1
|
Millinger J, Langenskiöld M, Nygren A, Österberg K, Nordanstig J. Renal Artery Blood Flow and Surface Parenchymal Perfusion During Renal Artery Endoshunting in a Porcine Model. EJVES Vasc Forum 2024; 62:104-109. [PMID: 39640086 PMCID: PMC11617771 DOI: 10.1016/j.ejvsvf.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 12/07/2024] Open
Abstract
Objective Ischaemia and reperfusion can result in permanent tissue damage. During complex open abdominal aortic surgery, transient clamping of the renovisceral arteries may be required to successfully complete the vascular repair. Endovascular shunting (endoshunting) presents an alternative technique for managing such temporary renovisceral ischaemia. This study aimed to investigate the performance of endoshunting to the renal circulation in a porcine model. Methods This study of five domestic pigs investigated arterial volume flow rates during endoshunting of a single renal artery and the associated impact on renal perfusion parameters (laser Doppler renal parenchymal perfusion, renal oxygen extraction, and selective urinary output). The study was performed in three steps: baseline registrations (30 minutes), endoshunting (120 minutes), and restoration (60 minutes). The right kidney was used as the experimental side and the left kidney as control. Results The median arterial flow rate in the left control kidney remained constant throughout the experiment. On the right (endoshunted) side, the baseline median arterial flow rate was 267 (range, 160-404) mL/min. Following activation of the endoshunt, the median arterial volume flow dropped by 59%-110 (range, 45-150) mL/min (p = .018). During endoshunting, the median kidney surface perfusion decreased to 42% of the baseline value. On the control side, a rise in the median parenchymal perfusion was observed after endoshunt activation, which was again normalised following restoration of native right renal artery flow. During endoshunting, the median regional urine production was 0.32 (range, 0.12-0.50) mL/hour but resumed after renal artery flow restoration. Conclusion On average, the endoshunted kidneys showed a rapid restoration of blood flow, parenchymal perfusion, and urine production after 120 minutes of endoshunting. This suggests that endoshunting to the kidney using an endoshunt system might be a promising strategy to preserve renal function when temporary interruption of native renal artery blood flow is needed during complex vascular surgical repairs involving the renal arteries.
Collapse
Affiliation(s)
- Johan Millinger
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Vascular Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marcus Langenskiöld
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Vascular Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Nygren
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Anaesthetics and Intensive Care Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Klas Österberg
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Vascular Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joakim Nordanstig
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Vascular Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
2
|
Zhang H, Zhang T, Hou L, Zhao J, Fan Q, Wang L, Lu Z, Dong H, Lei C. Association of intraoperative cerebral and somatic tissue oxygen saturation with postoperative acute kidney injury in adult patients undergoing multiple valve surgery. BMC Anesthesiol 2023; 23:319. [PMID: 37726660 PMCID: PMC10507988 DOI: 10.1186/s12871-023-02279-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND The association between tissue oxygenation with postoperative acute kidney injury (AKI) in adult patients undergoing multiple valve surgery has not been specifically studied. METHODS In this prospective exploratory cohort study, 99 patients were enrolled. The left forehead, the left forearm, the left upper thigh, and the left renal region tissue oxygen saturation using near-infrared spectroscopy were monitored. The association between each threshold and AKI was assessed. The relative and absolute thresholds were < 70%, < 75%, < 80%, < 85%, < 90%, < 95%, and < 100% baseline, and baseline-standard deviation (SD), -1.5 SD, -2 SD, -2.5 SD, and -3 SD. Multivariate logistic regression analysis was adopted to explore the association. RESULTS AKI occurred in 53 (54%) patients. The absolute value-based SrrO2 thresholds associated with AKI were baseline-3 SD (odds ratio [OR], 4.629; 95% confidence interval [CI], 1.238-17.314; P = 0.023) and baseline-2.5 SD (OR, 2.842; 95% CI, 1.025-7.881; P = 0.045) after adjusting for the potential confounders, those are renal region tissue oxygen saturation of 55% and 60%, but not statistically significant after correcting for multiple testing (corrected P = 0.114 and 0.179, respectively). CONCLUSION The SrrO2 desaturation, defined as < baseline - 2.5 SD or < baseline - 3 SD, may be associated with AKI. The thresholds need to be verified in future large-scale studies. TRIAL REGISTRATIONS The study was registered at ClinicalTrials.gov, first trial registration: 26/10/2017, identifier: NCT03323203.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Taoyuan Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Lihong Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Jing Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Qianqian Fan
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Lini Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Zhihong Lu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China
| | - Chong Lei
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an, 710032, China.
| |
Collapse
|
3
|
Wang F, Otsuka T, Adelnia F, Takahashi K, Delgado R, Harkins KD, Zu Z, de Caestecker MP, Harris RC, Gore JC, Takahashi T. Multiparametric magnetic resonance imaging in diagnosis of long-term renal atrophy and fibrosis after ischemia reperfusion induced acute kidney injury in mice. NMR IN BIOMEDICINE 2022; 35:e4786. [PMID: 35704387 PMCID: PMC10805124 DOI: 10.1002/nbm.4786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
Tubular atrophy and fibrosis are pathological changes that determine the prognosis of kidney disease induced by acute kidney injury (AKI). We aimed to evaluate multiple magnetic resonance imaging (MRI) parameters, including pool size ratio (PSR) from quantitative magnetization transfer, relaxation rates, and measures from spin-lock imaging ( R 1 ρ and S ρ ), for assessing the pathological changes associated with AKI-induced kidney disease. Eight-week-old male C57BL/6 J mice first underwent unilateral ischemia reperfusion injury (IRI) induced by reperfusion after 45 min of ischemia. They were imaged using a 7T MRI system 56 days after the injury. Paraffin tissue sections were stained using Masson trichrome and picrosirius red to identify histopathological changes such as tubular atrophy and fibrosis. Histology detected extensive tubular atrophy and moderate fibrosis in the cortex and outer stripe of the outer medulla (CR + OSOM) and more prominent fibrosis in the inner stripe of the outer medulla (ISOM) of IRI kidneys. In the CR + OSOM region, evident decreases in PSR, R 1 , R 2 , R 1 ρ , and S ρ showed in IRI compared with contralateral kidneys, with PSR and S ρ exhibiting the most significant changes. In addition, the exchange parameter S ρ dropped by the largest degree among all the MRI parameters, whileR 2 * increased significantly. In the ISOM of IRI kidneys, PSR increased while S ρ kept decreasing. R 2 , R 1 ρ , andR 2 * all increased due to more severe fibrosis in this region. Among MRI measures, PSR and R 1 ρ showed the highest detectability of renal changes no matter whether tubular atrophy or fibrosis dominated.R 2 * and S ρ could be more specific to a single pathological event than other MRI measures because onlyR 2 * increased and S ρ decreased consistently when either fibrosis or tubular atrophy dominated, and their correlations with fibrosis scores were higher than other MRI measures. Multiparametric MRI may enable a more comprehensive analysis of histopathological changes following AKI.
Collapse
Affiliation(s)
- Feng Wang
- Vanderbilt University Institute of Imaging Science,
Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Radiology and Radiological Sciences,
Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt O’Brien Kidney Research Center,
Vanderbilt University Medical Center, Nashville, TN 37232
| | - Tadashi Otsuka
- Division of Nephrology and Hypertension, Vanderbilt
University Medical Center, Nashville, TN 37232
| | - Fatemeh Adelnia
- Vanderbilt University Institute of Imaging Science,
Vanderbilt University Medical Center, Nashville, TN 37232
| | - Keiko Takahashi
- Division of Nephrology and Hypertension, Vanderbilt
University Medical Center, Nashville, TN 37232
- Vanderbilt O’Brien Kidney Research Center,
Vanderbilt University Medical Center, Nashville, TN 37232
| | - Rachel Delgado
- Division of Nephrology and Hypertension, Vanderbilt
University Medical Center, Nashville, TN 37232
- Vanderbilt O’Brien Kidney Research Center,
Vanderbilt University Medical Center, Nashville, TN 37232
| | - Kevin D. Harkins
- Vanderbilt University Institute of Imaging Science,
Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Radiology and Radiological Sciences,
Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biomedical Engineering, Vanderbilt
University, Nashville, TN 37232
| | - Zhongliang Zu
- Vanderbilt University Institute of Imaging Science,
Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Radiology and Radiological Sciences,
Vanderbilt University Medical Center, Nashville, TN 37232
| | - Mark P. de Caestecker
- Division of Nephrology and Hypertension, Vanderbilt
University Medical Center, Nashville, TN 37232
- Vanderbilt O’Brien Kidney Research Center,
Vanderbilt University Medical Center, Nashville, TN 37232
| | - Raymond C. Harris
- Division of Nephrology and Hypertension, Vanderbilt
University Medical Center, Nashville, TN 37232
- Vanderbilt O’Brien Kidney Research Center,
Vanderbilt University Medical Center, Nashville, TN 37232
| | - John C. Gore
- Vanderbilt University Institute of Imaging Science,
Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Radiology and Radiological Sciences,
Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biomedical Engineering, Vanderbilt
University, Nashville, TN 37232
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt
University Medical Center, Nashville, TN 37232
- Vanderbilt O’Brien Kidney Research Center,
Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
4
|
Urbán D, Cserni T, Boros M, Juhász Á, Érces D, Varga G. Bladder augmentation from an insider's perspective: a review of the literature on microcirculatory studies. Int Urol Nephrol 2021; 53:2221-2230. [PMID: 34435307 DOI: 10.1007/s11255-021-02971-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Augmentation cystoplasty is an exemplary multiorgan intervention in urology which is particularly associated with microvascular damage. Our aim was to review the available intravital imaging techniques and data obtained from clinical and experimental microcirculatory studies involving the most important donor organs applied in bladder augmentation. Although numerous direct or indirect methods are available to assess the condition of microvessels the implementation of microcirculatory diagnostic methods in humans is still challenging and the assessment of organ microcirculation in the operating theatre has limitations. Nevertheless, preclinical studies generally report good internal validity and although prospective human protocols with reduced variability are needed, a possible positive impact of microcirculatory diagnostics on the clinical outcomes of urologic surgery can be anticipated.
Collapse
Affiliation(s)
- Dániel Urbán
- Institute of Surgical Research, University of Szeged, Pulz u. 1., Szeged, 6724, Hungary.,Department of General and Thoracic Surgery, Hetényi Géza County Hospital, Tószegi u. 21., Szolnok, 5000, Hungary
| | - Tamás Cserni
- Institute of Surgical Research, University of Szeged, Pulz u. 1., Szeged, 6724, Hungary.,Department of Paediatric Urology, The Royal Manchester Children's Hospital, Oxford Road, Manchester, M13 9WL, UK
| | - Mihály Boros
- Institute of Surgical Research, University of Szeged, Pulz u. 1., Szeged, 6724, Hungary
| | - Árpád Juhász
- Department of General and Thoracic Surgery, Hetényi Géza County Hospital, Tószegi u. 21., Szolnok, 5000, Hungary
| | - Dániel Érces
- Institute of Surgical Research, University of Szeged, Pulz u. 1., Szeged, 6724, Hungary
| | - Gabriella Varga
- Institute of Surgical Research, University of Szeged, Pulz u. 1., Szeged, 6724, Hungary.
| |
Collapse
|
5
|
Behem CR, Graessler MF, Friedheim T, Kluttig R, Pinnschmidt HO, Duprée A, Debus ES, Reuter DA, Wipper SH, Trepte CJC. The use of pulse pressure variation for predicting impairment of microcirculatory blood flow. Sci Rep 2021; 11:9215. [PMID: 33911116 PMCID: PMC8080713 DOI: 10.1038/s41598-021-88458-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Dynamic parameters of preload have been widely recommended to guide fluid therapy based on the principle of fluid responsiveness and with regard to cardiac output. An equally important aspect is however to also avoid volume-overload. This accounts particularly when capillary leakage is present and volume-overload will promote impairment of microcirculatory blood flow. The aim of this study was to evaluate, whether an impairment of intestinal microcirculation caused by volume-load potentially can be predicted using pulse pressure variation in an experimental model of ischemia/reperfusion injury. The study was designed as a prospective explorative large animal pilot study. The study was performed in 8 anesthetized domestic pigs (German landrace). Ischemia/reperfusion was induced during aortic surgery. 6 h after ischemia/reperfusion-injury measurements were performed during 4 consecutive volume-loading-steps, each consisting of 6 ml kg−1 bodyweight−1. Mean microcirculatory blood flow (mean Flux) of the ileum was measured using direct laser-speckle-contrast-imaging. Receiver operating characteristic analysis was performed to determine the ability of pulse pressure variation to predict a decrease in microcirculation. A reduction of ≥ 10% mean Flux was considered a relevant decrease. After ischemia–reperfusion, volume-loading-steps led to a significant increase of cardiac output as well as mean arterial pressure, while pulse pressure variation and mean Flux were significantly reduced (Pairwise comparison ischemia/reperfusion-injury vs. volume loading step no. 4): cardiac output (l min−1) 1.68 (1.02–2.35) versus 2.84 (2.15–3.53), p = 0.002, mean arterial pressure (mmHg) 29.89 (21.65–38.12) versus 52.34 (43.55–61.14), p < 0.001, pulse pressure variation (%) 24.84 (17.45–32.22) versus 9.59 (1.68–17.49), p = 0.004, mean Flux (p.u.) 414.95 (295.18–534.72) versus 327.21 (206.95–447.48), p = 0.006. Receiver operating characteristic analysis revealed an area under the curve of 0.88 (CI 95% 0.73–1.00; p value < 0.001) for pulse pressure variation for predicting a decrease of microcirculatory blood flow. The results of our study show that pulse pressure variation does have the potential to predict decreases of intestinal microcirculatory blood flow due to volume-load after ischemia/reperfusion-injury. This should encourage further translational research and might help to prevent microcirculatory impairment due to excessive fluid resuscitation and to guide fluid therapy in the future.
Collapse
Affiliation(s)
- Christoph R Behem
- Department of Anesthesiology, Center of Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Michael F Graessler
- Department of Anesthesiology, Center of Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Till Friedheim
- Department of Anesthesiology, Center of Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Rahel Kluttig
- Department of Anesthesiology, Center of Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Hans O Pinnschmidt
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Duprée
- Department of Visceral- and Thoracic Surgery, Center of Operative Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - E Sebastian Debus
- Department of Vascular Medicine, University Heart and Vascular Center Hamburg GmbH (UHZ), Hamburg, Germany
| | - Daniel A Reuter
- Department of Anesthesiology and Intensive Care Medicine, Rostock University Medical Center, Rostock, Germany
| | - Sabine H Wipper
- University Department for Vascular Surgery, Department of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Constantin J C Trepte
- Department of Anesthesiology, Center of Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
6
|
Bienert A, Sobczyński P, Młodawska K, Hartmann-Sobczyńska R, Grześkowiak E, Wiczling P. The influence of cardiac output on propofol and fentanyl pharmacokinetics and pharmacodynamics in patients undergoing abdominal aortic surgery. J Pharmacokinet Pharmacodyn 2020; 47:583-596. [PMID: 32840723 PMCID: PMC7652808 DOI: 10.1007/s10928-020-09712-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/13/2020] [Indexed: 12/27/2022]
Abstract
Cardiac output (CO) is expected to affect elimination and distribution of highly extracted and perfusion rate-limited drugs. This work was undertaken to quantify the effect of CO measured by the pulse pressure method on pharmacokinetics and pharmacodynamics of propofol and fentanyl administrated during total intravenous anesthesia (TIVA). The data were obtained from 22 ASA III patients undergoing abdominal aortic surgery. Propofol was administered via target-controlled infusion system (Diprifusor) and fentanyl was administered at a dose of 2-3 µg/kg each time analgesia appeared to be inadequate. Hemodynamic measurements as well as bispectral index were monitored and recorded throughout the surgery. Data analysis was performed by using a non-linear mixed-effect population modeling (NONMEM 7.4 software). Three compartment models that incorporated blood flows as parameters were used to describe propofol and fentanyl pharmacokinetics. The delay of the anesthetic effect, with respect to plasma concentrations, was described using a biophase (effect) compartment. The bispectral index was linked to the propofol and fentanyl effect site concentrations through a synergistic Emax model. An empirical linear model was used to describe CO changes observed during the surgery. Cardiac output was identified as an important predictor of propofol and fentanyl pharmacokinetics. Consequently, it affected the depth of anesthesia and the recovery time after propofol-fentanyl TIVA infusion cessation. The model predicted (not observed) CO values correlated best with measured responses. Patients' age was identified as a covariate affecting the rate of CO changes during the anesthesia leading to age-related difference in individual patient's responses to both drugs.
Collapse
Affiliation(s)
- Agnieszka Bienert
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Sw. Marii Magdaleny 14 Street, 61-861, Poznan, Poland.
| | - Paweł Sobczyński
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 1/2 Długa Str., 61-848, Poznań, Poland
| | - Katarzyna Młodawska
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Sw. Marii Magdaleny 14 Street, 61-861, Poznan, Poland
| | - Roma Hartmann-Sobczyńska
- Department of Experimental Anaesthesiology, Poznan University of Medical Sciences, Sw. Marii Magdaleny 14 Street, 61-861, Poznan, Poland
| | - Edmund Grześkowiak
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Sw. Marii Magdaleny 14 Street, 61-861, Poznan, Poland
| | - Paweł Wiczling
- Department of Biopharmacy and Pharmacodynamics, Medical University of Gdansk, Hallera 107 Street, 80-416, Gdansk, Poland
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW Currently, the treatment of patients with shock is focused on the clinical symptoms of shock. In the early phase, this is usually limited to heart rate, blood pressure, lactate levels and urine output. However, as the ultimate goal of resuscitation is the improvement in microcirculatory perfusion the question is whether these currently used signs of shock and the improvement in these signs actually correspond to the changes in the microcirculation. RECENT FINDINGS Recent studies have shown that during the development of shock the deterioration in the macrocirculatory parameters are followed by the deterioration of microcirculatory perfusion. However, in many cases the restoration of adequate macrocirculatory parameters is frequently not associated with improvement in microcirculatory perfusion. This relates not only to the cause of shock, where there are some differences between different forms of shock, but also to the type of treatment. SUMMARY The improvement in macrohemodynamics during the resuscitation is not consistently followed by subsequent changes in the microcirculation. This may result in both over-resuscitation and under-resuscitation leading to increased morbidity and mortality. In this article the principles of coherence and the monitoring of the microcirculation are reviewed.
Collapse
|
8
|
Tavy ALM, de Bruin AFJ, Smits AB, Boerma EC, Ince C, Noordzij PG, Boerma D, van Iterson M. Intestinal Mucosal and Serosal Microcirculation at the Planned Anastomosis during Abdominal Surgery. Eur Surg Res 2020; 60:248-256. [PMID: 31935725 DOI: 10.1159/000505325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/10/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Intestinal blood flow is often named as a key factor in the pathophysiology of anastomotic leakage. The distribution between mucosal and serosal microperfusion during surgery remains to be elucidated. OBJECTIVE The aim of this study was to assess if the mucosal microcirculation of the intestine is more vulnerable to a surgical hit than the serosal microcirculation during surgery. METHODS In an observational cohort study (n = 9 patients), the microcirculation of the bowel serosa and mucosa was visualized with incident dark-field imaging during surgery. At the planned anastomosis, the following microcirculatory parameters were determined: microvascular flow index (MFI), percentage of perfused vessels (PPV), perfused vessel density (PVD), and total vessel density (TVD). Data are presented as median (interquartile range [IQR]). RESULTS Perfusion parameters and vessel density were significantly higher for the mucosa than the serosal microcirculation at the planned site for anastomosis or stoma. Mucosal MFI was 3.00 (IQR 3.00-3.00) compared to a serosal MFI of 2.75 (IQR 2.21-2.94), p = 0.03. The PPV was 99% (IQR 98-100) versus 92% (IQR 66-94), p = 0.01. The TVD was 16.77 mm/mm2 (IQR 13.04-18.01) versus 10.42 mm/mm2 (IQR 9.36-11.81), p = 0.01, and the PVD was 15.44 mm/mm2 (IQR 13.04-17.78) versus 9.02 mm/mm2 (IQR 6.43-9.43), p = 0.01. CONCLUSIONS The mucosal microcirculation was preserved, while lower perfusion of the serosa was found at the planned anastomosis or stoma during surgery. Further research is needed to link our observations to the clinically relevant endpoint of anastomotic leakage.
Collapse
Affiliation(s)
- Arthur L M Tavy
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Nieuwegein, The Netherlands,
| | - Anton F J de Bruin
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Anke B Smits
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - E Christiaan Boerma
- Department of Intensive Care, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Can Ince
- Department of Intensive Care Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Peter G Noordzij
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Mat van Iterson
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
9
|
Gaibi T, Ghatak-Roy A. Approach to Acute Kidney Injuries in the Emergency Department. Emerg Med Clin North Am 2019; 37:661-677. [DOI: 10.1016/j.emc.2019.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
10
|
Ferrara G, Kanoore Edul VS, Caminos Eguillor JF, Buscetti MG, Canales HS, Lattanzio B, Gatti L, Ince C, Dubin A. Effects of fluid and norepinephrine resuscitation in a sheep model of endotoxin shock and acute kidney injury. J Appl Physiol (1985) 2019; 127:788-797. [DOI: 10.1152/japplphysiol.00172.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The pathophysiology of renal failure in septic shock is complex. Although microvascular dysfunction has been proposed as a mechanism, there are controversial findings about the characteristics of microvascular redistribution and the effects of resuscitation. Our hypothesis was that the normalization of systemic hemodynamics with fluids and norepinephrine fails to improve acute kidney injury. To test this hypothesis, we assessed systemic and renal hemodynamics and oxygen metabolism in 24 anesthetized and mechanically ventilated sheep. Renal cortical microcirculation was evaluated by SDF-videomicroscopy. Shock ( n = 12) was induced by intravenous administration of endotoxin. After 60 min of shock, 30 mL/kg of saline solution was infused and norepinephrine was titrated to reach a mean blood pressure of 70 mmHg for 2 h. These animals were compared with a sham group ( n = 12). After endotoxin administration, mean blood pressure, cardiac index, and systemic O2 transport and consumption decreased ( P < 0.05 for all). Resuscitation improved these variables. Endotoxin shock also reduced renal blood flow and O2 transport and consumption (205[157–293] vs. 131 [99–185], 28.4[19.0–38.2] vs. 15.8[13.5–23.2], and 5.4[4.0–8.8] vs. 3.7[3.3–4.5] mL·min−1·100 g−1, respectively); cortical perfused capillary density (23.8[23.5–25.9] vs. 17.5[15.1–19.0] mm/mm2); and creatinine clearance (62.4[39.2–99.4] vs. 10.7[4.4–23.5] mL/min). After 2 h of resuscitation, these variables did not improve (174[91–186], 20.5[10.8–22.7], and 3.8[1.9–4.8] mL·min−1·100 g−1, 19.9[18.6–22.1] mm/mm2, and 5.9[1.0–11.9] mL/min). In conclusion, endotoxin shock induced severe renal failure associated with decreased renal flow, O2 transport and consumption, and cortical microcirculation. Normalization of systemic hemodynamics with fluids and norepinephrine failed to improve renal perfusion, oxygenation, and function. NEW & NOTEWORTHY This experimental model of endotoxin shock induced severe renal failure, which was associated with abnormalities in renal regional blood flow, microcirculation, and oxygenation. Derangements included the compromise of peritubular microvascular perfusion. Improvements in systemic hemodynamics through fluids and norepinephrine were unable to correct these abnormalities.
Collapse
Affiliation(s)
- Gonzalo Ferrara
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Cátedra de Farmacología Aplicada, La Plata, Argentina
| | - Vanina Siham Kanoore Edul
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Cátedra de Farmacología Aplicada, La Plata, Argentina
| | | | - María Guillermina Buscetti
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Cátedra de Farmacología Aplicada, La Plata, Argentina
| | - Héctor Saúl Canales
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Cátedra de Farmacología Aplicada, La Plata, Argentina
| | - Bernardo Lattanzio
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Cátedra de Farmacología Aplicada, La Plata, Argentina
| | - Luis Gatti
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Cátedra de Farmacología Aplicada, La Plata, Argentina
| | - Can Ince
- Department of Intensive Care, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Arnaldo Dubin
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Cátedra de Farmacología Aplicada, La Plata, Argentina
| |
Collapse
|
11
|
Tavy ALM, de Bruin AFJ, van der Sloot K, Boerma EC, Ince C, Noordzij PG, Boerma D, van Iterson M. Effects of Thoracic Epidural Anaesthesia on the Serosal Microcirculation of the Human Small Intestine. World J Surg 2019; 42:3911-3917. [PMID: 30097706 DOI: 10.1007/s00268-018-4746-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The effect of thoracic epidural analgesia (TEA) on splanchnic blood flow during abdominal surgery remains unclear. The purpose of this study was to examine whether the hemodynamic effects of TEA resulted in microcirculatory alterations to the intestinal serosa, which was visualized using incident dark-field (IDF) videomicroscopy. METHODS An observational cohort study was performed. In 18 patients, the microcirculation of the intestinal serosa was visualized with IDF. Microcirculatory and hemodynamic measurements were performed prior to (T1) and after administering a bolus of levobupivacaine (T2). If correction of blood pressure was indicated, a third measurement was performed (T3). The following microcirculatory parameters were calculated: microvascular flow index, proportion of perfused vessels, perfused vessel density and total vessel density. Data are presented as median [IQR]. RESULTS Mean arterial pressure decreased from 73 mmHg (68-83) at T1 to 63 mmHg (±11) at T2 (p = 0.001) with a systolic blood pressure of 114 mmHg (98-128) and 87 (81-97), respectively (p = 0.001). The microcirculatory parameters of the bowel serosa, however, were unaltered. In seven patients, blood pressure was corrected to baseline values from a MAP of 56 mmHg (55-57), while microcirculatory parameters remained constant. CONCLUSION We examined the effects of TEA on the intestinal serosal microcirculation during abdominal surgery using IDF imaging for the first time in patients. Regardless of a marked decrease in hemodynamics, microcirculatory parameters of the bowel serosa were not significantly affected. TRIAL REGISTRY NUMBER ClinicalTrials.gov identifier NCT02688946.
Collapse
Affiliation(s)
- A L M Tavy
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands.
| | - A F J de Bruin
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands
| | - K van der Sloot
- Department of Anesthesiology and Pain Medicine, The Hague Medical Center, The Hague, The Netherlands
| | - E C Boerma
- Department of Intensive Care, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - C Ince
- Department of Translational Physiology, Academic Medical Center, Amsterdam, The Netherlands
| | - P G Noordzij
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands
| | - D Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - M van Iterson
- Department of Anesthesiology, Intensive Care and Pain Medicine, St. Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands
| |
Collapse
|
12
|
Loggi S, Mininno N, Damiani E, Marini B, Adrario E, Scorcella C, Domizi R, Carsetti A, Pantanetti S, Pagliariccio G, Carbonari L, Donati A. Changes in the sublingual microcirculation following aortic surgery under balanced or total intravenous anaesthesia: a prospective observational study. BMC Anesthesiol 2019; 19:1. [PMID: 30611197 PMCID: PMC6320625 DOI: 10.1186/s12871-018-0673-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022] Open
Abstract
Background In vascular surgery with aortic cross-clamping, ischemia/reperfusion injury induces systemic haemodynamic and microcirculatory disturbances. Different anaesthetic regimens may have a varying impact on tissue perfusion. The aim of this study was to explore changes in microvascular perfusion in patients undergoing elective open abdominal aortic aneurysm repair under balanced or total intravenous anaesthesia. Methods Prospective observational study. Patients undergoing elective open infrarenal abdominal aortic aneurysm repair received balanced (desflurane + remifentanil, n = 20) or total intravenous anaesthesia (TIVA, propofol + remifentanil using target-controlled infusion, n = 20) according to the clinician’s decision. A goal-directed haemodynamic management was applied in all patients. Measurements were obtained before anaesthesia induction (baseline) and at end-surgery and included haemodynamics, arterial/venous blood gases, sublingual microvascular flow and density (incident dark field illumination imaging), peripheral muscle tissue oxygenation and microcirculatory reactivity (thenar near infrared spectroscopy with a vascular occlusion test). Results The two groups did not differ for baseline characteristics, mean aortic-clamping time and requirement of vasoactive agents during surgery. Changes in mean arterial pressure, systemic vascular resistance index, haemoglobin and blood lactate levels were similar between the two groups, while the cardiac index increased at end-surgery in patients undergoing balanced anaesthesia. The sublingual microcirculation was globally unaltered in the TIVA group at end-surgery, while patients undergoing balanced anaesthesia showed an increase in the total and perfused small vessel densities (from 16.6 ± 4.2 to 19.1 ± 5.4 mm/mm2, p < 0.05). Changes in microvascular density were negatively correlated with changes in the systemic vascular resistance index. The area of reactive hyperaemia during the VOT increased in the balanced anaesthesia group (from 14.8 ± 8.1 to 25.6 ± 14.8%*min, p < 0.05). At end-surgery, the tissue haemoglobin index in the TIVA group was lower than that in the balanced anaesthesia group. Conclusions In patients undergoing elective open abdominal aortic aneurysm repair with a goal-directed hemodynamic management, indices of sublingual or peripheral microvascular perfusion/oxygenation were globally preserved with both balanced anaesthesia and TIVA. Patients undergoing balanced anaesthesia showed microvascular recruitment at end-surgery. Trial registration NCT03510793, https://www.clinicaltrials.gov, date of registration April 27th 2018, retrospectively registered.
Collapse
Affiliation(s)
- Silvia Loggi
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Nicoletta Mininno
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Elisa Damiani
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Benedetto Marini
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Erica Adrario
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Claudia Scorcella
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Roberta Domizi
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Andrea Carsetti
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Simona Pantanetti
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy
| | - Gabriele Pagliariccio
- Unit of Vascular Surgery, Azienda Ospedaliera Universitaria "Ospedali Riuniti Umberto I - Lancisi - Salesi" of Ancona, Ancona, Italy
| | - Luciano Carbonari
- Unit of Vascular Surgery, Azienda Ospedaliera Universitaria "Ospedali Riuniti Umberto I - Lancisi - Salesi" of Ancona, Ancona, Italy
| | - Abele Donati
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, via Tronto 10/a, 60126, Torrette di Ancona, Italy.
| |
Collapse
|
13
|
Evaluation of renal oxygen saturation using photoacoustic imaging for the early prediction of chronic renal function in a model of ischemia-induced acute kidney injury. PLoS One 2018; 13:e0206461. [PMID: 30557371 PMCID: PMC6296502 DOI: 10.1371/journal.pone.0206461] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/13/2018] [Indexed: 11/19/2022] Open
Abstract
PURPOSE To evaluate the utility of photoacoustic imaging in measuring changes in renal oxygen saturation after ischemia-induced acute kidney injury, and to compare these measurements with histological findings and serum levels of kidney function. MATERIAL AND METHODS Acute kidney injury was induced by clamping the left renal pedicle in C57Bl/6 mice, with a 35-min ischemic period used to induce mild renal injury (14 mice) and a 50-min period for severe injury (13 mice). The oxygen saturation was measured before induction, and at 5 time-points over the first 48 h after induction, starting at 4 h after induction. Oxygen saturation, histological score, kidney volume, and the 24 h creatinine clearance rate and serum blood urea nitrogen were also measured on day 28. Between-group differences were evaluated using a Mann-Whitney U-test and Dunn's multiple comparisons. The association between oxygen saturation and measured variables was evaluated using Spearman's correlation. A receiver operator characteristic curve was constructed from oxygen saturation values at 24 h after heminephrectomy to predict chronic renal function. RESULTS The oxygen saturation was higher in the mild than severe renal injury group at 24 h after induction (73.7% and 66.9%, respectively, P<0.05). Between-group comparison on day 28 revealed a higher kidney volume (P = 0.007), lower tubular injury (P<0.001), lower serum level of blood urea nitrogen level (P = 0.016), and lower 24 h creatinine clearance rate (P = 0.042) in the mild compared with the severe injury group. The oxygen saturation at 24 h correlated with the 24 h creatinine clearance rate (P = 0.036) and serum blood urea nitrogen (P<0.001) on day 28, with an area under the receiver operating curve of 0.825. CONCLUSION Oxygen saturation, measured by photoacoustic imaging at 24 h after acute kidney injury can predict the extent of subsequent histological alterations in the kidney early after injury.
Collapse
|
14
|
de Bruin AFJ, Tavy ALM, van der Sloot K, Smits A, Ince C, Boerma EC, Noordzij PG, Boerma D, van Iterson M. Can sidestream dark field (SDF) imaging identify subtle microvascular changes of the bowel during colorectal surgery? Tech Coloproctol 2018; 22:793-800. [PMID: 30413998 DOI: 10.1007/s10151-018-1872-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/20/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recognition of a non-viable bowel during colorectal surgery is a challenging task for surgeons. Identifying the turning point in serosal microcirculatory deterioration leading up to a non-viable bowel is crucial. The aim of the present study was to determine whether sidestream darkfield (SDF) imaging can detect subtle changes in serosal microcirculation of the sigmoid after vascular transection during colorectal surgery. METHODS A prospective observational clinical study was performed at a single medical centre. All eligible participants underwent laparoscopic sigmoid resection and measurements were taken during the extra-abdominal phase. Microcirculation was measured at the transected bowel and 20 cm proximal to this point. Microcirculatory parameters such as Microvascular Flow Index (MFI), proportion of perfused vessels (PPV), perfused vessel density (PVD), total vessel density (TVD) and the Heterogeneity Index were determined. Data are presented as median (interquartile range) or mean ± standard deviation. RESULTS A total of 60 SDF images were acquired for 10 patients. Perfusion parameters and perfused vessel density were significantly lower at the transected bowel compared with the non-transected measurements [MFI 2.29 (1.96-2.63) vs 2.96 (2.73-3.00), p = 0.007; PPV 74% (55-83) vs 94% (86-97), p = 0.007; and PVD 7.61 ± 2.99 mm/mm2 versus 10.67 ± 1.48 mm/mm2, p = 0.009]. Total vessel density was similar between the measurement locations. CONCLUSIONS SDF imaging can identify changes of the bowel serosal microcirculation. Significantly lower serosal microcirculatory parameters of the vascular transected bowel was seen compared with the non-transected bowel. The ability of SDF imaging to detect subtle differences holds promise for future research on microvascular cut-off values leading to a non-viable bowel.
Collapse
Affiliation(s)
- A F J de Bruin
- Department of Anesthesiology, Intensive Care and Pain Medicine, St Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands.
| | - A L M Tavy
- Department of Anesthesiology, Intensive Care and Pain Medicine, St Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands
| | - K van der Sloot
- Department of Anesthesiologie and Pain Medicine, Haaglanden Medisch Centrum, The Hague, The Netherlands
| | - A Smits
- Department of Surgery, St Antonius Hospital, Nieuwegein, The Netherlands
| | - C Ince
- Department of Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| | - E C Boerma
- Department of Intensive Care, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - P G Noordzij
- Department of Anesthesiology, Intensive Care and Pain Medicine, St Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands
| | - D Boerma
- Department of Surgery, St Antonius Hospital, Nieuwegein, The Netherlands
| | - M van Iterson
- Department of Anesthesiology, Intensive Care and Pain Medicine, St Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands
| |
Collapse
|
15
|
Volume Based Resuscitation and Intestinal Microcirculation after Ischaemia/Reperfusion Injury: Results of an Exploratory Aortic Clamping Study in Pigs. Eur J Vasc Endovasc Surg 2018; 57:284-291. [PMID: 30309783 DOI: 10.1016/j.ejvs.2018.08.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 08/27/2018] [Indexed: 11/23/2022]
Abstract
OBJECTIVES In the presence of ischaemia/reperfusion (I/R) induced endothelial injury, volume administration may not correlate with increased microcirculation. The aim of this study was to evaluate intestinal microcirculation after standardised sequential volume loading in an animal model of I/R injury following supracoeliac aortic clamping. METHODS This was a prospective exploratory pilot animal study. Intestinal I/R injury was induced in eight pigs during experimental thoraco-abdominal aortic repair. After 6 h of I/R, microcirculatory blood flow (mFlux, measured in the ileum using direct laser speckle contrast imaging) and macrohaemodynamic parameters (using trans-cardiopulmonary thermodilution) were measured and measurements were repeated after each of four sequential volume loading steps (VLS1 - 4). Each load was administered over 5 min followed by another 5 min for equilibration. RESULTS All animals survived until after VLS4. After 6 h of I/R cardiac output (CO) (p < .001) and mFlux (p < .001) had both decreased. CO increased again after VLS1 (p < .001) and VLS2 (p = .036), whereas mFlux did not change. In contrast, mFlux further decreased after VLS3 (p < .01) and VLS4 (p < .001), whereas CO did not change anymore. Extravascular lung water continued to increase after VLS2 (p = .046) and VLS4 (p = .049). CONCLUSIONS I/R leads to impaired intestinal microcirculation, which was not restored by volume administration in spite of improved CO. In contrast, further volume administration exceeding preload reserves was associated with additional decreases in the intestinal microcirculation. The potentially negative effect of excessive volume resuscitation after I/R injury should encourage further translational research.
Collapse
|
16
|
Ow CPC, Ngo JP, Ullah MM, Barsha G, Meex RC, Watt MJ, Hilliard LM, Koeners MP, Evans RG. Absence of renal hypoxia in the subacute phase of severe renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 2018; 315:F1358-F1369. [PMID: 30110566 PMCID: PMC6293301 DOI: 10.1152/ajprenal.00249.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Tissue hypoxia has been proposed as an important event in renal ischemia-reperfusion injury (IRI), particularly during the period of ischemia and in the immediate hours following reperfusion. However, little is known about renal oxygenation during the subacute phase of IRI. We employed four different methods to assess the temporal and spatial changes in tissue oxygenation during the subacute phase (24 h and 5 days after reperfusion) of a severe form of renal IRI in rats. We hypothesized that the kidney is hypoxic 24 h and 5 days after an hour of bilateral renal ischemia, driven by a disturbed balance between renal oxygen delivery (Do2) and oxygen consumption (V̇o2). Renal Do2 was not significantly reduced in the subacute phase of IRI. In contrast, renal V̇o2 was 55% less 24 h after reperfusion and 49% less 5 days after reperfusion than after sham ischemia. Inner medullary tissue Po2, measured by radiotelemetry, was 25 ± 12% (mean ± SE) greater 24 h after ischemia than after sham ischemia. By 5 days after reperfusion, tissue Po2 was similar to that in rats subjected to sham ischemia. Tissue Po2 measured by Clark electrode was consistently greater 24 h, but not 5 days, after ischemia than after sham ischemia. Cellular hypoxia, assessed by pimonidazole adduct immunohistochemistry, was largely absent at both time points, and tissue levels of hypoxia-inducible factors were downregulated following renal ischemia. Thus, in this model of severe IRI, tissue hypoxia does not appear to be an obligatory event during the subacute phase, likely because of the markedly reduced oxygen consumption.
Collapse
Affiliation(s)
- Connie P C Ow
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Jennifer P Ngo
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Md Mahbub Ullah
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Giannie Barsha
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Ruth C Meex
- Department of Human Biology, NUTRIM School of Nutritional and Translational Research in Metabolism, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Matthew J Watt
- Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Lucinda M Hilliard
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Maarten P Koeners
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol , Bristol , United Kingdom.,Institute of Biomedical and Clinical Science, University of Exeter Medical School , Exeter , United Kingdom
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Physiology, Monash University , Melbourne, Victoria , Australia
| |
Collapse
|
17
|
Ow CPC, Ngo JP, Ullah MM, Hilliard LM, Evans RG. Renal hypoxia in kidney disease: Cause or consequence? Acta Physiol (Oxf) 2018; 222:e12999. [PMID: 29159875 DOI: 10.1111/apha.12999] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/10/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Tissue hypoxia has been proposed as an important factor in the pathophysiology of both chronic kidney disease (CKD) and acute kidney injury (AKI), initiating and propagating a vicious cycle of tubular injury, vascular rarefaction, and fibrosis and thus exacerbation of hypoxia. Here, we critically evaluate this proposition by systematically reviewing the literature relevant to the following six questions: (i) Is kidney disease always associated with tissue hypoxia? (ii) Does tissue hypoxia drive signalling cascades that lead to tissue damage and dysfunction? (iii) Does tissue hypoxia per se lead to kidney disease? (iv) Does tissue hypoxia precede pathology? (v) Does tissue hypoxia colocalize with pathology? (vi) Does prevention of tissue hypoxia prevent kidney disease? We conclude that tissue hypoxia is a common feature of both AKI and CKD. Furthermore, at least under in vitro conditions, renal tissue hypoxia drives signalling cascades that lead to tissue damage and dysfunction. Tissue hypoxia itself can lead to renal pathology, independent of other known risk factors for kidney disease. There is also some evidence that tissue hypoxia precedes renal pathology, at least in some forms of kidney disease. However, we have made relatively little progress in determining the spatial relationships between tissue hypoxia and pathological processes (i.e. colocalization) or whether therapies targeted to reduce tissue hypoxia can prevent or delay the progression of renal disease. Thus, the hypothesis that tissue hypoxia is a "common pathway" to both AKI and CKD still remains to be adequately tested.
Collapse
Affiliation(s)
- C. P. C. Ow
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - J. P. Ngo
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - M. M. Ullah
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - L. M. Hilliard
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - R. G. Evans
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| |
Collapse
|
18
|
|
19
|
Rahmania L, Orbegozo D, Su F, Taccone FS, Vincent JL, De Backer D. Administration of Tetrahydrobiopterin (BH4) Protects the Renal Microcirculation From Ischemia and Reperfusion Injury. Anesth Analg 2017; 125:1253-1260. [PMID: 28632534 DOI: 10.1213/ane.0000000000002131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Abdominal aortic aneurysm surgery with suprarenal cross-clamping is often associated with renal injury. Although the mechanism underlying such injury is unclear, tissue ischemia and reperfusion, which induces endothelial dysfunction and decreases the availability of tetrahydrobiopterin (BH4), may play a role. We evaluated whether BH4 administration prevents renal ischemia/reperfusion injury in an animal model of aortic cross-clamping. METHODS Nineteen anesthetized, mechanically ventilated, and invasively monitored adult sheep were randomized into 3 groups: sham animals (n = 5) that underwent surgical preparation but no aortic clamping; an ischemia/reperfusion group (n = 7), where the aorta was clamped above the renal arteries for 1 hour, and a BH4 group (n = 7), in which animals received 20 mg/kg of BH4 followed by aortic cross-clamp for 1 hour. Animals were followed for a maximum of 6 hours after reperfusion. The renal microcirculation was evaluated at baseline (before clamping), and 1, 4, and 6 hours after reperfusion using side-stream dark field videomicroscopy. The renal lactate-to-pyruvate ratio was evaluated using microdialysis. The primary outcome was the change in proportion of small perfused vessels before and after injury. Secondary outcomes were renal tissue redox state and renal function. RESULTS Ischemia/reperfusion injury was associated with increases in heart rate and mean arterial pressure, which were blunted by BH4 administration. From the first to the sixth hour after reperfusion, the small vessel density (estimated mean difference [EMD], 1.03; 95% confidence interval [CI], 0.41-1.64; P = .003), perfused small vessel density (EMD, 0.84; 95% CI, 0.29-1.39; P = .005), and proportion of perfused small vessels (EMD, 8.60; 95% CI, 0.85-16.30; P = .031) were altered less in the BH4 than in the ischemia/reperfusion group. The renal lactate-to-pyruvate ratios were lower in the cortex in the BH4 than in the ischemia/reperfusion group from the first to the sixth hour after reperfusion (EMD, -19.16; 95% CI, -11.06 to 33.16; P = .002) and in the medulla from the first to the fourth hour (EMD, -26.62; 95% CI, -18.32 to 38.30; P = .020; and EMD, -8.68; 95% CI, -5.96 to 12.65; P = .019). At the sixth hour, serum creatinine was lower in the BH4 than in the ischemia/reperfusion group (EMD, -3.36; 95% CI, -0.29 to 1.39; P = .026). CONCLUSIONS In this sheep model of renal ischemia/reperfusion, BH4 pretreatment reduced renal microvascular injury and improved renal metabolism and function. Further work is needed to clarify the potential role of BH4 in ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Lokmane Rahmania
- From the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
20
|
Mongardon N, Kohlhauer M, Lidouren F, Hauet T, Giraud S, Hutin A, Costes B, Barau C, Bruneval P, Micheau P, Cariou A, Dhonneur G, Berdeaux A, Ghaleh B, Tissier R. A Brief Period of Hypothermia Induced by Total Liquid Ventilation Decreases End-Organ Damage and Multiorgan Failure Induced by Aortic Cross-Clamping. Anesth Analg 2017; 123:659-69. [PMID: 27482772 DOI: 10.1213/ane.0000000000001432] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND In animal models, whole-body cooling reduces end-organ injury after cardiac arrest and other hypoperfusion states. The benefits of cooling in humans, however, are uncertain, possibly because detrimental effects of prolonged cooling may offset any potential benefit. Total liquid ventilation (TLV) provides both ultrafast cooling and rewarming. In previous reports, ultrafast cooling with TLV potently reduced neurological injury after experimental cardiac arrest in animals. We hypothesized that a brief period of rapid cooling and rewarming via TLV could also mitigate multiorgan failure (MOF) after ischemia-reperfusion induced by aortic cross-clamping. METHODS Anesthetized rabbits were submitted to 30 minutes of supraceliac aortic cross-clamping followed by 300 minutes of reperfusion. They were allocated either to a normothermic procedure with conventional ventilation (control group) or to hypothermic TLV (33°C) before, during, and after cross-clamping (pre-clamp, per-clamp, and post-clamp groups, respectively). In all TLV groups, hypothermia was maintained for 75 minutes and switched to a rewarming mode before resumption to conventional mechanical ventilation. End points included cardiovascular, renal, liver, and inflammatory parameters measured 300 minutes after reperfusion. RESULTS In the normothermic (control) group, ischemia-reperfusion injury produced evidence of MOF including severe vasoplegia, low cardiac output, acute kidney injury, and liver failure. In the TLV group, we observed gradual improvements in cardiac output in post-clamp, per-clamp, and pre-clamp groups versus control (53 ± 8, 64 ± 12, and 90 ± 24 vs 36 ± 23 mL/min/kg after 300 minutes of reperfusion, respectively). Liver biomarker levels were also lower in pre-clamp and per-clamp groups versus control. However, acute kidney injury was prevented in pre-clamp, and to a limited extent in per-clamp groups, but not in the post-clamp group. For instance, creatinine clearance was 4.8 ± 3.1 and 0.5 ± 0.6 mL/kg/min at the end of the follow-up in pre-clamp versus control animals (P = .0004). Histological examinations of the heart, kidney, liver, and jejunum in TLV and control groups also demonstrated reduced injury with TLV. CONCLUSIONS A brief period of ultrafast cooling with TLV followed by rapid rewarming attenuated biochemical and histological markers of MOF after aortic cross-clamping. Cardiovascular and liver dysfunctions were limited by a brief period of hypothermic TLV, even when started after reperfusion. Conversely, acute kidney injury was limited only when hypothermia was started before reperfusion. Further work is needed to determine the clinical significance of our results and to identify the optimal duration and timing of TLV-induced hypothermia for end-organ protection in hypoperfusion states.
Collapse
Affiliation(s)
- Nicolas Mongardon
- From the *Inserm, U955, Equipe 3, Créteil, France; †Université Paris Est, UMR_S 955, UPEC, DHU A-TVB, Créteil, France; ‡Université Paris Est, Ecole Nationale Vétérinaire Alfort, Maisons Alfort, France; §Service d' Anesthésie et des Réanimations Chirurgicales, DHU A-TVB, Hôpitaux Universitaires Henri Mondor, Assistance Publique - Hôpitaux de Paris, Créteil, France; ‖Inserm, U1082, Poitiers, France; ¶Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France; #CHU de Poitiers, Service de Biochimie, Poitiers, France; **Inserm, UMR 970, Paris Cardiovascular Research Center, Paris, France; ††Université de Sherbrooke, Sherbrooke, Canada; and ‡‡Service de Réanimation Médicale, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bullen A, Liu ZZ, Hepokoski M, Li Y, Singh P. Renal Oxygenation and Hemodynamics in Kidney Injury. Nephron Clin Pract 2017; 137:260-263. [PMID: 28614837 DOI: 10.1159/000477830] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/26/2022] Open
Abstract
Acute kidney injury (AKI) continues to be a major therapeutic challenge. Despite significant advances made in cellular and molecular pathophysiology of AKI, major gaps in knowledge exist regarding the changes in renal hemodynamics and oxygenation in the early stages and through the continuum of AKI. Particular features of renal hemodynamics and oxygenation increase the susceptibility of the kidney to sustain injury due to oxygen demand-supply mismatch and also play an important role in the recovery and repair from AKI as well as the transition of AKI to chronic kidney disease. However, lack of well-established physiological biomarkers and noninvasive imaging techniques limit our understanding of the interactions between renal macro and microcirculation and tissue oxygenation in AKI. Advances in our ability to assess these parameters in preclinical and clinical AKI will enable the development of targeted therapeutics to improve clinical outcomes.
Collapse
Affiliation(s)
- Alexander Bullen
- Division of Nephrology-Hypertension, University of California, San Diego School of Medicine and VA San Diego Healthcare System, San Diego, CA, USA
| | | | | | | | | |
Collapse
|
22
|
Zammert M, Gelman S. The pathophysiology of aortic cross-clamping. Best Pract Res Clin Anaesthesiol 2016; 30:257-69. [PMID: 27650338 DOI: 10.1016/j.bpa.2016.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022]
Abstract
During open aortic surgery, interrupting the blood flow through the aorta by applying a cross-clamp is often a key step to allow for surgical repair. As a consequence, ischemia is induced in parts of the body distal to the clamp site. This significant alteration in the blood flow is almost always associated with hemodynamic changes. Upon release of the cross-clamp, the blood flow is restored, triggering an ischemia-reperfusion response, leading to many pathophysiological processes such as inflammation, humoral changes, and metabolite circulation that could lead to injury in many organ systems and may significantly influence the postoperative outcome. It is therefore important to understand these processes and how they can be treated in order to allow for safe surgical aortic repairs while ensuring the best possible outcomes.
Collapse
Affiliation(s)
- Martin Zammert
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Simon Gelman
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Layton AT. Recent advances in renal hypoxia: insights from bench experiments and computer simulations. Am J Physiol Renal Physiol 2016; 311:F162-5. [PMID: 27147670 DOI: 10.1152/ajprenal.00228.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 04/26/2016] [Indexed: 11/22/2022] Open
Abstract
The availability of oxygen in renal tissue is determined by the complex interactions among a host of processes, including renal blood flow, glomerular filtration, arterial-to-venous oxygen shunting, medullary architecture, Na(+) transport, and oxygen consumption. When this delicate balance is disrupted, the kidney may become susceptible to hypoxic injury. Indeed, renal hypoxia has been implicated as one of the major causes of acute kidney injury and chronic kidney diseases. This review highlights recent advances in our understanding of renal hypoxia; some of these studies were published in response to a recent Call for Papers of this journal: Renal Hypoxia.
Collapse
Affiliation(s)
- Anita T Layton
- Department of Mathematics, Duke University, North Carolina
| |
Collapse
|
24
|
Prowle JR, Bellomo R. Sepsis-associated acute kidney injury: macrohemodynamic and microhemodynamic alterations in the renal circulation. Semin Nephrol 2015; 35:64-74. [PMID: 25795500 DOI: 10.1016/j.semnephrol.2015.01.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Traditionally, renal ischemia has been regarded as central to the pathogenesis of sepsis-associated acute kidney injury (SA-AKI). Accordingly, hemodynamic management of SA-AKI has emphasized restoration of renal perfusion, whereas, experimentally, ischemia reperfusion models have been emphasized. However, in human beings, SA-AKI usually is accompanied by hyperdynamic circulation. Moreover, clinical and experimental evidence now suggests the importance of inflammatory mechanisms in the development of AKI and microcirculatory dysfunction more than systemic alteration in renal perfusion. In this review, we examine systemic, regional, and microcirculatory hemodynamics in SA-AKI, and attempt to rationalize the hemodynamic management of this condition.
Collapse
Affiliation(s)
- John R Prowle
- Adult Critical Care Unit, Department of Renal Medicine and Transplantation, The Royal London Hospital, Barts Health NHS Trust, London, UK; William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, Australia; Australian and New Zealand Intensive Care Research Centre, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia.
| |
Collapse
|
25
|
Balestra GM, Aalders MCG, Specht PAC, Ince C, Mik EG. Oxygenation measurement by multi-wavelength oxygen-dependent phosphorescence and delayed fluorescence: catchment depth and application in intact heart. JOURNAL OF BIOPHOTONICS 2015; 8:615-628. [PMID: 25250821 DOI: 10.1002/jbio.201400054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/11/2014] [Accepted: 08/18/2014] [Indexed: 06/03/2023]
Abstract
Oxygen delivery and metabolism represent key factors for organ function in health and disease. We describe the optical key characteristics of a technique to comprehensively measure oxygen tension (PO(2)) in myocardium, using oxygen-dependent quenching of phosphorescence and delayed fluorescence of porphyrins, by means of Monte Carlo simulations and ex vivo experiments. Oxyphor G2 (microvascular PO(2)) was excited at 442 nm and 632 nm and protoporphyrin IX (mitochondrial PO(2)) at 510 nm. This resulted in catchment depths of 161 (86) µm, 350 (307) µm and 262 (255) µm respectively, as estimated by Monte Carlo simulations and ex vivo experiments (brackets). The feasibility to detect changes in oxygenation within separate anatomical compartments is demonstrated in rat heart in vivo. Schematic of ex vivo measurements.
Collapse
Affiliation(s)
- Gianmarco M Balestra
- Department of Anesthesiology, Laboratory of Experimental Anesthesiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Medical Intensive Care, University Hospital Basel, Switzerland
- Department of Translational Physiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Maurice C G Aalders
- Department of Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands
| | - Patricia A C Specht
- Department of Anesthesiology, Laboratory of Experimental Anesthesiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Can Ince
- Department of Translational Physiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Egbert G Mik
- Department of Anesthesiology, Laboratory of Experimental Anesthesiology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Layton AT. Recent advances in renal hemodynamics: insights from bench experiments and computer simulations. Am J Physiol Renal Physiol 2015; 308:F951-5. [PMID: 25715984 DOI: 10.1152/ajprenal.00008.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/23/2015] [Indexed: 01/08/2023] Open
Abstract
It has been long known that the kidney plays an essential role in the control of body fluids and blood pressure and that impairment of renal function may lead to the development of diseases such as hypertension (Guyton AC, Coleman TG, Granger Annu Rev Physiol 34: 13-46, 1972). In this review, we highlight recent advances in our understanding of renal hemodynamics, obtained from experimental and theoretical studies. Some of these studies were published in response to a recent Call for Papers of this journal: Renal Hemodynamics: Integrating with the Nephron and Beyond.
Collapse
Affiliation(s)
- Anita T Layton
- Department of Mathematics, Duke University, Durham, North Carolina
| |
Collapse
|
27
|
Abdelkader A, Ho J, Ow CPC, Eppel GA, Rajapakse NW, Schlaich MP, Evans RG. Renal oxygenation in acute renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 2014; 306:F1026-38. [PMID: 24598805 DOI: 10.1152/ajprenal.00281.2013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tissue hypoxia has been demonstrated, in both the renal cortex and medulla, during the acute phase of reperfusion after ischemia induced by occlusion of the aorta upstream from the kidney. However, there are also recent clinical observations indicating relatively well preserved oxygenation in the nonfunctional transplanted kidney. To test whether severe acute kidney injury can occur in the absence of widespread renal tissue hypoxia, we measured cortical and inner medullary tissue Po2 as well as total renal O2 delivery (Do2) and O2 consumption (Vo2) during the first 2 h of reperfusion after 60 min of occlusion of the renal artery in anesthetized rats. To perform this experiment, we used a new method for measuring kidney Do2 and Vo2 that relies on implantation of fluorescence optodes in the femoral artery and renal vein. We were unable to detect reductions in renal cortical or inner medullary tissue Po2 during reperfusion after ischemia localized to the kidney. This is likely explained by the observation that Vo2 (-57%) was reduced by at least as much as Do2 (-45%), due to a large reduction in glomerular filtration (-94%). However, localized tissue hypoxia, as evidence by pimonidazole adduct immunohistochemistry, was detected in kidneys subjected to ischemia and reperfusion, particularly in, but not exclusive to, the outer medulla. Thus, cellular hypoxia, particularly in the outer medulla, may still be present during reperfusion even when reductions in tissue Po2 are not detected in the cortex or inner medulla.
Collapse
Affiliation(s)
- Amany Abdelkader
- Dept. of Physiology, PO Box 13F, Monash Univ., Victoria 3800, Australia.
| | | | | | | | | | | | | |
Collapse
|
28
|
Renal blood flow, fractional excretion of sodium and acute kidney injury: time for a new paradigm? Curr Opin Crit Care 2013; 18:585-92. [PMID: 22954663 DOI: 10.1097/mcc.0b013e328358d480] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Global renal blood flow is considered pivotal to renal function. Decreased global renal blood flow (decreased perfusion) is further considered the major mechanism of reduced glomerular filtration rate responsible for the development of acute kidney injury (AKI) in critically ill patients. Additionally, urinary biochemical tests are widely taught to allow the differential diagnosis of prerenal (functional) AKI and intrinsic [structural AKI (so-called acute tubular necrosis)]. In this review we will examine recent evidence regarding these two key clinical paradigms. RECENT FINDINGS Recent animal experiments and clinical studies in humans using cine-phase contrast magnetic resonance technology are not consistent with the decreased perfusion paradigm. They suggest instead that changes in the intra-renal circulation including modification in efferent arteriolar function and intra-renal shunting are much more likely to be responsible for AKI, especially in sepsis. Similarly, recent human studies indicate the urinary biochemistry has limited diagnostic or prognostic ability and is dissociated form biomarker and microscopic evidence of tubular injury. SUMMARY Intra-renal microcirculatory changes are likely more important than changes in global blood flow in the development of AKI. Urinary biochemistry is not a clinically useful diagnostic or prognostic tool in critically ill patients at risk of or with AKI.
Collapse
|
29
|
Evans RG, Ince C, Joles JA, Smith DW, May CN, O'Connor PM, Gardiner BS. Haemodynamic influences on kidney oxygenation: Clinical implications of integrative physiology. Clin Exp Pharmacol Physiol 2013; 40:106-22. [DOI: 10.1111/1440-1681.12031] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 10/21/2012] [Accepted: 11/15/2012] [Indexed: 01/08/2023]
Affiliation(s)
- Roger G Evans
- Department of Physiology; Monash University; Melbourne; Victoria; Australia
| | - Can Ince
- Department of Translational Physiology; Academic Medical Center; University of Amsterdam; Amsterdam; The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension; University Medical Center; Utrecht; The Netherlands
| | - David W Smith
- School of Computer Science and Software Engineering; The University of Western Australia; Perth; Western Australia; Australia
| | - Clive N May
- Florey Institute of Neuroscience and Mental Health; University of Melbourne; Melbourne; Victoria; Australia
| | - Paul M O'Connor
- Department of Physiology; Georgia Health Sciences University; Augusta; GA; USA
| | - Bruce S Gardiner
- School of Computer Science and Software Engineering; The University of Western Australia; Perth; Western Australia; Australia
| |
Collapse
|