Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Resuscitation ; 195: 110121, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38272387
2.
FASEB J ; 38(1): e23379, 2024 01.
Article in English | MEDLINE | ID: mdl-38133921

ABSTRACT

Dynamin-related protein 1 (Drp1) is a cytosolic GTPase protein that when activated translocates to the mitochondria, meditating mitochondrial fission and increasing reactive oxygen species (ROS) in cardiomyocytes. Drp1 has shown promise as a therapeutic target for reducing cardiac ischemia/reperfusion (IR) injury; however, the lack of specificity of some small molecule Drp1 inhibitors and the reliance on the use of Drp1 haploinsufficient hearts from older mice have left the role of Drp1 in IR in question. Here, we address these concerns using two approaches, using: (a) short-term (3 weeks), conditional, cardiomyocyte-specific, Drp1 knockout (KO) and (b) a novel, highly specific Drp1 GTPase inhibitor, Drpitor1a. Short-term Drp1 KO mice exhibited preserved exercise capacity and cardiac contractility, and their isolated cardiac mitochondria demonstrated increased mitochondrial complex 1 activity, respiratory coupling, and calcium retention capacity compared to controls. When exposed to IR injury in a Langendorff perfusion system, Drp1 KO hearts had preserved contractility, decreased reactive oxygen species (ROS), enhanced mitochondrial calcium capacity, and increased resistance to mitochondrial permeability transition pore (MPTP) opening. Pharmacological inhibition of Drp1 with Drpitor1a following ischemia, but before reperfusion, was as protective as Drp1 KO for cardiac function and mitochondrial calcium homeostasis. In contrast to the benefits of short-term Drp1 inhibition, prolonged Drp1 ablation (6 weeks) resulted in cardiomyopathy. Drp1 KO hearts were also associated with decreased ryanodine receptor 2 (RyR2) protein expression and pharmacological inhibition of the RyR2 receptor decreased ROS in post-IR hearts suggesting that changes in RyR2 may have a role in Drp1 KO mediated cardioprotection. We conclude that Drp1-mediated increases in myocardial ROS production and impairment of mitochondrial calcium handling are key mechanisms of IR injury. Short-term inhibition of Drp1 is a promising strategy to limit early myocardial IR injury which is relevant for the therapy of acute myocardial infarction, cardiac arrest, and heart transplantation.


Subject(s)
Dynamins , Myocardial Infarction , Myocardial Reperfusion Injury , Animals , Mice , Calcium/metabolism , Dynamins/metabolism , Homeostasis , Mitochondria, Heart/metabolism , Mitochondrial Dynamics , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
3.
Genes (Basel) ; 14(7)2023 06 28.
Article in English | MEDLINE | ID: mdl-37510271

ABSTRACT

Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, with septic cardiomyopathy being a common and severe complication. Despite its significant clinical impact, the molecular mechanisms underlying sepsis-induced cardiomyopathy (SICM) remain incompletely understood. In this study, we performed a comparative analysis of whole transcriptome profiles using RNA sequencing in mouse hearts in two widely used mouse models of septic cardiomyopathy. CLP-induced sepsis was achieved by surgical cecal ligation and puncture, while LPS-induced sepsis was induced using a 5 mg/kg intraperitoneal (IP) injection of lipopolysaccharide (LPS). For consistency, we utilized sham-operated mice as the control for septic models. Our aim was to identify key genes and pathways involved in the development of septic cardiomyopathy and to evaluate the similarities and differences between the two models. Our findings demonstrated that both the CLP and lipopolysaccharide LPS methods could induce septic heart dysfunction within 24 h. We identified common transcriptional regulatory regions in the septic hearts of both models, such as Nfkb1, Sp1, and Jun. Moreover, differentially expressed genes (DEGs) in comparison to control were involved in shared pathways, including regulation of inflammatory response, regulation of reactive oxygen species metabolic process, and the JAK-STAT signaling pathway. However, each model presented distinctive whole transcriptome expression profiles and potentially diverse pathways contributing to sepsis-induced heart failure. This extensive comparison enhances our understanding of the molecular basis of septic cardiomyopathy, providing invaluable insights. Accordingly, our study also contributes to the pursuit of effective and personalized treatment strategies for SICM, highlighting the importance of considering the specific causative factors.


Subject(s)
Cardiomyopathies , Sepsis , Mice , Animals , Lipopolysaccharides/toxicity , Transcriptome , Cardiomyopathies/genetics , Sepsis/complications , Sepsis/genetics , Sepsis/drug therapy , Heart
5.
J Am Heart Assoc ; 12(13): e029131, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37345832

ABSTRACT

Background As partial pressure of oxygen (pO2) rises with the first breath, the ductus arteriosus (DA) constricts, diverting blood flow to the pulmonary circulation. The DA's O2 sensor resides within smooth muscle cells. The DA smooth muscle cells' mitochondrial electron transport chain (ETC) produces reactive oxygen species (ROS) in proportion to oxygen tension, causing vasoconstriction by regulating redox-sensitive ion channels and enzymes. To identify which ETC complex contributes most to DA O2 sensing and determine whether ROS mediate O2 sensing independent of metabolism, we used electron leak suppressors, S1QEL (suppressor of site IQ electron leak) and S3QEL (suppressor of site IIIQo electron leak), which decrease ROS production by inhibiting electron leak from quinone sites IQ and IIIQo, respectively. Methods and Results The effects of S1QEL, S3QEL, and ETC inhibitors (rotenone and antimycin A) on DA tone, mitochondrial metabolism, O2-induced changes in intracellular calcium, and ROS were studied in rabbit DA rings, and human and rabbit DA smooth muscle cells. S1QEL's effects on DA patency were assessed in rabbit kits, using micro computed tomography. In DA rings, S1QEL, but not S3QEL, reversed O2-induced constriction (P=0.0034) without reducing phenylephrine-induced constriction. S1QEL did not inhibit mitochondrial metabolism or ETC-I activity. In human DA smooth muscle cells, S1QEL and rotenone inhibited O2-induced increases in intracellular calcium (P=0.02 and 0.001, respectively), a surrogate for DA constriction. S1QEL inhibited O2-induced ROS generation (P=0.02). In vivo, S1QEL prevented O2-induced DA closure (P<0.0001). Conclusions S1QEL, but not S3QEL, inhibited O2-induced rises in ROS and DA constriction ex vivo and in vivo. DA O2 sensing relies on pO2-dependent changes in electron leak at site IQ in ETC-I, independent of metabolism. S1QEL offers a therapeutic means to maintain DA patency.


Subject(s)
Ductus Arteriosus , Animals , Humans , Rabbits , Oxygen , Reactive Oxygen Species/metabolism , Electron Transport , Calcium/metabolism , Electrons , Rotenone/metabolism , Rotenone/pharmacology , X-Ray Microtomography
7.
bioRxiv ; 2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36993497

ABSTRACT

RATIONALE: Cardiac microvascular leakage and inflammation are triggered during myocardial infarction (MI) and contribute to heart failure. Hypoxia-inducible factor 2α (Hif2α) is highly expressed in endothelial cells (ECs) and rapidly activated by myocardial ischemia, but whether it has a role in endothelial barrier function during MI is unclear. OBJECTIVE: To test our hypothesis that the expression of Hif2α and its binding partner aryl hydrocarbon nuclear translocator (ARNT) in ECs regulate cardiac microvascular permeability in infarcted hearts. METHODS AND RESULTS: Experiments were conducted with mice carrying an inducible EC-specific Hif2α-knockout (ecHif2α-/-) mutation, with mouse cardiac microvascular endothelial cells (CMVECs) isolated from the hearts of ecHif2α-/- mice after the mutation was induced, and with human CMVECs and umbilical-vein endothelial cells transfected with ecHif2α siRNA. After MI induction, echocardiographic assessments of cardiac function were significantly lower, while measures of cardiac microvascular leakage (Evans blue assay), plasma IL6 levels, and cardiac neutrophil accumulation and fibrosis (histology) were significantly greater, in ecHif2α-/- mice than in control mice, and RNA-sequencing analysis of heart tissues from both groups indicated that the expression of genes involved in vascular permeability and collagen synthesis was enriched in ecHif2α-/- hearts. In cultured ECs, ecHif2α deficiency was associated with declines in endothelial barrier function (electrical cell impedance assay) and the reduced abundance of tight-junction proteins, as well as an increase in the expression of inflammatory markers, all of which were largely reversed by the overexpression of ARNT. We also found that ARNT, but not Hif2α, binds directly to the IL6 promoter and suppresses IL6 expression. CONCLUSIONS: EC-specific deficiencies in Hif2α expression significantly increase cardiac microvascular permeability, promote inflammation, and reduce cardiac function in infarcted mouse hearts, and ARNT overexpression can reverse the upregulation of inflammatory genes and restore endothelial-barrier function in Hif2α-deficient ECs.

9.
Elife ; 112022 07 13.
Article in English | MEDLINE | ID: mdl-35822617

ABSTRACT

Cellular metabolism is a critical regulator of macrophage effector function. Tissue-resident alveolar macrophages (TR-AMs) inhabit a unique niche marked by high oxygen and low glucose. We have recently shown that in contrast to bone marrow-derived macrophages (BMDMs), TR-AMs do not utilize glycolysis and instead predominantly rely on mitochondrial function for their effector response. It is not known how changes in local oxygen concentration that occur during conditions such as acute respiratory distress syndrome (ARDS) might affect TR-AM metabolism and function; however, ARDS is associated with progressive loss of TR-AMs, which correlates with the severity of disease and mortality. Here, we demonstrate that hypoxia robustly stabilizes HIF-1α in TR-AMs to promote a glycolytic phenotype. Hypoxia altered TR-AM metabolite signatures, cytokine production, and decreased their sensitivity to the inhibition of mitochondrial function. By contrast, hypoxia had minimal effects on BMDM metabolism. The effects of hypoxia on TR-AMs were mimicked by FG-4592, a HIF-1α stabilizer. Treatment with FG-4592 decreased TR-AM death and attenuated acute lung injury in mice. These findings reveal the importance of microenvironment in determining macrophage metabolic phenotype and highlight the therapeutic potential in targeting cellular metabolism to improve outcomes in diseases characterized by acute inflammation.


Subject(s)
Acute Lung Injury , Respiratory Distress Syndrome , Animals , Cell Survival , Glycolysis , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages, Alveolar/metabolism , Mice , Oxygen/metabolism
10.
Neurocrit Care ; 36(1): 61-70, 2022 02.
Article in English | MEDLINE | ID: mdl-34268646

ABSTRACT

BACKGROUND: Neurological injury following successful resuscitation from sudden cardiac arrest (CA) is common. The pathophysiological basis of this injury remains poorly understood, and treatment options are limited. Microglial activation and neuroinflammation are established contributors to many neuropathologies, such as Alzheimer disease and traumatic brain injury, but their potential role in post-CA injury has only recently been recognized. Here, we hypothesize that microglial activation that occurs following brief asystolic CA is associated with neurological injury and represents a potential therapeutic target. METHODS: Adult C57BL/6 male and female mice were randomly assigned to 12-min, KCl-induced asystolic CA, under anesthesia and ventilation, followed by successful cardiopulmonary resuscitation (n = 19) or sham intervention (n = 11). Neurological assessments of mice were performed using standardized neurological scoring, video motion tracking, and sensory/motor testing. Mice were killed at 72 h for histological studies; neuronal degeneration was assessed using Fluoro-Jade C staining. Microglial characteristics were assessed by immunohistochemistry using the marker of ionized calcium binding adaptor molecule 1, followed by ImageJ analyses for cell integrity density and skeletal analyses. RESULTS: Neurological injury in post-cardiopulmonary-resuscitation mice vs. sham mice was evident by poorer neurological scores (difference of 3.626 ± 0.4921, 95% confidence interval 2.618-4.634), sensory and motor functions (worsened by sixfold and sevenfold, respectively, compared with baseline), and locomotion (75% slower with a 76% decrease in total distance traveled). Post-CA brains demonstrated evidence of neurodegeneration and neuroinflammatory microglial activation. CONCLUSIONS: Extensive microglial activation and neurodegeneration in the CA1 region and the dentate gyrus of the hippocampus are evident following brief asystolic CA and are associated with severe neurological injury.


Subject(s)
Cardiopulmonary Resuscitation , Heart Arrest , Animals , Disease Models, Animal , Female , Heart Arrest/complications , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism
11.
Acad Emerg Med ; 29(1): 118-122, 2022 01.
Article in English | MEDLINE | ID: mdl-34250678

ABSTRACT

Emergency medicine (EM) investigators lag in research funding from the National Institutes of Health (NIH) when compared to other specialties. NIH funding determinations are made in part by a process of NIH study section peer review. Low participation by EM investigators in NIH peer review could be one explanation for low levels of NIH funding by EM investigators. The objective of this study was to establish a current-state metric of EM faculty researchers serving on standing NIH study sections from 2019 to 2020. Publicly available lists of NIH study section membership rosters within the Center for Scientific Review and within individual NIH institutions were reviewed for standing members. Committee members listed as being members of a department of emergency medicine were identified as emergency care researchers. Special emphasis panels and ad hoc members were excluded. Members degrees were categorized as PhD, MD (with or without non-PhD degree), MD/PhD, and other. Similar analysis was performed of AHRQ study sections. A total of 6,113 members on NIH study sections were identified. Degrees held by committee members included PhDs 74% (4,547), MDs 14%(883), MD/PhDs 10% (584), and other (99). Twenty (0.3%) NIH study section members were identified as members of an emergency department (ED). A total of 20% (four) held PhDs, 75% (15) held MDs, and 5%(one) held MD/PhD degrees. A total of 25% (five) of EM faculty were pediatric and 75% (15) were adult. Clustering of study sections within similar institutions was noted with 40% (two) of the pediatric faculty at the same institution while 27% (four) of the adult faculty were at the same institution. AHRQ study section review identified 3% (four/127) as members of an ED. Our data show that 20 EM faculty comprised 0.3% of NIH standing study section members and four EM faculty comprised 3% of AHRQ standing study section members from 2019 to 2020 and that these members were clustered at a few institutions.


Subject(s)
Emergency Medicine , National Institutes of Health (U.S.) , Adult , Child , Faculty , Humans , United States
12.
Neurocrit Care ; 34(1): 64-72, 2021 02.
Article in English | MEDLINE | ID: mdl-32358767

ABSTRACT

BACKGROUND: Cardiac arrest (CA) patients who survived by cardiopulmonary resuscitation (CPR) can present different levels of neurological deficits ranging from minor cognitive impairments to persistent vegetative state and brain death. The pathophysiology of the resulting brain injury is poorly understood, and whether changes in post-CA brain metabolism contribute to the injury are unknown. Here we utilized [18F]fluorodeoxyglucose (FDG)-Positron emission tomography (PET) to study in vivo cerebral glucose metabolism 72 h following CA in a murine CA model. METHODS: Anesthetized and ventilated adult C57BL/6 mice underwent 12-min KCl-induced CA followed by CPR. Seventy-two hours following CA, surviving mice were intraperitoneally injected with [18F]FDG (~ 186 µCi/200 µL) and imaged on Molecubes preclinical micro-PET/computed tomography (CT) imaging systems after a 30-min awake uptake period. Brain [18F]FDG uptake was determined by the VivoQuant software on fused PET/CT images with the 3D brain atlas. Upon completion of Positron emission tomography (PET) imaging, remaining [18F]FDG radioactivity in the brain, heart, and liver was determined using a gamma counter. RESULTS: Global increases in brain [18F]FDG uptake in post-CA mice were observed compared to shams and controls. The median standardized uptake value of [18F]FDG for CA animals was 1.79 versus sham 1.25 (p < 0.05) and control animals 0.78 (p < 0.01). This increased uptake was consistent throughout the 60-min imaging period and across all brain regions reaching statistical significance in the midbrain, pons, and medulla. Biodistribution analyses of various key organs yielded similar observations that the median [18F]FDG uptake for brain was 7.04%ID/g tissue for CA mice versus 5.537%ID/g tissue for sham animals, p < 0.05). CONCLUSIONS: This study has successfully applied [18F]FDG-PET/CT to measure changes in brain metabolism in a murine model of asystolic CA. Our results demonstrate increased [18F]FDG uptake in the brain 72 h following CA, suggesting increased metabolic demand in the case of severe neurological injury. Further study is warranted to determine the etiology of these changes.


Subject(s)
Fluorodeoxyglucose F18 , Heart Arrest , Animals , Brain/diagnostic imaging , Glucose , Heart Arrest/diagnostic imaging , Humans , Mice , Mice, Inbred C57BL , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography , Radiopharmaceuticals , Tissue Distribution
13.
West J Emerg Med ; 22(4): 979-987, 2021 Jul 20.
Article in English | MEDLINE | ID: mdl-35354003

ABSTRACT

INTRODUCTION: Patients with coronavirus disease 2019 (COVID-19) can develop rapidly progressive respiratory failure. Ventilation strategies during the COVID-19 pandemic seek to minimize patient mortality. In this study we examine associations between the availability of emergency department (ED)-initiated high-flow nasal cannula (HFNC) for patients presenting with COVID-19 respiratory distress and outcomes, including rates of endotracheal intubation (ETT), mortality, and hospital length of stay. METHODS: We performed a retrospective, non-concurrent cohort study of patients with COVID-19 respiratory distress presenting to the ED who required HFNC or ETT in the ED or within 24 hours following ED departure. Comparisons were made between patients presenting before and after the introduction of an ED-HFNC protocol. RESULTS: Use of HFNC was associated with a reduced rate of ETT in the ED (46.4% vs 26.3%, P <0.001) and decreased the cumulative proportion of patients who required ETT within 24 hours of ED departure (85.7% vs 32.6%, P <0.001) or during their entire hospitalization (89.3% vs 48.4%, P <0.001). Using HFNC was also associated with a trend toward increased survival to hospital discharge; however, this was not statistically significant (50.0% vs 68.4%, P = 0.115). There was no impact on intensive care unit or hospital length of stay. Demographics, comorbidities, and illness severity were similar in both cohorts. CONCLUSIONS: The institution of an ED-HFNC protocol for patients with COVID-19 respiratory distress was associated with reductions in the rate of ETT. Early initiation of HFNC is a promising strategy for avoiding ETT and improving outcomes in patients with COVID-19.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , COVID-19/therapy , Cannula , Cohort Studies , Emergency Service, Hospital , Humans , Pandemics , Retrospective Studies
15.
Crit Care Med ; 48(2): e133-e140, 2020 02.
Article in English | MEDLINE | ID: mdl-31939812

ABSTRACT

OBJECTIVES: Cardiogenic shock following cardiopulmonary resuscitation for sudden cardiac arrest is common, occurring even in the absence of acute coronary artery occlusion, and contributes to high rates of postcardiopulmonary resuscitation mortality. The pathophysiology of this shock is unclear, and effective therapies for improving clinical outcomes are lacking. DESIGN: Laboratory investigation. SETTING: University laboratory. SUBJECTS: C57BL/6 adult female mice. INTERVENTIONS: Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent a 4, 8, 12, or 16-minute potassium chloride-induced cardiac arrest followed by 90 seconds of cardiopulmonary resuscitation. Mice were then blindly randomized to a single IV injection of vehicle (phosphate-buffered saline) or suppressor of site IQ electron leak, an inhibitor of superoxide production by complex I of the mitochondrial electron transport chain. Suppressor of site IQ electron leak and vehicle were administered during cardiopulmonary resuscitation. MEASUREMENTS AND MAIN RESULTS: Using a murine model of asystolic cardiac arrest, we discovered that duration of cardiac arrest prior to cardiopulmonary resuscitation determined postresuscitation success rates, degree of neurologic injury, and severity of myocardial dysfunction. Post-cardiopulmonary resuscitation cardiac dysfunction was not associated with myocardial necrosis, apoptosis, inflammation, or mitochondrial permeability transition pore opening. Furthermore, left ventricular function recovered within 72 hours of cardiopulmonary resuscitation, indicative of myocardial stunning. Postcardiopulmonary resuscitation, the myocardium exhibited increased reactive oxygen species and evidence of mitochondrial injury, specifically reperfusion-induced reactive oxygen species generation at electron transport chain complex I. Suppressor of site IQ electron leak, which inhibits complex I-dependent reactive oxygen species generation by suppression of site IQ electron leak, decreased myocardial reactive oxygen species generation and improved postcardiopulmonary resuscitation myocardial function, neurologic outcomes, and survival. CONCLUSIONS: The severity of cardiogenic shock following asystolic cardiac arrest is dependent on the length of cardiac arrest prior to cardiopulmonary resuscitation and is mediated by myocardial stunning resulting from mitochondrial electron transport chain complex I dysfunction. A novel pharmacologic agent targeting this mechanism, suppressor of site IQ electron leak, represents a potential, practical therapy for improving sudden cardiac arrest resuscitation outcomes.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Heart Arrest/therapy , Hydrogen Peroxide/antagonists & inhibitors , Mitochondria/drug effects , Myocardial Stunning/prevention & control , Superoxides/antagonists & inhibitors , Animals , Cardiopulmonary Resuscitation , Female , Heart Arrest/physiopathology , Mice , Mice, Inbred C57BL , Myocardial Stunning/physiopathology , Random Allocation , Reactive Oxygen Species/metabolism
16.
Resusc Plus ; 3: 100017, 2020 Sep.
Article in English | MEDLINE | ID: mdl-34223300

ABSTRACT

BACKGROUND: Cardiopulmonary resuscitation (CPR) in adults following non-traumatic out of hospital cardiac arrest (OHCA) can cause thoracic complications including rib fractures, sternal fractures, and pneumothorax. Post-CPR complication rates are poorly studied and the optimum imaging modality to detect these complications post-resuscitation has not been established. METHODS: We performed a retrospective review of adult patients transported to a single, urban, academic hospital following atraumatic OHCA between September 2015 and January 2020. Patients who achieved sustained return of spontaneous circulation (ROSC) and who underwent computed tomography (CT) imaging of the chest following radiographic chest x-ray were included in the analyses. Patient demographics and prehospital data were collected. Descriptive statistics and multivariate logistic regression analysis were performed. Sensitivity and specificity of chest x-ray for the detection of thoracic injury in this population were estimated. RESULTS: 786 non-traumatic OHCA patients were transported to the ED, 417 of whom obtained sustained ROSC and were admitted to the hospital (53%). 137 (32.9%) admitted patients underwent CT imaging of the chest in the ED. Of these imaged patients median age was 62 years old (IQR 53-70) with 54.0% female and 38.0% of patients having received bystander CPR. 40/137 (29.2%) patients had skeletal fractures noted on CT imaging and 12/137 (8.8%) had pneumothorax present on CT imaging. X-ray yielded a sensitivity of 7.5% for rib fracture and 50% for pneumothorax with a specificity of 100% for both. Logistic regression analysis revealed no significant association between age, sex, bystander CPR, or resuscitation length with thoracic fractures or pneumothorax. CONCLUSIONS: Complications from OHCA CPR were high with 29.2% of CT imaged patients having rib fractures and 8.8% having pneumothoraces. X-ray had poor sensitivity for these post-resuscitation complications. Post-CPR CT imaging of the chest should be considered for detecting post-CPR complications.

17.
Resuscitation ; 136: 126-130, 2019 03.
Article in English | MEDLINE | ID: mdl-30716427

ABSTRACT

BACKGROUND: Extracorporeal cardiopulmonary resuscitation (ECPR) is a resource-intensive tool that provides haemodynamic and respiratory support in patients who have suffered cardiac arrest. In this study, we investigated the cost-utility of ECPR (cost/QALY) in cardiac arrest patients treated at our institution. METHODS: We performed a retrospective review of patients who received ECPR following cardiac arrest between 2012 and 2018. All medical care-associated charges with ECPR and subsequent hospital admission were recorded. The quality-of-life of survivors was assessed with the Health Utilities Index Mark II. The cost-utility of ECPR was calculated with cost and quality-of-life data. RESULTS: ECPR was used in 32 patients (15/32 in-hospital, 47%) with a median age of 55.0 years (IQR 46.3-63.3 years), 59% male and 66% African American. The median duration of ECPR support was 2.1 days (IQR 0.9-3.8 days). Survival to hospital discharge was 16%. The median score of the Health Utilities Index Mark II at discharge for the survivors was 0.44 (IQR 0.32-0.52). The median operating cost for patients undergoing ECMO was $125,683 per patient (IQR $49,751-$206,341 per patient). The calculated cost-utility for ECPR was $56,156/QALY gained. CONCLUSIONS: The calculated cost-utility is within the threshold considered cost-effective in the United States (<$150,000/QALY gained). These results are comparable to the cost-effectiveness of heart transplantation for end-stage heart failure. Larger studies are needed to assess the cost-utility of ECPR and to identify whether other factors, such as patient characteristics, affect the cost-utility benefit.


Subject(s)
Cardiopulmonary Resuscitation/methods , Extracorporeal Membrane Oxygenation/economics , Hospital Costs/statistics & numerical data , Out-of-Hospital Cardiac Arrest/therapy , Adult , Aged , Cost-Benefit Analysis , Extracorporeal Membrane Oxygenation/mortality , Female , Humans , Length of Stay/economics , Male , Middle Aged , Out-of-Hospital Cardiac Arrest/economics , Out-of-Hospital Cardiac Arrest/mortality , Quality-Adjusted Life Years , Registries , Retrospective Studies
18.
PLoS One ; 12(9): e0185046, 2017.
Article in English | MEDLINE | ID: mdl-28934276

ABSTRACT

RATIONALE: Post-ischemic changes in cellular metabolism alter myocardial and neurological function. Pyruvate dehydrogenase (PDH), the limiting step in mitochondrial glucose oxidation, is inhibited by increased expression of PDH kinase (PDK) during ischemia/reperfusion injury. This results in decreased utilization of glucose to generate cellular ATP. Post-cardiac arrest (CA) hypothermia improves outcomes and alters metabolism, but its influence on PDH and PDK activity following CA are unknown. We hypothesized that therapeutic hypothermia (TH) following CA is associated with the inhibition of PDK activity and increased PDH activity. We further hypothesized that an inhibitor of PDK activity, dichloroacetate (DCA), would improve PDH activity and post-CA outcomes. METHODS AND RESULTS: Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent a 12-minute KCl-induced CA followed by cardiopulmonary resuscitation. Compared to normothermic (37°C) CA controls, administering TH (30°C) improved overall survival (72-hour survival rate: 62.5% vs. 28.6%, P<0.001), post-resuscitation myocardial function (ejection fraction: 50.9±3.1% vs. 27.2±2.0%, P<0.001; aorta systolic pressure: 132.7±7.3 vs. 72.3±3.0 mmHg, P<0.001), and neurological scores at 72-hour post CA (9.5±1.3 vs. 5.4±1.3, P<0.05). In both heart and brain, CA increased lactate concentrations (1.9-fold and 3.1-fold increase, respectively, P<0.01), decreased PDH enzyme activity (24% and 50% reduction, respectively, P<0.01), and increased PDK protein expressions (1.2-fold and 1.9-fold, respectively, P<0.01). In contrast, post-CA treatment with TH normalized lactate concentrations (P<0.01 and P<0.05) and PDK expressions (P<0.001 and P<0.05), while increasing PDH activity (P<0.01 and P<0.01) in both the heart and brain. Additionally, treatment with DCA (0.2 mg/g body weight) 30 min prior to CA improved both myocardial hemodynamics 2 hours post-CA (aortic systolic pressure: 123±3 vs. 96±4 mmHg, P<0.001) and 72-hour survival rates (50% vs. 19%, P<0.05) in normothermic animals. CONCLUSIONS: Enhanced PDH activity in the setting of TH or DCA administration is associated with improved post-CA resuscitation outcomes. PDH is a promising therapeutic target for improving post-CA outcomes.


Subject(s)
Dichloroacetic Acid/therapeutic use , Heart Arrest/therapy , Hypothermia, Induced , Pyruvate Dehydrogenase Complex/metabolism , Animals , Cardiopulmonary Resuscitation , Combined Modality Therapy , Female , Heart Arrest/enzymology , Heart Arrest/mortality , Hemodynamics , Mice , Mice, Inbred C57BL , Survival Rate
19.
Oncotarget ; 8(21): 34082-34098, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28423723

ABSTRACT

Doxorubicin is the chemotherapeutic drug of choice for a wide variety of cancers, and cardiotoxicity is one of the major side effects of doxorubicin treatment. One of the main cellular targets of doxorubicin in the heart is mitochondria. Mitochondrial sirtuin, SIRT3 has been shown to protect against doxorubicin-induced cardiotoxicity. We have recently identified honokiol (HKL) as an activator of SIRT3, which protects the heart from developing pressure overload hypertrophy. Here, we show that HKL-mediated activation of SIRT3 also protects the heart from doxorubicin-induced cardiac damage without compromising the tumor killing potential of doxorubicin. Doxorubicin-induced cardiotoxicity is associated with increased ROS production and consequent fragmentation of mitochondria and cell death. HKL-mediated activation of SIRT3 prevented Doxorubicin induced ROS production, mitochondrial damage and cell death in rat neonatal cardiomyocytes. HKL also promoted mitochondrial fusion. We also show that treatment with HKL blocked doxorubicin-induced cardiac toxicity in mice. This was associated with reduced mitochondrial DNA damage and improved mitochondrial function. Furthermore, treatments of mice, bearing prostrate tumor-xenografts, with HKL and doxorubicin showed inhibition of tumor growth with significantly reduced cardiac toxicity. Our results suggest that HKL-mediated activation of SIRT3 protects the heart from doxorubicin-induced cardiotoxicity and represents a potentially novel adjunct for chemotherapy treatments.


Subject(s)
Biphenyl Compounds/administration & dosage , Cardiomyopathies/prevention & control , Doxorubicin/adverse effects , Lignans/administration & dosage , Mitochondria, Heart/drug effects , Animals , Biphenyl Compounds/pharmacology , Cardiomyopathies/chemically induced , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Lignans/pharmacology , Mice , Mitochondria, Heart/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Reactive Oxygen Species/metabolism , Sirtuin 3 , Up-Regulation
20.
J Mol Med (Berl) ; 95(4): 381-393, 2017 04.
Article in English | MEDLINE | ID: mdl-28265681

ABSTRACT

Right ventricular (RV) function determines prognosis in pulmonary arterial hypertension (PAH). We hypothesize that ischemia causes RV dysfunction in PAH by triggering dynamin-related protein 1 (Drp1)-mediated mitochondrial fission. RV function was compared in control rats (n = 50) versus rats with monocrotaline-induced PAH (MCT-PAH; n = 60) both in vivo (echocardiography) and ex vivo (RV Langendorff). Mitochondrial membrane potential and morphology and RV function were assessed before or after 2 cycles of ischemia-reperfusion injury challenge (RV-IR). The effects of Mdivi-1 (25 µM), a Drp1 GTPase inhibitor, and P110 (1 µM), a peptide inhibitor of Drp1-Fis1 interaction, were studied. We found that MCT caused RV hypertrophy, RV vascular rarefaction, and RV dysfunction. Prior to IR, the mitochondria in MCT-PAH RV were depolarized and swollen with increased Drp1 content and reduced aconitase activity. RV-IR increased RV end diastolic pressure (RVEDP) and mitochondrial Drp1 expression in both control and MCT-PAH RVs. IR depolarized mitochondria in control RV but did not exacerbate the basally depolarized MCT-PAH RV mitochondria. During RV IR mdivi-1 and P110 reduced Drp1 translocation to mitochondria, improved mitochondrial structure and function, and reduced RVEDP. In conclusion, RV ischemia occurs in PAH and causes Drp1-Fis1-mediated fission leading to diastolic dysfunction. Inhibition of mitochondrial fission preserves RV function in RV-IR. KEY MESSAGES: Right ventricular ischemia reperfusion (RV-IR) causes RV diastolic dysfunction. IR-induced mitochondrial fission causes RV diastolic dysfunction. In RV-IR Drp1 translocates to mitochondria, binds Fis1 and causes fission and injury. A baseline RV mitochondriopathy in MCT PAH resembles IR-induced mitochondrial injury. Drp1 inhibitors (Mdivi-1 and P110) preserve RV diastolic function post RV-IR.


Subject(s)
Dynamins/metabolism , Hypertension, Pulmonary/etiology , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Myocardial Reperfusion Injury/complications , Ventricular Dysfunction, Right/etiology , Animals , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Mitochondria/metabolism , Mitochondria/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Rats, Sprague-Dawley , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/pathology , Ventricular Dysfunction, Right/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...