/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The first FDA-approved test to assess risk for acute kidney injury (AKI), [TIMP-2]•[IGFBP7], is clinically available in many parts of the world, including the USA and Europe. We sought to understand how the test is currently being used clinically. METHODS We invited a group of experts knowledgeable on the utility of this test for kidney injury to a panel discussion regarding the appropriate use of the test. Specifically, we wanted to identify which patients would be appropriate for testing, how the results are interpreted, and what actions would be taken based on the results of the test. We used a modified Delphi method to prioritize specific populations for testing and actions based on biomarker test results. No attempt was made to evaluate the evidence in support of various actions however. RESULTS Our results indicate that clinical experts have developed similar practice patterns for use of the [TIMP-2]•[IGFBP7] test in Europe and North America. Patients undergoing major surgery (both cardiac and non-cardiac), those who were hemodynamically unstable, or those with sepsis appear to be priority patient populations for testing kidney stress. It was agreed that, in patients who tested positive, management of potentially nephrotoxic drugs and fluids would be a priority. Patients who tested negative may be candidates for "fast-track" protocols. CONCLUSION In the experience of our expert panel, biomarker testing has been a priority after major surgery, hemodynamic instability, or sepsis. Our panel members reported that a positive test prompts management of nephrotoxic drugs as well as fluids, while patients with negative results are considered to be excellent candidates for "fast-track" protocols.
Collapse
Affiliation(s)
- Louis M. Guzzi
- Florida Hospital, 601 E. Rollins Street, Orlando, FL 32803 USA
| | - Tobias Bergler
- University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Brian Binnall
- Baystate Medical Center, 759 Chestnut Street, Springfield, MA 01107 USA
| | | | - Lui Forni
- The Royal Surrey County Hospital NHS Foundation Trust, Egerton Rd, Guildford, Surrey GU2 7XX UK
- University of Surrey, 388 Stag Hill, Guildford, Surrey GU2 7XH UK
| | | | - Eric Gluck
- Swedish Covenant Hospital, 5145 N California Ave, Chicago, IL 60625 USA
| | - Ivan Göcze
- University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Jay L. Koyner
- Section of Nephrology, Department of Medicine, University of Chicago, 5841 South Maryland Ave, Suite S-507, MC5100, Chicago, IL 60637 USA
| | - V. Seenu Reddy
- Tristar Centennial Medical Center, 2400 Patterson St #307, Nashville, TN 37203 USA
| | - Thomas Rimmelé
- Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d’Arsonval, 69003 Lyon, France
| | - Claudio Ronco
- Department of Nephrology University of Padua, Padua Italy; San Bortolo Hospital, Vicenza, Italy; International Renal Research Institute Vicenza, Vicenza, Italy
| | - Julien Textoris
- Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d’Arsonval, 69003 Lyon, France
- bioMérieux, 5 Place d’Arsonval, 69003 Lyon, France
| | - Alexander Zarbock
- University Hospital Münster, Albert-Schweitzer Campus 1, Building A1, 48149 Münster, Germany
| | - John A. Kellum
- The Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, 3347 Forbes Avenue, Suite 220, Pittsburgh, PA 15213 USA
- Critical Care Medicine, Clinical & Translational Science, and Bioengineering, Center for Critical Care Nephrology, 3347 Forbes Avenue, Suite 220, Pittsburgh, PA 15213 USA
| |
Collapse
|
139
|
|
140
|
Peerapornratana S, Manrique-Caballero CL, Gómez H, Kellum JA. Acute kidney injury from sepsis: current concepts, epidemiology, pathophysiology, prevention and treatment. Kidney Int 2019; 96:1083-1099. [PMID: 31443997 DOI: 10.1016/j.kint.2019.05.026] [Citation(s) in RCA: 893] [Impact Index Per Article: 148.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/19/2019] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a frequent complication of the critically ill patient and is associated with unacceptable morbidity and mortality. Prevention of S-AKI is difficult because by the time patients seek medical attention, most have already developed acute kidney injury. Thus, early recognition is crucial to provide supportive treatment and limit further insults. Current diagnostic criteria for acute kidney injury has limited early detection; however, novel biomarkers of kidney stress and damage have been recently validated for risk prediction and early diagnosis of acute kidney injury in the setting of sepsis. Recent evidence shows that microvascular dysfunction, inflammation, and metabolic reprogramming are 3 fundamental mechanisms that may play a role in the development of S-AKI. However, more mechanistic studies are needed to better understand the convoluted pathophysiology of S-AKI and to translate these findings into potential treatment strategies and add to the promising pharmacologic approaches being developed and tested in clinical trials.
Collapse
Affiliation(s)
- Sadudee Peerapornratana
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; The Clinical Research, Investigation and Systems Modeling of Acute Illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Excellence Center for Critical Care Nephrology, Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Laboratory Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Carlos L Manrique-Caballero
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; The Clinical Research, Investigation and Systems Modeling of Acute Illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hernando Gómez
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; The Clinical Research, Investigation and Systems Modeling of Acute Illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; The Clinical Research, Investigation and Systems Modeling of Acute Illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
141
|
Miller L, Singbartl K, Chroneos ZC, Ruiz-Velasco V, Lang CH, Bonavia A. Resistin directly inhibits bacterial killing in neutrophils. Intensive Care Med Exp 2019; 7:30. [PMID: 31147868 PMCID: PMC6542889 DOI: 10.1186/s40635-019-0257-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022] Open
Abstract
Background Sepsis-induced immunosuppression is a key factor contributing to the morbidity and mortality of critically ill patients, and polymorphonuclear neutrophil dysfunction is believed to be a hallmark of this immunosuppression. Circulating myeloid cells produce the cytokine resistin (RETN), which has been associated with poor outcomes in sepsis/septic shock and can directly inhibit neutrophil function. We previously demonstrated that resistin caused a dose-dependent impairment in neutrophil migration, reactive oxygen species production, and bacterial clearance in neutrophil cell lines. However, the relative antimicrobial responses of other innate immune cells to Gram-positive and Gram-negative infections in the presence of elevated levels of resistin have not been evaluated. We hypothesized that resistin directly contributes to sepsis-induced immunosuppression by selectively targeting the neutrophil component of the innate cellular immune system. Thus, the goal of the present study was to compare the effect of resistin on bacterial killing using monocultures or co-cultures of monocyte and neutrophil cell lines, as well as to extend our findings to primary immune cells. Results Our results indicate that human resistin impairs the ability of neutrophils to kill the Gram-negative bacterium Pseudomonas aeruginosa and the Gram-positive bacterium Staphylococcus aureus. In contrast, with the exception of macrophages incubated with P. aeruginosa, resistin did not affect the ability of macrophages or monocytes to kill either Gram-positive or Gram-negative organisms. Furthermore, co-incubation of neutrophils with increasing proportions of monocytes did not enhance bacterial killing. Resistin blocked bactericidal activity through partial reduction of F-actin polymerization and suppression of the oxidative burst in neutrophils. Conclusions Our studies indicate that resistin selectively impairs neutrophil bacterial killing. These findings further support the notion that resistin can mimic cell type-dependent immunosuppressive effects. This is consistent with its putative role in the pathogenesis of bacterial sepsis.
Collapse
Affiliation(s)
- Lauren Miller
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, 500 University Dr, Mail Code H-187, Hershey, PA, 17033, USA
| | - Kai Singbartl
- Department of Critical Care Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Zissis C Chroneos
- Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Microbiology and Immunology, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, 500 University Dr, Mail Code H-187, Hershey, PA, 17033, USA
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Anthony Bonavia
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, 500 University Dr, Mail Code H-187, Hershey, PA, 17033, USA.
| |
Collapse
|
142
|
Fiorentino M, Tohme FA, Murugan R, Kellum JA. Plasma Biomarkers in Predicting Renal Recovery from Acute Kidney Injury in Critically Ill Patients. Blood Purif 2019; 48:253-261. [PMID: 31079110 DOI: 10.1159/000500423] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/13/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Numerous studies have suggested a possible role for acute kidney injury (AKI) biomarkers in predicting renal recovery both before and after renal replacement therapy (RRT). However, definitions for recovery and whether to include patients dying but free of RRT may influence results. OBJECTIVES To validate plasma neutrophil gelatinase-associated lipocalin (pNGAL) as a useful biomarker for predicting or improving the ability of clinical predictors alone to predict recovery following AKI, including in our model plasma B-type natriuretic peptide (pBNP) to account for cardiovascular events. METHODS We analyzed 69 patients enrolled in the Acute Renal Failure Trial Network study. pNGAL and pBNP were measured on days 2, 7, and 14. We analyzed their predictive ability for subsequent recovery, defined as alive and independent from dialysis in 60 days. In sensitivity analyses, we explored changes in results with alternative definitions of recovery. RESULTS Twenty-nine patients (42%) recovered from AKI. Neither pNGAL nor pBNP, alone or in combination, was accurate predictors of renal recovery-the best area under the receiver-operating characteristics curve (AUC) was for pNGAL using the largest relative change (AUC 0.59, 95% CI 0.45-0.74). The best clinical model achieved superior performance to biomarkers (AUC 0.69, 95% CI 0.56-0.81). The AUC was greatest (0.75, 95% CI 0.60-0.91) when pNGAL + pBNP on day 14 were added to the clinical model but this increase did not achieve statistical significance. However, integrated discrimination improvement analysis showed that the addition of pNGAL and pBNP on day 14 to the clinical model significantly improved the prediction of renal recovery (p = 0.008). CONCLUSIONS pNGAL and pBNP can improve the accuracy of clinical parameters in predicting AKI recovery and a full model using biomarkers together with age achieved adequate discrimination.
Collapse
Affiliation(s)
- Marco Fiorentino
- The Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari, Bari, Italy
| | - Fadi A Tohme
- The Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Raghavan Murugan
- The Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The CRISMA Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John A Kellum
- The Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA, .,The CRISMA Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,
| |
Collapse
|
143
|
Kellum JA, Wen X, de Caestecker MP, Hukriede NA. Sepsis-Associated Acute Kidney Injury: A Problem Deserving of New Solutions. Nephron Clin Pract 2019; 143:174-178. [PMID: 31018211 DOI: 10.1159/000500167] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/04/2019] [Indexed: 12/13/2022] Open
Abstract
Sepsis-associated acute kidney injury (S-AKI) significantly worsens patient prognosis, and recent evidence suggests that the injury process begins early and may be sustained by therapies used to treat the sepsis (e.g., fluids resuscitation, antibiotics). While efforts to develop less-injurious treatments are making progress, some degree of secondary injury is to be expected. So too is the inevitable nature of organ injury, which is often present at the time the patient seeks medical attention. We recently found that most patients presenting with septic shock and developing AKI had evidence of kidney damage at the time of, or within 24 h of their admission. In such patients, prevention is not a viable option, as injury has already occurred by the time of presentation. Since S-AKI patients are at increased risk of developing chronic kidney disease, a fundamental target for interventions in S-AKI is to prevent fibrosis (maladaptive repair) while stimulating regeneration (proliferation of viable epithelial cells). Using a pathway-agnostic, proliferation-based phenotypic assay, we discovered phenylthiobutanoic acid, a small molecule histone deacetylase inhibitor, that enhances renal recovery and reduces fibrosis in both zebrafish and mouse models of AKI.
Collapse
Affiliation(s)
- John A Kellum
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA, .,Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,
| | - Xiaoyan Wen
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark P de Caestecker
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Neil A Hukriede
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
144
|
Affiliation(s)
- Ryan M Brown
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Matthew W Semler
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
145
|
Lung-Kidney Cross-Talk. CRITICAL CARE NEPHROLOGY 2019. [PMCID: PMC7969729 DOI: 10.1016/b978-0-323-44942-7.00121-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Awareness of the multifaceted lung and kidney interactions in the critically ill has increased considerably. Cardiogenic and noncardiogenic pulmonary edema represent two entities of pulmonary edema and differ significantly in terms of alveolar fluid clearance. Acute lung injury describes the breakdown of normal lung architecture with development of a high-permeability noncardiogenic pulmonary edema resulting from an inflammation/oxidant-mediated injury to the alveolar-capillary barrier and downregulation of the epithelial active ion transport system. Acute kidney injury is the most common organ dysfunction in patients with acute respiratory distress syndrome. It may develop as a result of blood gas disturbances that compromise renal blood flow and renal compensatory mechanisms; pulmonary hypertension, which may aggravate renal impairment by causing renal congestion and tissue edema; and mechanical ventilation–induced alterations, including systemic release of mediators, all which promote end-organ cell injury. Acute kidney injury, on the other hand, may initiate and perpetuate lung injury resulting from fluid overload and the systemic release of mediators that promote increased pulmonary vascular permeability, lung inflammation, and apoptosis, and breakdown of the transepithelial electrolyte and water transport, ultimately leading to respiratory failure. It is hoped that an in-depth understanding of the factors influencing lung-kidney interactions will encourage physicians to explore and develop new strategies for the benefit of the patient. This chapter will:Review the pathophysiology of acute lung injury. Summarize the emerging understanding of lung-kidney cross-talk in the critically ill patient. Identify the mechanisms by which acute kidney injury may potentiate acute lung injury.
Collapse
|
146
|
|
147
|
Effects of Fluid Bolus Therapy on Renal Perfusion, Oxygenation, and Function in Early Experimental Septic Kidney Injury. Crit Care Med 2019; 47:e36-e43. [DOI: 10.1097/ccm.0000000000003507] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
148
|
Truche AS, Ragey SP, Souweine B, Bailly S, Zafrani L, Bouadma L, Clec'h C, Garrouste-Orgeas M, Lacave G, Schwebel C, Guebre-Egziabher F, Adrie C, Dumenil AS, Zaoui P, Argaud L, Jamali S, Goldran Toledano D, Marcotte G, Timsit JF, Darmon M. ICU survival and need of renal replacement therapy with respect to AKI duration in critically ill patients. Ann Intensive Care 2018; 8:127. [PMID: 30560526 PMCID: PMC6297118 DOI: 10.1186/s13613-018-0467-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Transient and persistent acute kidney injury (AKI) could share similar physiopathological mechanisms. The objective of our study was to assess prognostic impact of AKI duration on ICU mortality. DESIGN Retrospective analysis of a prospective database via cause-specific model, with 28-day ICU mortality as primary end point, considering discharge alive as a competing event and taking into account time-dependent nature of renal recovery. Renal recovery was defined as a decrease of at least one KDIGO class compared to the previous day. SETTING 23 French ICUs. PATIENTS Patients of a French multicentric observational cohort were included if they suffered from AKI at ICU admission between 1996 and 2015. INTERVENTION None. RESULTS A total of 5242 patients were included. Initial severity according to KDIGO creatinine definition was AKI stage 1 for 2458 patients (46.89%), AKI stage 2 for 1181 (22.53%) and AKI stage 3 for 1603 (30.58%). Crude 28-day ICU mortality according to AKI severity was 22.74% (n = 559), 27.69% (n = 327) and 26.26% (n = 421), respectively. Renal recovery was experienced by 3085 patients (58.85%), and its rate was significantly different between AKI severity stages (P < 0.01). Twenty-eight-day ICU mortality was independently lower in patients experiencing renal recovery [CSHR 0.54 (95% CI 0.46-0.63), P < 0.01]. Lastly, RRT requirement was strongly associated with persistent AKI whichever threshold was chosen between day 2 and 7 to delineate transient from persistent AKI. CONCLUSIONS Short-term renal recovery, according to several definitions, was independently associated with higher mortality and RRT requirement. Moreover, distinction between transient and persistent AKI is consequently a clinically relevant surrogate outcome variable for diagnostic testing in critically ill patients.
Collapse
Affiliation(s)
- A S Truche
- UMR 1137 - IAME Team 5 - DeSCID : Decision SCiences in Infectious Diseases, Control and Care, Inserm/Paris Diderot University, Sorbonne Paris Cité, Paris, France
- Medical Intensive Care Unit, Grenoble University Hospital, Grenoble 1 University, U823, La Tronche, France
- Nephrology Dialysis Renal Transplantation, Grenoble University Hospital, La Tronche, France
| | - S Perinel Ragey
- Medical Intensive Care Unit, Croix Rousse Hospital, Lyon University Hospital, Lyon, France
| | - B Souweine
- Medical Intensive Care Unit, Gabriel Montpied University Hospital, Clermont-Ferrand, France
| | - S Bailly
- UMR 1137 - IAME Team 5 - DeSCID : Decision SCiences in Infectious Diseases, Control and Care, Inserm/Paris Diderot University, Sorbonne Paris Cité, Paris, France
- Medical Intensive Care Unit, Grenoble University Hospital, Grenoble 1 University, U823, La Tronche, France
| | - L Zafrani
- Medical Intensive Care Unit, AP-HP, Saint Louis Hospital, Paris, France
- Medicine University, Paris 7 University, Paris, France
| | - L Bouadma
- Medical and Infectious Diseases Intensive Care Unit, AP-HP, Bichat Hospital, Paris Diderot University, 75018, Paris, France
| | - C Clec'h
- Intensive Care Unit, AP-HP, Avicenne Hospital, Paris, France
- Medicine University, Paris 13 University, Bobigny, France
| | - M Garrouste-Orgeas
- Intensive Care Unit, Saint Joseph Hospital Network, Paris, France
- Medicine University, Paris Descartes University, Sorbonne Cite, Paris, France
| | - G Lacave
- Medical Intensive Care Unit, André Mignot Hospital, Versailles, France
| | - C Schwebel
- Medical Intensive Care Unit, Grenoble University Hospital, Grenoble 1 University, U823, La Tronche, France
| | - F Guebre-Egziabher
- Nephrology Dialysis Renal Transplantation, Grenoble University Hospital, La Tronche, France
| | - C Adrie
- Physiology Department, Cochin University Hospital, Assistance Publique, Hôpitaux de Paris (AP-HP), Paris Descartes University des, Sorbonne Cite, Paris, France
| | - A S Dumenil
- Medical-Surgical Intensive Care Unit, AP-HP, Antoine Béclère University Hospital, Clamart, France
| | - Ph Zaoui
- Nephrology Dialysis Renal Transplantation, Grenoble University Hospital, La Tronche, France
| | - L Argaud
- Medical Intensive Care Unit, Edouard Herriot University Hospital, Lyon, France
| | - S Jamali
- Critical Care Medicine Unit, Dourdan Hospital, Dourdan, France
| | | | - G Marcotte
- Surgical ICU, Edouard Herriot University Hospital, Lyon, France
| | - J F Timsit
- UMR 1137 - IAME Team 5 - DeSCID : Decision SCiences in Infectious Diseases, Control and Care, Inserm/Paris Diderot University, Sorbonne Paris Cité, Paris, France
- Medical and Infectious Diseases Intensive Care Unit, AP-HP, Bichat Hospital, Paris Diderot University, 75018, Paris, France
| | - M Darmon
- Medical Intensive Care Unit, AP-HP, Saint Louis Hospital, Paris, France.
- Medicine University, Paris 7 University, Paris, France.
- ECSTRA Team, Biostatistics and Clinical Epidemiology, UMR 1153 (Center of Epidemiology and Biostatistics Sorbonne Paris Cité, CRESS), INSERM, Paris, France.
| |
Collapse
|
149
|
Affiliation(s)
- Matthieu Legrand
- AP-HP, GH Saint Louis-Lariboisière, Department of Anesthesiology and Critical Care and Burn Unit, Paris, France
- University Paris Diderot, Université Sorbonne Paris Cité, France
- UMR INSERM 942, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- F-CRIN, INICRCT Network, Paris, France
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
150
|
Pickkers P, Mehta RL, Murray PT, Joannidis M, Molitoris BA, Kellum JA, Bachler M, Hoste EAJ, Hoiting O, Krell K, Ostermann M, Rozendaal W, Valkonen M, Brealey D, Beishuizen A, Meziani F, Murugan R, de Geus H, Payen D, van den Berg E, Arend J. Effect of Human Recombinant Alkaline Phosphatase on 7-Day Creatinine Clearance in Patients With Sepsis-Associated Acute Kidney Injury: A Randomized Clinical Trial. JAMA 2018; 320:1998-2009. [PMID: 30357272 PMCID: PMC6248164 DOI: 10.1001/jama.2018.14283] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE Sepsis-associated acute kidney injury (AKI) adversely affects long-term kidney outcomes and survival. Administration of the detoxifying enzyme alkaline phosphatase may improve kidney function and survival. OBJECTIVE To determine the optimal therapeutic dose, effect on kidney function, and adverse effects of a human recombinant alkaline phosphatase in patients who are critically ill with sepsis-associated AKI. DESIGN, SETTING, AND PARTICIPANTS The STOP-AKI trial was an international (53 recruiting sites), randomized, double-blind, placebo-controlled, dose-finding, adaptive phase 2a/2b study in 301 adult patients admitted to the intensive care unit with a diagnosis of sepsis and AKI. Patients were enrolled between December 2014 and May 2017, and follow-up was conducted for 90 days. The final date of follow-up was August 14, 2017. INTERVENTIONS In the intention-to-treat analysis, in part 1 of the trial, patients were randomized to receive recombinant alkaline phosphatase in a dosage of 0.4 mg/kg (n = 31), 0.8 mg/kg (n = 32), or 1.6 mg/kg (n = 29) or placebo (n = 30), once daily for 3 days, to establish the optimal dose. The optimal dose was identified as 1.6 mg/kg based on modeling approaches and adverse events. In part 2, 1.6 mg/kg (n = 82) was compared with placebo (n = 86). MAIN OUTCOMES AND MEASURES The primary end point was the time-corrected area under the curve of the endogenous creatinine clearance for days 1 through 7, divided by 7 to provide a mean daily creatinine clearance (AUC1-7 ECC). Incidence of fatal and nonfatal (serious) adverse events ([S]AEs) was also determined. RESULTS Overall, 301 patients were enrolled (men, 70.7%; median age, 67 years [interquartile range {IQR}, 59-73]). From day 1 to day 7, median ECC increased from 26.0 mL/min (IQR, 8.8 to 59.5) to 65.4 mL/min (IQR, 26.7 to 115.4) in the recombinant alkaline phosphatase 1.6-mg/kg group vs from 35.9 mL/min (IQR, 12.2 to 82.9) to 61.9 mL/min (IQR, 22.7 to 115.2) in the placebo group (absolute difference, 9.5 mL/min [95% CI, -23.9 to 25.5]; P = .47). Fatal adverse events occurred in 26.3% of patients in the 0.4-mg/kg recombinant alkaline phosphatase group; 17.1% in the 0.8-mg/kg group, 17.4% in the 1.6-mg/kg group, and 29.5% in the placebo group. Rates of nonfatal SAEs were 21.0% for the 0.4-mg/kg recombinant alkaline phosphatase group, 14.3% for the 0.8-mg/kg group, 25.7% for the 1.6-mg/kg group, and 20.5% for the placebo group. CONCLUSIONS AND RELEVANCE Among patients who were critically ill with sepsis-associated acute kidney injury, human recombinant alkaline phosphatase compared with placebo did not significantly improve short-term kidney function. Further research is necessary to assess other clinical outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02182440.
Collapse
Affiliation(s)
- Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Ravindra L. Mehta
- Department of Medicine, University of California, San Diego, Medical Center
| | | | - Michael Joannidis
- Department of Internal Medicine, Division of Intensive Care and Emergency Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | - John A. Kellum
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mirjam Bachler
- Department of General and Surgical Critical Care Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Eric A. J. Hoste
- Intensive Care, Ghent University, Ghent, Belgium
- Clinical Research Foundation Flanders, Brussels, Belgium
| | - Oscar Hoiting
- Intensive Care, Canisius Wilhelmina Hospital, Nijmegen, the Netherlands
| | - Kenneth Krell
- Internal Medicine, Eastern Idaho Regional Medical Center, Idaho Falls
| | - Marlies Ostermann
- Guy’s and St Thomas’ Hospital, King’s College London, London, United Kingdom
| | - Wim Rozendaal
- Intensive Care, Jeroen Bosch Hospital, ‘s-Hertogenbosch, the Netherlands
| | - Miia Valkonen
- Division of Anesthesia and Intensive Care Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - David Brealey
- Division of Critical Care, University College London Hospitals National Institute for Health Research Biomedical Research Centre, London, United Kingdom
- Bloomsbury Institute of Intensive Care Medicine, University College Hospital, London, United Kingdom
| | | | - Ferhat Meziani
- Faculté de Médecine, Service de Réanimation, Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Strasbourg, France
| | - Raghavan Murugan
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hilde de Geus
- Department of Intensive Care, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Didier Payen
- Unité Mixte de Recherche INSERM 1160, University Paris 7 Denis Diderot, Paris, France
- Department of Anaesthesiology and Critical Care, Hôpital Lariboisière, Assistance Publique—Hôpitaux de Paris, Paris, France
| | | | - Jacques Arend
- Clinical Department, AM-Pharma BV, Bunnik, the Netherlands
| |
Collapse
|