Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell Biochem ; 478(6): 1231-1244, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36282352

ABSTRACT

Sodium fluoroacetate (FA) is a metabolic poison that systemically inhibits the tricarboxylic acid (TCA) cycle, causing energy deficiency and ultimately multi-organ failure. It poses a significant threat to society because of its high toxicity, potential use as a chemical weapon and lack of effective antidotal therapy. In this study, we investigated cell-permeable succinate prodrugs as potential treatment for acute FA intoxication. We hypothesized that succinate prodrugs would bypass FA-induced mitochondrial dysfunction, provide metabolic support, and prevent metabolic crisis during acute FA intoxication. To test this hypothesis, rats were exposed to FA (0.75 mg/kg) and treated with the succinate prodrug candidate NV354. Treatment efficacy was evaluated based on cardiac and cerebral mitochondrial respiration, mitochondrial content, metabolic profiles and tissue pathology. In the heart, FA increased concentrations of the TCA metabolite citrate (+ 4.2-fold, p < 0.01) and lowered ATP levels (- 1.9-fold, p < 0.001), confirming the inhibition of the TCA cycle by FA. High-resolution respirometry of cardiac mitochondria further revealed an impairment of mitochondrial complex V (CV)-linked metabolism, as evident by a reduced phosphorylation system control ratio (- 41%, p < 0.05). The inhibition of CV-linked metabolism is a novel mechanism of FA cardiac toxicity, which has implications for drug development and which NV354 was unable to counteract at the given dose. In the brain, FA induced the accumulation of ß-hydroxybutyrate (+ 1.4-fold, p < 0.05) and the reduction of mitochondrial complex I (CI)-linked oxidative phosphorylation (OXPHOSCI) (- 20%, p < 0.01), the latter of which was successfully alleviated by NV354. This promising effect of NV354 warrants further investigations to determine its potential neuroprotective effects.


Subject(s)
Prodrugs , Rats , Animals , Prodrugs/pharmacology , Prodrugs/metabolism , Succinic Acid/metabolism , Mitochondria/metabolism , Oxidative Phosphorylation , Electron Transport Complex I/metabolism , Fluoroacetates/pharmacology , Fluoroacetates/metabolism
2.
J Cardiothorac Vasc Anesth ; 36(7): 1985-1994, 2022 07.
Article in English | MEDLINE | ID: mdl-34969566

ABSTRACT

OBJECTIVES: The augmented inflammatory response to cardiac surgery is a recognized cause of postoperative acute kidney injury. The present study aimed to investigate the effects of preoperative cyclosporine treatment on cytokine production and delineate factors associated with postoperative kidney impairment. DESIGN: A randomized, double-blind, placebo-controlled, single-center study. SETTING: At a tertiary care, university hospital. PARTICIPANTS: Patients eligible for elective coronary artery bypass grafting surgery; 67 patients were enrolled. INTERVENTIONS: Patients were randomized to receive 2.5 mg/kg cyclosporine or placebo before surgery. Cytokine levels were measured after the induction of anesthesia and 4 hours after the end of cardiopulmonary bypass. MEASUREMENTS AND MAIN RESULTS: Tissue-aggressive (interleukin [IL]-1ß, macrophage inflammatory protein [MIP]-1ß, granulocyte colony-stimulating factor [G-CSF], IL-6, IL-8, IL-17, MCP-1), as well tissue-lenient (IL-4) cytokines, were significantly elevated in response to surgery. Changes in cytokine levels were not affected by cyclosporine pretreatment. CONCLUSIONS: Elective coronary artery bypass grafting surgery with cardiopulmonary bypass triggers cytokine activation. This activation was not impacted by preoperative cyclosporine treatment.


Subject(s)
Coronary Artery Bypass , Cyclosporine , Cardiopulmonary Bypass , Coronary Artery Bypass/adverse effects , Cyclosporine/pharmacology , Cytokines/pharmacology , Humans , Kidney/physiology
3.
Int J Mol Sci ; 22(1)2021 Jan 03.
Article in English | MEDLINE | ID: mdl-33401621

ABSTRACT

Statins are the cornerstone of lipid-lowering therapy. Although generally well tolerated, statin-associated muscle symptoms (SAMS) represent the main reason for treatment discontinuation. Mitochondrial dysfunction of complex I has been implicated in the pathophysiology of SAMS. The present study proposed to assess the concentration-dependent ex vivo effects of three statins on mitochondrial respiration in viable human platelets and to investigate whether a cell-permeable prodrug of succinate (complex II substrate) can compensate for statin-induced mitochondrial dysfunction. Mitochondrial respiration was assessed by high-resolution respirometry in human platelets, acutely exposed to statins in the presence/absence of the prodrug NV118. Statins concentration-dependently inhibited mitochondrial respiration in both intact and permeabilized cells. Further, statins caused an increase in non-ATP generating oxygen consumption (uncoupling), severely limiting the OXPHOS coupling efficiency, a measure of the ATP generating capacity. Cerivastatin (commercially withdrawn due to muscle toxicity) displayed a similar inhibitory capacity compared with the widely prescribed and tolerable atorvastatin, but did not elicit direct complex I inhibition. NV118 increased succinate-supported mitochondrial oxygen consumption in atorvastatin/cerivastatin-exposed platelets leading to normalization of coupled (ATP generating) respiration. The results acquired in isolated human platelets were validated in a limited set of experiments using atorvastatin in HepG2 cells, reinforcing the generalizability of the findings.


Subject(s)
Blood Platelets/physiology , Cell Respiration , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitochondria/physiology , Oxygen Consumption , Succinic Acid/pharmacology , Adult , Aged , Blood Platelets/drug effects , Female , Humans , Male , Mitochondria/drug effects
4.
Int J Mol Sci ; 21(16)2020 Aug 13.
Article in English | MEDLINE | ID: mdl-32823731

ABSTRACT

Oxidative stress is associated with many renal disorders, both acute and chronic, and has also been described to contribute to the disease progression. Therefore, oxidative stress is a potential therapeutic target. The human antioxidant α1-microglobulin (A1M) is a plasma and tissue protein with heme-binding, radical-scavenging and reductase activities. A1M can be internalized by cells, localized to the mitochondria and protect mitochondrial function. Due to its small size, A1M is filtered from the blood into the glomeruli, and taken up by the renal tubular epithelial cells. A1M has previously been described to reduce renal damage in animal models of preeclampsia, radiotherapy and rhabdomyolysis, and is proposed as a pharmacological agent for the treatment of kidney damage. In this paper, we examined the in vitro protective effects of recombinant human A1M (rA1M) in human proximal tubule epithelial cells. Moreover, rA1M was found to protect against heme-induced cell-death both in primary cells (RPTEC) and in a cell-line (HK-2). Expression of stress-related genes was upregulated in both cell cultures in response to heme exposure, as measured by qPCR and confirmed with in situ hybridization in HK-2 cells, whereas co-treatment with rA1M counteracted the upregulation. Mitochondrial respiration, analyzed with the Seahorse extracellular flux analyzer, was compromised following exposure to heme, but preserved by co-treatment with rA1M. Finally, heme addition to RPTE cells induced an upregulation of the endogenous cellular expression of A1M, via activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-pathway. Overall, data suggest that A1M/rA1M protects against stress-induced damage to tubule epithelial cells that, at least partly, can be attributed to maintaining mitochondrial function.


Subject(s)
Alpha-Globulins/pharmacology , Epithelial Cells/pathology , Heme/toxicity , Kidney Tubules, Proximal/pathology , Protective Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cytoprotection/drug effects , HSP70 Heat-Shock Proteins/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Stress, Physiological/drug effects , Up-Regulation/drug effects
5.
EMBO J ; 34(8): 1042-55, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25698262

ABSTRACT

Obesity is a major global public health problem, and understanding its pathogenesis is critical for identifying a cure. In this study, a gene knockout strategy was used in post-neonatal mice to delete synoviolin (Syvn)1/Hrd1/Der3, an ER-resident E3 ubiquitin ligase with known roles in homeostasis maintenance. Syvn1 deficiency resulted in weight loss and lower accumulation of white adipose tissue in otherwise wild-type animals as well as in genetically obese (ob/ob and db/db) and adipose tissue-specific knockout mice as compared to control animals. SYVN1 interacted with and ubiquitinated the thermogenic coactivator peroxisome proliferator-activated receptor coactivator (PGC)-1ß, and Syvn1 mutants showed upregulation of PGC-1ß target genes and increase in mitochondrion number, respiration, and basal energy expenditure in adipose tissue relative to control animals. Moreover, the selective SYVN1 inhibitor LS-102 abolished the negative regulation of PGC-1ß by SYVN1 and prevented weight gain in mice. Thus, SYVN1 is a novel post-translational regulator of PGC-1ß and a potential therapeutic target in obesity treatment.


Subject(s)
Body Weight/genetics , Mitochondria/physiology , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/physiology , 3T3-L1 Cells , Animals , Cells, Cultured , Down-Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
6.
N Engl J Med ; 373(11): 1021-31, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26321103

ABSTRACT

BACKGROUND: Experimental and clinical evidence suggests that cyclosporine may attenuate reperfusion injury and reduce myocardial infarct size. We aimed to test whether cyclosporine would improve clinical outcomes and prevent adverse left ventricular remodeling. METHODS: In a multicenter, double-blind, randomized trial, we assigned 970 patients with an acute anterior ST-segment elevation myocardial infarction (STEMI) who were undergoing percutaneous coronary intervention (PCI) within 12 hours after symptom onset and who had complete occlusion of the culprit coronary artery to receive a bolus injection of cyclosporine (administered intravenously at a dose of 2.5 mg per kilogram of body weight) or matching placebo before coronary recanalization. The primary outcome was a composite of death from any cause, worsening of heart failure during the initial hospitalization, rehospitalization for heart failure, or adverse left ventricular remodeling at 1 year. Adverse left ventricular remodeling was defined as an increase of 15% or more in the left ventricular end-diastolic volume. RESULTS: A total of 395 patients in the cyclosporine group and 396 in the placebo group received the assigned study drug and had data that could be evaluated for the primary outcome at 1 year. The rate of the primary outcome was 59.0% in the cyclosporine group and 58.1% in the control group (odds ratio, 1.04; 95% confidence interval [CI], 0.78 to 1.39; P=0.77). Cyclosporine did not reduce the incidence of the separate clinical components of the primary outcome or other events, including recurrent infarction, unstable angina, and stroke. No significant difference in the safety profile was observed between the two treatment groups. CONCLUSIONS: In patients with anterior STEMI who had been referred for primary PCI, intravenous cyclosporine did not result in better clinical outcomes than those with placebo and did not prevent adverse left ventricular remodeling at 1 year. (Funded by the French Ministry of Health and NeuroVive Pharmaceutical; CIRCUS ClinicalTrials.gov number, NCT01502774; EudraCT number, 2009-013713-99.).


Subject(s)
Cyclophilins/antagonists & inhibitors , Cyclosporine/administration & dosage , Enzyme Inhibitors/administration & dosage , Myocardial Infarction/drug therapy , Percutaneous Coronary Intervention , Ventricular Remodeling/drug effects , Aged , Combined Modality Therapy , Cyclosporine/adverse effects , Double-Blind Method , Electrocardiography , Enzyme Inhibitors/adverse effects , Female , Heart Failure/epidemiology , Humans , Injections, Intravenous , Kaplan-Meier Estimate , Male , Middle Aged , Mortality , Myocardial Infarction/therapy
7.
Anesthesiology ; 128(4): 710-717, 2018 04.
Article in English | MEDLINE | ID: mdl-29369890

ABSTRACT

BACKGROUND: Acute kidney injury is a common complication after cardiac surgery, leading to increased morbidity and mortality. One suggested cause for acute kidney injury is extracorporeal circulation-induced ischemia-reperfusion injury. In animal studies, cyclosporine has been shown to reduce ischemia-reperfusion injury in the kidneys. We hypothesized that administering cyclosporine before extracorporeal circulation could protect the kidneys in patients undergoing cardiac surgery. METHODS: The Cyclosporine to Protect Renal Function in Cardiac Surgery (CiPRICS) study was an investigator-initiated, double-blind, randomized, placebo-controlled, single-center study. The primary objective was to assess if cyclosporine could reduce acute kidney injury in patients undergoing coronary artery bypass grafting surgery with extracorporeal circulation. In the study, 154 patients with an estimated glomerular filtration rate of 15 to 90 ml · min · 1.73 m were enrolled. Study patients were randomized to receive 2.5 mg/kg cyclosporine or placebo intravenously before surgery. The primary endpoint was relative plasma cystatin C changes from the preoperative day to postoperative day 3. Secondary endpoints included biomarkers of kidney, heart, and brain injury. RESULTS: All enrolled patients were analyzed. The cyclosporine group (136.4 ± 35.6%) showed a more pronounced increase from baseline plasma cystatin C to day 3 compared to placebo (115.9 ± 30.8%), difference, 20.6% (95% CI, 10.2 to 31.2%, P < 0.001). The same pattern was observed for the other renal markers. The cyclosporine group had more patients in Risk Injury Failure Loss End-stage (RIFLE) groups R (risk), I (injury), or F (failure; 31% vs. 8%, P < 0.001). There were no differences in safety parameter distribution between groups. CONCLUSIONS: Administration of cyclosporine did not protect coronary artery bypass grafting patients from acute kidney injury. Instead, cyclosporine caused a decrease in renal function compared to placebo that resolved after 1 month.


Subject(s)
Acute Kidney Injury/epidemiology , Coronary Artery Bypass/trends , Cyclosporine/administration & dosage , Glomerular Filtration Rate/drug effects , Postoperative Complications/epidemiology , Preoperative Care/methods , Acute Kidney Injury/chemically induced , Acute Kidney Injury/physiopathology , Aged , Coronary Artery Bypass/methods , Cyclosporine/adverse effects , Double-Blind Method , Female , Glomerular Filtration Rate/physiology , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Postoperative Complications/chemically induced , Postoperative Complications/physiopathology , Preoperative Care/adverse effects
8.
Pediatr Res ; 83(2): 455-465, 2018 02.
Article in English | MEDLINE | ID: mdl-28981487

ABSTRACT

BackgroundDiagnosing mitochondrial disease (MD) is a challenge. In addition to genetic analyses, clinical practice is to perform invasive procedures such as muscle biopsy for biochemical and histochemical analyses. Blood cell respirometry is rapid and noninvasive. Our aim was to explore its possible role in diagnosing MD.MethodsBlood samples were collected from 113 pediatric patients, for whom MD was a differential diagnosis. A respiratory analysis model based on ratios (independent of mitochondrial specific content) was derived from a group of healthy controls and tested on the patients. The diagnostic accuracy of platelet respirometry was evaluated against routine diagnostic investigation.ResultsMD prevalence in the cohort was 16%. A ratio based on the respiratory response to adenosine diphosphate in the presence of complex I substrates had 96% specificity for disease and a positive likelihood ratio of 5.3. None of the individual ratios had sensitivity above 50%, but a combined model had 72% sensitivity.ConclusionNormal findings of platelet respirometry are not able to rule out MD, but pathological results make the diagnosis more likely and could strengthen the clinical decision to perform further invasive analyses. Our results encourage further study into the role of blood respirometry as an adjunct diagnostic tool for MD.


Subject(s)
Blood Platelets/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/blood , Mitochondrial Diseases/diagnosis , Oxygen Consumption , Biopsy , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Lactic Acid/blood , Male , Oxygen/chemistry , Prevalence , Sensitivity and Specificity
9.
J Nucl Cardiol ; 24(6): 1912-1921, 2017 12.
Article in English | MEDLINE | ID: mdl-27506700

ABSTRACT

BACKGROUND: Large body size can cause a higher proportion of emitted photons being attenuated within the patient. Therefore, clinical myocardial perfusion SPECT (MPS) protocols often include unproportionally higher radioisotope activity to obese patients. The aim was to evaluate if a linear weight-adjusted low-dose protocol can be applied to obese patients and thereby decrease radiation exposure. METHODS AND RESULT: Two hundred patients (>110 kg, BMI 18-41, [n = 69], ≤ 110 kg, BMI 31-58, [n = 131]) underwent 99mTc-tetrofosmin stress examination on a Cadmium Zinc Telluride or a conventional gamma camera using new generations of reconstruction algorithm (Resolution Recovery). Patients <110 kg were administered 2.5 MBq/kg, patients between 110 and 120 kg received 430 MBq and patients >120 kg received 570 MBq according to clinical routine. Patients >110 kg had 130% total number of counts in the images compared to patients <110 kg. Recalculating the counts to correspond to an administered activity of 2.5 MBq/kg resulted in similar number of counts across the groups. Image analyses in a subgroup with images corresponding to high activity and 2.5 MBq/kg showed no difference in image quality or ischemia quantification. CONCLUSION: Linear low-dose weight-adjusted protocol of 2.5 MBq/kg in MPS can be applied over a large weight span without loss of counts or image quality, resulting in a significant reduction in radiation exposure to obese patients.


Subject(s)
Myocardial Perfusion Imaging , Overweight/diagnostic imaging , Radiation Dosage , Tomography, Emission-Computed, Single-Photon , Aged , Body Weight , Clinical Protocols , Female , Humans , Male , Middle Aged , Organophosphorus Compounds , Organotechnetium Compounds , Radiation Exposure
10.
Am Heart J ; 169(6): 758-766.e6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26027612

ABSTRACT

BACKGROUND: Both acute myocardial ischemia and reperfusion contribute to cardiomyocyte death in ST-elevation myocardial infarction (STEMI). The final infarct size is the principal determinant of subsequent clinical outcome in STEMI patients. In a proof-of-concept phase II trial, the administration of cyclosporine prior to primary percutaneous coronary intervention (PPCI) has been associated with a reduction of infarct size in STEMI patients. METHODS: CIRCUS is an international, prospective, multicenter, randomized, double-blinded, placebo-controlled trial. The study is designed to compare the efficacy and safety of cyclosporine versus placebo, in addition to revascularization by PPCI, in patients presenting with acute anterior myocardial infarction within 12 hours of symptoms onset and initial TIMI flow ≤1 in the culprit left anterior descending coronary artery. Patients are randomized in a 1:1 fashion to 2.5 mg/kg intravenous infusion of cyclosporine or matching placebo performed in the minutes preceding PCI. The primary efficacy end point of CIRCUS is a composite of 1-year all-cause mortality, rehospitalization for heart failure or heart failure worsening during initial hospitalization, and left ventricular adverse remodeling as determined by sequential transthoracic echochardiography. Secondary outcomes will be tested using a hierarchical sequence of left ventricular (LV) ejection fraction and absolute measurements of LV volumes. The composite of death and rehospitalization for heart failure or heart failure worsening during initial hospitalization will be further assessed at three years after the initial infarction. RESULTS: Recruitment lasted from April 2011 to February 2014. The CIRCUS trial has recruited 975 patients with acute anterior myocardial infarction. The 12-months results are expected to be available in 2015. CONCLUSIONS: The CIRCUS trial is testing the hypothesis that cyclosporine in addition to early revascularization with PPCI compared to placebo in patients with acute anterior myocardial infarction reduces the incidence of death, heart failure and adverse LV remodeling at one-year follow-up.


Subject(s)
Cyclosporine/adverse effects , Cyclosporine/therapeutic use , Myocardial Infarction/surgery , Percutaneous Coronary Intervention , Biomarkers/blood , Coronary Angiography , Double-Blind Method , Echocardiography , Electrocardiography , Female , Humans , Male , Myocardial Infarction/physiopathology , Prospective Studies , Time Factors , Treatment Outcome
11.
J Nucl Cardiol ; 21(4): 695-702, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24807623

ABSTRACT

INTRODUCTION: Myocardial perfusion single photon emission computed tomography (MPS) is one of the most widely used diagnostic methods in patients with suspected ischemic heart disease (IHD). Recently, a novel technique based on cadmium-zinc-telluride (CZT) detectors, pinhole collimators, and a stationary gantry was introduced for MPS. The aim of this work was to investigate how patient positioning affects the reconstructed MPS images using this novel technique. MATERIALS AND METHODS: Eighteen patients referred for a clinical MPS due to suspected IHD were included in the study. All patients underwent MPS imaging on a GE Discovery NM 530c CZT camera. After image acquisition with the heart positioned in the center of the quality field of view (QFOV), the patients were re-imaged in different positions 5-20 mm off-center. The heart was still positioned within the limits of the QFOV during the off-center scans. The summed stress score and/or the summed rest score (SSS and/or SRS) for the acquisition performed in the center was compared to the same parameter for the acquisitions performed off-center. RESULTS: There was a statistically significant increase in SSS and/or SRS when imaging was performed with the heart 5-20 mm outside the center of the QFOV compared to optimal positioning (7.7 ± 1.3 vs 6.6 ± 1.3, P = .006). The SSS and/or SRS increased with ≥2 U in 35% (14/40) of the off-center examinations. CONCLUSION: It is important to carefully position the patient's heart within the center of the QFOV when performing MPS with the Discovery NM 530c CZT camera to avoid positioning-related image artifacts that could affect the diagnostic accuracy.


Subject(s)
Gamma Cameras , Myocardial Perfusion Imaging/methods , Patient Positioning , Tomography, Emission-Computed, Single-Photon/methods , Aged , Aged, 80 and over , Cadmium , Female , Humans , Male , Middle Aged , Tellurium , Zinc
12.
Heliyon ; 10(5): e26745, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38439844

ABSTRACT

There is a growing interest for the possibility of using peripheral blood cells (including platelets) as markers for mitochondrial function in less accessible tissues. Only a few studies have examined the correlation between respiration in blood and muscle tissue, with small sample sizes and conflicting results. This study investigated the correlation of mitochondrial respiration within and across tissues. Additional analyses were performed to elucidate which blood cell type would be most useful for assessing systemic mitochondrial function. There was a significant but weak within tissue correlation between platelets and peripheral blood mononuclear cells (PBMCs). Neither PBMCs nor platelet respiration correlated significantly with muscle respiration. Muscle fibers from a group of athletes had higher mass-specific respiration, due to higher mitochondrial content than non-athlete controls, but this finding was not replicated in either of the blood cell types. In a group of patients with primary mitochondrial diseases, there were significant differences in blood cell respiration compared to healthy controls, particularly in platelets. Platelet respiration generally correlated better with the citrate synthase activity of each sample, in comparison to PBMCs. In conclusion, this study does not support the theory that blood cells can be used as accurate biomarkers to detect minor alterations in muscle respiration. However, in some instances, pronounced mitochondrial abnormalities might be reflected across tissues and detectable in blood cells, with more promising findings for platelets than PBMCs.

13.
BMC Cell Biol ; 14: 40, 2013 Sep 22.
Article in English | MEDLINE | ID: mdl-24053891

ABSTRACT

BACKGROUND: Ischemic preconditioning has been proposed to involve changes in mitochondrial H(+) and K(+) fluxes, in particular through activation of uncoupling proteins and ATP-sensitive K(+) channels (MitoKATP). The objectives of the present study were to explore how increased H(+) and K(+) fluxes influence heart mitochondrial physiology with regard to production and scavenging of reactive oxygen species (ROS), volume changes and resistance to calcium-induced mitochondrial permeability transition (mPT). RESULTS: Isolated rat heart mitochondria were exposed to a wide concentration range of the protonophore CCCP or the potassium ionophore valinomycin to induce increased H(+) and K(+) conductance, respectively. Simultaneous monitoring of mitochondrial respiration and calcium retention capacity (CRC) demonstrated that the relative increase in respiration caused by valinomycin or CCCP correlated with a decrease in CRC, and that no level of respiratory uncoupling was associated with enhanced resistance to mPT. Mitochondria suspended in hyperosmolar buffer demonstrated a dose-dependent reduction in CRC with increasing osmolarity. However, mitochondria in hypoosmolar buffer to increase matrix volume did not display increased CRC. ROS generation was reduced by both K(+)- and H(+)-mediated respiratory uncoupling. The ability of heart mitochondria to detoxify H2O2 was substantially greater than the production rate. The H2O2 detoxification was dependent on respiratory substrates and was dramatically decreased following calcium-induced mPT, but was unaffected by uncoupling via increased K(+) and H(+) conductance. CONCLUSION: It is concluded that respiratory uncoupling is not directly beneficial to rat heart mitochondrial resistance to calcium overload irrespective of whether H(+) or K(+) conductance is increased. The negative effects of respiratory uncoupling thus probably outweigh the reduction in ROS generation and a potential positive effect by increased matrix volume, resulting in a net sensitization of heart mitochondria to mPT activation.


Subject(s)
Mitochondria, Heart/metabolism , Oxygen/metabolism , Potassium Channels/metabolism , Potassium/metabolism , Protons , Reactive Oxygen Species/metabolism , Animals , Calcium/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cations, Monovalent , Cyclosporine/pharmacology , Diazoxide/pharmacology , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Osmolar Concentration , Oxidative Phosphorylation/drug effects , Permeability , Proton Ionophores/pharmacology , Rats , Uncoupling Agents/pharmacology , Valinomycin/pharmacology
14.
Crit Care ; 17(4): R152, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23883738

ABSTRACT

INTRODUCTION: In sepsis, mitochondria have been associated with both initial dysfunction and subsequent upregulation (biogenesis). However, the evolvement of mitochondrial function in sepsis over time is largely unknown, and we therefore investigated mitochondrial respiration in peripheral blood immune cells (PBICs) in sepsis patients during the first week after admission to the intensive care unit (ICU). METHODS: PBICs from 20 patients with severe sepsis or septic shock were analyzed with high-resolution respirometry 3 times after admission to the ICU (within 48 hours, days 3 to 4 and days 6 to 7). Mitochondrial DNA (mtDNA), cytochrome c (Cyt c), and citrate synthase (CS) were measured as indicators of cellular mitochondrial content. RESULTS: In intact PBICs with endogenous substrates, a gradual increase in cellular respiration reached 173% of controls after 1 week (P = 0.001). In permeabilized cells, respiration using substrates of complex I, II, and IV were significantly increased days 1 to 2, reaching 137%, 130%, and 173% of controls, respectively. In parallel, higher levels of CS activity, mtDNA, and Cyt c content in PBICs (211%, 243%, and 331% of controls for the respective indicators were found at days 6 to 7; P < 0.0001). No differences in respiratory capacities were noted between survivors and nonsurvivors at any of the time points measured. CONCLUSIONS: PBICs from patients with sepsis displayed higher mitochondrial respiratory capacities compared with controls, due to an increased mitochondrial content, as indicated by increased mitochondrial DNA, protein content, and enzyme activity. The results argue against mitochondrial respiratory dysfunction in this type of cells in sepsis.


Subject(s)
Intensive Care Units , Leukocytes, Mononuclear/metabolism , Mitochondria/metabolism , Sepsis/metabolism , Adult , Aged , Cell Respiration/physiology , Female , Humans , Immunity, Cellular/physiology , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Mitochondria/immunology , Sepsis/immunology , Young Adult
15.
Acta Neurochir Suppl ; 118: 311-5, 2013.
Article in English | MEDLINE | ID: mdl-23564156

ABSTRACT

Traumatic brain injury and ischemia can result in marked neuronal degeneration and residual impairment of cerebral function. However, no effective pharmacological treatment directed at tissues of the central nervous system (CNS) for acute intervention has been developed. The detailed pathophysiological cascade leading to -neurodegeneration in these conditions has not been elucidated, but cellular calcium overload and mitochondrial dysfunction have been implicated in a wide range of animal models involving degeneration of the CNS. In particular, activation of the calcium-induced mitochondrial permeability transition (mPT) is considered to be a major cause of cell death inferred by the broad and potent neuroprotective effects of -pharmacological inhibitors of mPT, especially modulators of cyclophilin activity and, more specifically, genetic inactivation of the mitochondrial cyclophilin, cyclophilin D. Reviewed are evidence and challenges that could bring on the dawning of mitochondrial medicine aimed at safeguarding energy supply following acute injury to the CNS.


Subject(s)
Cyclophilins/metabolism , Mitochondria/drug effects , Neuroprotective Agents , Animals , Arsenicals/pharmacology , Arsenicals/therapeutic use , Brain Injuries/drug therapy , Calcium/metabolism , Peptidyl-Prolyl Isomerase F , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Ischemia/drug therapy
16.
Neurotherapeutics ; 20(6): 1482-1495, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37561274

ABSTRACT

Drug development in traumatic brain injury (TBI) has been impeded by the complexity and heterogeneity of the disease pathology, as well as limited understanding of the secondary injury cascade that follows the initial trauma. As a result, patients with TBI have an unmet need for effective pharmacological therapies. One promising drug candidate is cyclosporine, a polypeptide traditionally used to achieve immunosuppression in transplant recipients. Cyclosporine inhibits mitochondrial permeability transition, thereby reducing secondary brain injury, and has shown neuroprotective effects in multiple preclinical models of TBI. Moreover, the cyclosporine formulation NeuroSTAT® displayed positive effects on injury biomarker levels in patients with severe TBI enrolled in the Phase Ib/IIa Copenhagen Head Injury Ciclosporin trial (NCT01825044). Future research on neuroprotective compounds such as cyclosporine should take advantage of recent advances in fluid-based biomarkers and neuroimaging to select patients with similar disease pathologies for clinical trials. This would increase statistical power and allow for more accurate assessment of long-term outcomes.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Neuroprotective Agents , Humans , Cyclosporine/therapeutic use , Cyclosporine/pharmacology , Brain Injuries, Traumatic/drug therapy , Brain Injuries/drug therapy , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Biomarkers
17.
Sci Rep ; 12(1): 20329, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36434021

ABSTRACT

Pesticides account for hundreds of millions of cases of acute poisoning worldwide each year, with organophosphates (OPs) being responsible for the majority of all pesticide-related deaths. OPs inhibit the enzyme acetylcholinesterase (AChE), which leads to impairment of the central- and peripheral nervous system. Current standard of care (SOC) alleviates acute neurologic-, cardiovascular- and respiratory symptoms and reduces short term mortality. However, survivors often demonstrate significant neurologic sequelae. This highlights the critical need for further development of adjunctive therapies with novel targets. While the inhibition of AChE is thought to be the main mechanism of injury, mitochondrial dysfunction and resulting metabolic crisis may contribute to the overall toxicity of these agents. We hypothesized that the mitochondrially targeted succinate prodrug NV354 would support mitochondrial function and reduce brain injury during acute intoxication with the OP diisopropylfluorophosphate (DFP). To this end, we developed a rat model of acute DFP intoxication and evaluated the efficacy of NV354 as adjunctive therapy to SOC treatment with atropine and pralidoxime. We demonstrate that NV354, in combination with atropine and pralidoxime therapy, significantly improved cerebral mitochondrial complex IV-linked respiration and reduced signs of brain injury in a rodent model of acute DFP exposure.


Subject(s)
Brain Injuries , Organophosphate Poisoning , Prodrugs , Animals , Rats , Organophosphate Poisoning/drug therapy , Atropine/pharmacology , Atropine/therapeutic use , Prodrugs/pharmacology , Isoflurophate/toxicity , Succinic Acid , Acetylcholinesterase/metabolism , Rodentia/metabolism , Succinates , Mitochondria/metabolism , Brain Injuries/drug therapy
18.
J Biol Chem ; 285(1): 741-50, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19880514

ABSTRACT

Modulation of K(+) conductance of the inner mitochondrial membrane has been proposed to mediate preconditioning in ischemia-reperfusion injury. The mechanism is not entirely understood, but it has been linked to a decreased activation of mitochondrial permeability transition (mPT). In the present study K(+) channel activity was mimicked by picomolar concentrations of valinomycin. Isolated brain mitochondria were exposed to continuous infusions of calcium. Monitoring of extramitochondrial Ca(2+) and mitochondrial respiration provided a quantitative assay for mPT sensitivity by determining calcium retention capacity (CRC). Valinomycin and cyclophilin D inhibition separately and additively increased CRC. Comparable degrees of respiratory uncoupling induced by increased K(+) or H(+) conductance had opposite effects on mPT sensitivity. Protonophores dose-dependently decreased CRC, demonstrating that so-called mild uncoupling was not beneficial per se. The putative mitoK(ATP) channel opener diazoxide did not mimic the effect of valinomycin. An alkaline matrix pH was required for mitochondria to retain calcium, but increased K(+) conductance did not result in augmented DeltapH. The beneficial effect of valinomycin on CRC was not mediated by H(2)O(2)-induced protein kinase Cepsilon activation. Rather, increased K(+) conductance reduced H(2)O(2) generation during calcium infusion. Lowering the osmolarity of the buffer induced an increase in mitochondrial volume and improved CRC similar to valinomycin without inducing uncoupling or otherwise affecting respiration. We propose that increased potassium conductance in brain mitochondria may cause a direct physiological effect on matrix volume inducing resistance to pathological calcium challenges.


Subject(s)
Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Size , Potassium/metabolism , Alkalies/metabolism , Animals , Calcium/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Respiration/drug effects , Diazoxide/pharmacology , Enzyme Activation/drug effects , Hydrogen/metabolism , Hydrogen Peroxide/pharmacology , Hydrogen-Ion Concentration/drug effects , Ion Transport/drug effects , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Size/drug effects , Molecular Mimicry/drug effects , Nigericin/pharmacology , Potassium Channels/metabolism , Protein Kinase C/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Valinomycin/pharmacology
19.
J Neurotrauma ; 38(13): 1870-1878, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33191835

ABSTRACT

All phase III trials evaluating medical treatments for traumatic brain injury (TBI), performed to date, have failed. To facilitate future success there is a need for novel outcome metrics that can bridge pre-clinical studies to clinical proof of concept trials. Our objective was to assess diffusion tensor imaging (DTI) and biofluid-based biomarkers as efficacy outcome metrics in a large animal study evaluating the efficacy of cyclosporine in TBI. This work builds on our previously published study that demonstrated a reduced volume of injury by 35% with cyclosporine treatment based on magnetic resonance imaging (MRI) results. A focal contusion injury was induced in piglets using a controlled cortical impact (CCI) device. Cyclosporine in a novel Cremophor/Kolliphor EL-free lipid emulsion, NeuroSTAT, was administered by continuous intravenous infusion for 5 days. The animals underwent DTI on day 5. Glial fibrillary acidic protein (GFAP), as a measure of astroglia injury, and neurofilament light (NF-L), as a measure of axonal injury, were measured in blood on days 1, 2, and 5, and in cerebrospinal fluid (CSF) on day 5 post-injury. Normalized fractional anisotropy (FA) was significantly (p = 0.027) higher in in the treatment group, indicating preserved tissue integrity with treatment. For the biomarkers, we observed a statistical trend of a decreased level of NF-L in CSF (p = 0.051), in the treatment group relative to placebo, indicating less axonal injury. Our findings suggest that DTI, and possibly CSF NF-L, may be feasible as translational end-points assessing neuroprotective drugs in TBI.


Subject(s)
Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Cyclosporine/therapeutic use , Diffusion Tensor Imaging/standards , Animals , Animals, Newborn , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Brain Injuries, Traumatic/drug therapy , Diffusion Tensor Imaging/methods , Female , Immunosuppressive Agents/therapeutic use , Swine
20.
Crit Care ; 14(6): R214, 2010.
Article in English | MEDLINE | ID: mdl-21106065

ABSTRACT

INTRODUCTION: Mitochondrial dysfunction has been suggested as a contributing factor to the pathogenesis of sepsis-induced multiple organ failure. Also, restoration of mitochondrial function, known as mitochondrial biogenesis, has been implicated as a key factor for the recovery of organ function in patients with sepsis. Here we investigated temporal changes in platelet mitochondrial respiratory function in patients with sepsis during the first week after disease onset. METHODS: Platelets were isolated from blood samples taken from 18 patients with severe sepsis or septic shock within 48 hours of their admission to the intensive care unit. Subsequent samples were taken on Day 3 to 4 and Day 6 to 7. Eighteen healthy blood donors served as controls. Platelet mitochondrial function was analyzed by high-resolution respirometry. Endogenous respiration of viable, intact platelets suspended in their own plasma or phosphate-buffered saline (PBS) glucose was determined. Further, in order to investigate the role of different dehydrogenases and respiratory complexes as well as to evaluate maximal respiratory activity of the mitochondria, platelets were permeabilized and stimulated with complex-specific substrates and inhibitors. RESULTS: Platelets suspended in their own septic plasma exhibited increased basal non-phosphorylating respiration (state 4) compared to controls and to platelets suspended in PBS glucose. In parallel, there was a substantial increase in respiratory capacity of the electron transfer system from Day 1 to 2 to Day 6 to 7 as well as compared to controls in both intact and permeabilized platelets oxidizing Complex I and/or II-linked substrates. No inhibition of respiratory complexes was detected in septic patients compared to controls. Non-survivors, at 90 days, had a more elevated respiratory capacity at Day 6 to 7 as compared to survivors. Cytochrome c increased over the time interval studied but no change in mitochondrial DNA was detected. CONCLUSIONS: The results indicate the presence of a soluble plasma factor in the initial stage of sepsis inducing uncoupling of platelet mitochondria without inhibition of the electron transfer system. The mitochondrial uncoupling was paralleled by a gradual and substantial increase in respiratory capacity. This may reflect a compensatory response to severe sepsis or septic shock, that was most pronounced in non-survivors, likely correlating to the severity of the septic insult.


Subject(s)
Blood Platelets/physiology , Cell Respiration/physiology , Mitochondria/pathology , Sepsis/metabolism , Sepsis/pathology , Adult , Aged , Female , Humans , Male , Middle Aged , Sepsis/mortality , Survival Rate/trends , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL