Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(1): 105470, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38118236

ABSTRACT

The prevailing notion that reduced cofactors NADH and FADH2 transfer electrons from the tricarboxylic acid cycle to the mitochondrial electron transfer system creates ambiguities regarding respiratory Complex II (CII). CII is the only membrane-bound enzyme in the tricarboxylic acid cycle and is part of the electron transfer system of the mitochondrial inner membrane feeding electrons into the coenzyme Q-junction. The succinate dehydrogenase subunit SDHA of CII oxidizes succinate and reduces the covalently bound prosthetic group FAD to FADH2 in the canonical forward tricarboxylic acid cycle. However, several graphical representations of the electron transfer system depict FADH2 in the mitochondrial matrix as a substrate to be oxidized by CII. This leads to the false conclusion that FADH2 from the ß-oxidation cycle in fatty acid oxidation feeds electrons into CII. In reality, dehydrogenases of fatty acid oxidation channel electrons to the Q-junction but not through CII. The ambiguities surrounding Complex II in the literature and educational resources call for quality control, to secure scientific standards in current communications of bioenergetics, and ultimately support adequate clinical applications. This review aims to raise awareness of the inherent ambiguity crisis, complementing efforts to address the well-acknowledged issues of credibility and reproducibility.


Subject(s)
Electron Transport Complex II , Electrons , Fatty Acids , Flavin-Adenine Dinucleotide , Succinate Dehydrogenase , Electron Transport , Fatty Acids/chemistry , Fatty Acids/metabolism , Flavin-Adenine Dinucleotide/analogs & derivatives , Flavin-Adenine Dinucleotide/metabolism , Oxidation-Reduction , Reproducibility of Results , Succinate Dehydrogenase/metabolism , Citric Acid Cycle , Mitochondria/metabolism , Ubiquinone/metabolism , Succinic Acid/metabolism , Electron Transport Complex II/metabolism , Energy Metabolism
2.
J Exp Biol ; 227(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38099471

ABSTRACT

Diapause exhibited by embryos of Artemia franciscana is accompanied by severe arrest of respiration. A large fraction of this depression is attributable to downregulation of trehalose catabolism that ultimately restricts fuel to mitochondria. This study now extends knowledge on the mechanism by revealing metabolic depression is heightened by inhibitions within mitochondria. Compared with that in embryo lysates during post-diapause, oxidative phosphorylation (OXPHOS) capacity P is depressed during diapause when either NADH-linked substrates (pyruvate and malate) for electron transfer (electron transfer capacity, E) through respiratory Complex I or the Complex II substrate succinate are used. When pyruvate, malate and succinate were combined, respiratory inhibition by the phosphorylation system in diapause lysates was discovered as judged by P/E flux control ratios (two-way ANOVA; F1,24=38.78; P<0.0001). Inhibition was eliminated as the diapause extract was diluted (significant interaction term; F2,24=9.866; P=0.0007), consistent with the presence of a diffusible inhibitor. One candidate is long-chain acyl-CoA esters known to inhibit the adenine nucleotide translocator. Addition of oleoyl-CoA to post-diapause lysates markedly decreased the P/E ratio to 0.40±0.07 (mean±s.d.; P=0.002) compared with 0.79±0.11 without oleoyl-CoA. Oleoyl-CoA inhibits the phosphorylation system and may be responsible for the depressed P/E in lysates from diapause embryos. With isolated mitochondria, depression of P/E by oleoyl-CoA was fully reversed by addition of l-carnitine (control versus recovery with l-carnitine, P=0.338), which facilitates oleoyl-CoA transport into the matrix and elimination by ß-oxidation. In conclusion, severe metabolic arrest during diapause promoted by restricting glycolytic carbon to mitochondria is reinforced by depression of OXPHOS capacity and the phosphorylation system.


Subject(s)
Diapause , Extremophiles , Animals , Oxidative Phosphorylation , Artemia/physiology , Malates , Pyruvates , Succinates , Carnitine
3.
Transpl Int ; 37: 12380, 2024.
Article in English | MEDLINE | ID: mdl-38463463

ABSTRACT

Donor organ biomarkers with sufficient predictive value in liver transplantation (LT) are lacking. We herein evaluate liver viability and mitochondrial bioenergetics for their predictive capacity towards the outcome in LT. We enrolled 43 consecutive patients undergoing LT. Liver biopsy samples taken upon arrival after static cold storage were assessed by histology, real-time confocal imaging analysis (RTCA), and high-resolution respirometry (HRR) for mitochondrial respiration of tissue homogenates. Early allograft dysfunction (EAD) served as primary endpoint. HRR data were analysed with a focus on the efficacy of ATP production or P-L control efficiency, calculated as 1-L/P from the capacity of oxidative phosphorylation P and non-phosphorylating respiration L. Twenty-two recipients experienced EAD. Pre-transplant histology was not predictive of EAD. The mean RTCA score was significantly lower in the EAD cohort (-0.75 ± 2.27) compared to the IF cohort (0.70 ± 2.08; p = 0.01), indicating decreased cell viability. P-L control efficiency was predictive of EAD (0.76 ± 0.06 in IF vs. 0.70 ± 0.08 in EAD-livers; p = 0.02) and correlated with the RTCA score. Both RTCA and P-L control efficiency in biopsy samples taken during cold storage have predictive capacity towards the outcome in LT. Therefore, RTCA and HRR should be considered for risk stratification, viability assessment, and bioenergetic testing in liver transplantation.


Subject(s)
Liver Transplantation , Primary Graft Dysfunction , Humans , Liver Transplantation/adverse effects , Graft Survival , Risk Factors , Liver/pathology , Energy Metabolism , Allografts/pathology , Primary Graft Dysfunction/etiology
4.
Mamm Genome ; 34(2): 229-243, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36565314

ABSTRACT

Ubiquinol cytochrome c reductase hinge protein (UQCRH) is required for the electron transfer between cytochrome c1 and c of the mitochondrial cytochrome bc1 Complex (CIII). A two-exon deletion in the human UQCRH gene has recently been identified as the cause for a rare familial mitochondrial disorder. Deletion of the corresponding gene in the mouse (Uqcrh-KO) resulted in striking biochemical and clinical similarities including impairment of CIII, failure to thrive, elevated blood glucose levels, and early death. Here, we set out to test how global ablation of the murine Uqcrh affects cardiac morphology and contractility, and bioenergetics. Hearts from Uqcrh-KO mutant mice appeared macroscopically considerably smaller compared to wildtype littermate controls despite similar geometries as confirmed by transthoracic echocardiography (TTE). Relating TTE-assessed heart to body mass revealed the development of subtle cardiac enlargement, but histopathological analysis showed no excess collagen deposition. Nonetheless, Uqcrh-KO hearts developed pronounced contractile dysfunction. To assess mitochondrial functions, we used the high-resolution respirometer NextGen-O2k allowing measurement of mitochondrial respiratory capacity through the electron transfer system (ETS) simultaneously with the redox state of ETS-reactive coenzyme Q (Q), or production of reactive oxygen species (ROS). Compared to wildtype littermate controls, we found decreased mitochondrial respiratory capacity and more reduced Q in Uqcrh-KO, indicative for an impaired ETS. Yet, mitochondrial ROS production was not generally increased. Taken together, our data suggest that Uqcrh-KO leads to cardiac contractile dysfunction at 9 weeks of age, which is associated with impaired bioenergetics but not with mitochondrial ROS production. Global ablation of the Uqcrh gene results in functional impairment of CIII associated with metabolic dysfunction and postnatal developmental arrest immediately after weaning from the mother. Uqcrh-KO mice show dramatically elevated blood glucose levels and decreased ability of isolated cardiac mitochondria to consume oxygen (O2). Impaired development (failure to thrive) after weaning manifests as a deficiency in the gain of body mass and growth of internal organ including the heart. The relative heart mass seemingly increases when organ mass calculated from transthoracic echocardiography (TTE) is normalized to body mass. Notably, the heart shows no signs of collagen deposition, yet does develop a contractile dysfunction reflected by a decrease in ejection fraction and fractional shortening.


Subject(s)
Blood Glucose , Failure to Thrive , Humans , Mice , Animals , Reactive Oxygen Species/metabolism , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Mice, Knockout , Energy Metabolism/genetics , Transcription Factors/metabolism
5.
Int J Mol Sci ; 23(9)2022 May 04.
Article in English | MEDLINE | ID: mdl-35563503

ABSTRACT

The oxidation of proline to pyrroline-5-carboxylate (P5C) leads to the transfer of electrons to ubiquinone in mitochondria that express proline dehydrogenase (ProDH). This electron transfer supports Complexes CIII and CIV, thus generating the protonmotive force. Further catabolism of P5C forms glutamate, which fuels the citric acid cycle that yields the reducing equivalents that sustain oxidative phosphorylation. However, P5C and glutamate catabolism depend on CI activity due to NAD+ requirements. NextGen-O2k (Oroboros Instruments) was used to measure proline oxidation in isolated mitochondria of various mouse tissues. Simultaneous measurements of oxygen consumption, membrane potential, NADH, and the ubiquinone redox state were correlated to ProDH activity and F1FO-ATPase directionality. Proline catabolism generated a sufficiently high membrane potential that was able to maintain the F1FO-ATPase operation in the forward mode. This was observed in CI-inhibited mouse liver and kidney mitochondria that exhibited high levels of proline oxidation and ProDH activity. This action was not observed under anoxia or when either CIII or CIV were inhibited. The duroquinone fueling of CIII and CIV partially reproduced the effects of proline. Excess glutamate, however, could not reproduce the proline effect, suggesting that processes upstream of the glutamate conversion from proline were involved. The ProDH inhibitors tetrahydro-2-furoic acid and, to a lesser extent, S-5-oxo-2-tetrahydrofurancarboxylic acid abolished all proline effects. The data show that ProDH-directed proline catabolism could generate sufficient CIII and CIV proton pumping, thus supporting ATP production by the F1FO-ATPase even under CI inhibition.


Subject(s)
Proline Oxidase , Ubiquinone , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Electron Transport Complex I/metabolism , Glutamic Acid/metabolism , Mice , Mitochondria/metabolism , Proline/metabolism , Proline Oxidase/metabolism , Ubiquinone/metabolism
6.
J Lipid Res ; 62: 100111, 2021.
Article in English | MEDLINE | ID: mdl-34450173

ABSTRACT

The molecular assembly of cells depends not only on the balance between anabolism and catabolism but to a large degree on the building blocks available in the environment. For cultured mammalian cells, this is largely determined by the composition of the applied growth medium. Here, we study the impact of lipids in the medium on mitochondrial membrane architecture and function by combining LC-MS/MS lipidomics and functional tests with lipid supplementation experiments in an otherwise serum-free and lipid-free cell culture model. We demonstrate that the composition of mitochondrial cardiolipins strongly depends on the lipid environment in cultured cells and favors the incorporation of essential linoleic acid over other fatty acids. Simultaneously, the mitochondrial respiratory complex I activity was altered, whereas the matrix-localized enzyme citrate synthase was unaffected. This raises the question on a link between membrane composition and respiratory control. In summary, we found a strong dependency of central mitochondrial features on the type of lipids contained in the growth medium. This underlines the importance of considering these factors when using and establishing cell culture models in biomedical research. In summary, we found a strong dependency of central mitochondrial features on the type of lipids contained in the growth medium.


Subject(s)
Cardiolipins/metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , Animals , Chromatography, High Pressure Liquid , HeLa Cells , Humans , Swine , Tandem Mass Spectrometry , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 115(16): 4158-4163, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29618609

ABSTRACT

Current strategies used to quantitatively describe the biological diversity of lipids by mass spectrometry are often limited in assessing the exact structural variability of individual molecular species in detail. A major challenge is represented by the extensive isobaric overlap present among lipids, hampering their accurate identification. This is especially true for cardiolipins, a mitochondria-specific class of phospholipids, which are functionally involved in many cellular functions, including energy metabolism, cristae structure, and apoptosis. Substituted with four fatty acyl side chains, cardiolipins offer a particularly high potential to achieve complex mixtures of molecular species. Here, we demonstrate how systematically generated high-performance liquid chromatography-mass spectral data can be utilized in a mathematical structural modeling approach, to comprehensively analyze and characterize the molecular diversity of mitochondrial cardiolipin compositions in cell culture and disease models, cardiolipin modulation experiments, and a broad variety of frequently studied model organisms.


Subject(s)
Cardiolipins/chemistry , Membrane Lipids/chemistry , Mitochondrial Membranes/chemistry , Animals , Bacteria/chemistry , Barth Syndrome/metabolism , Cardiolipins/isolation & purification , Cell Line , Chromatography, High Pressure Liquid , Fatty Acids/analysis , Fibroblasts/chemistry , Fungi/chemistry , Humans , Membrane Lipids/isolation & purification , Mice , Models, Molecular , Molecular Structure , Plants/chemistry , RAW 264.7 Cells , Tandem Mass Spectrometry , Vertebrates/metabolism
8.
Proc Natl Acad Sci U S A ; 114(24): 6382-6387, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28533386

ABSTRACT

The Himalayan Sherpas, a human population of Tibetan descent, are highly adapted to life in the hypobaric hypoxia of high altitude. Mechanisms involving enhanced tissue oxygen delivery in comparison to Lowlander populations have been postulated to play a role in such adaptation. Whether differences in tissue oxygen utilization (i.e., metabolic adaptation) underpin this adaptation is not known, however. We sought to address this issue, applying parallel molecular, biochemical, physiological, and genetic approaches to the study of Sherpas and native Lowlanders, studied before and during exposure to hypobaric hypoxia on a gradual ascent to Mount Everest Base Camp (5,300 m). Compared with Lowlanders, Sherpas demonstrated a lower capacity for fatty acid oxidation in skeletal muscle biopsies, along with enhanced efficiency of oxygen utilization, improved muscle energetics, and protection against oxidative stress. This adaptation appeared to be related, in part, to a putatively advantageous allele for the peroxisome proliferator-activated receptor A (PPARA) gene, which was enriched in the Sherpas compared with the Lowlanders. Our findings suggest that metabolic adaptations underpin human evolution to life at high altitude, and could have an impact upon our understanding of human diseases in which hypoxia is a feature.


Subject(s)
Adaptation, Physiological , Altitude , Ethnicity , Hypoxia/metabolism , Adaptation, Physiological/genetics , Adult , Atmospheric Pressure , Citric Acid Cycle , Energy Metabolism , Ethnicity/genetics , Fatty Acids/metabolism , Female , Gene Frequency , Glucose/metabolism , Glycolysis , Humans , Hypoxia/genetics , Hypoxia/physiopathology , Male , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Nepal , Nitric Oxide/blood , Oxidative Phosphorylation , Oxidative Stress , Oxygen Consumption , PPAR alpha/genetics , PPAR alpha/metabolism , Polymorphism, Single Nucleotide , Tibet/ethnology
9.
J Biol Chem ; 293(18): 6659-6671, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29540485

ABSTRACT

Metabolic responses to hypoxia play important roles in cell survival strategies and disease pathogenesis in humans. However, the homeostatic adjustments that balance changes in energy supply and demand to maintain organismal function under chronic low oxygen conditions remain incompletely understood, making it difficult to distinguish adaptive from maladaptive responses in hypoxia-related pathologies. We integrated metabolomic and proteomic profiling with mitochondrial respirometry and blood gas analyses to comprehensively define the physiological responses of skeletal muscle energy metabolism to 16 days of high-altitude hypoxia (5260 m) in healthy volunteers from the AltitudeOmics project. In contrast to the view that hypoxia down-regulates aerobic metabolism, results show that mitochondria play a central role in muscle hypoxia adaptation by supporting higher resting phosphorylation potential and enhancing the efficiency of long-chain acylcarnitine oxidation. This directs increases in muscle glucose toward pentose phosphate and one-carbon metabolism pathways that support cytosolic redox balance and help mitigate the effects of increased protein and purine nucleotide catabolism in hypoxia. Muscle accumulation of free amino acids favor these adjustments by coordinating cytosolic and mitochondrial pathways to rid the cell of excess nitrogen, but might ultimately limit muscle oxidative capacity in vivo Collectively, these studies illustrate how an integration of aerobic and anaerobic metabolism is required for physiological hypoxia adaptation in skeletal muscle, and highlight protein catabolism and allosteric regulation as unexpected orchestrators of metabolic remodeling in this context. These findings have important implications for the management of hypoxia-related diseases and other conditions associated with chronic catabolic stress.


Subject(s)
Acclimatization , Altitude Sickness/metabolism , Altitude Sickness/physiopathology , Altitude , Energy Metabolism/physiology , Metabolome , Muscle, Skeletal/metabolism , Proteomics , Amino Acids/metabolism , Carnitine/analogs & derivatives , Carnitine/metabolism , Fatty Acids/metabolism , Female , Glycolysis , Healthy Volunteers , Humans , Male , Mitochondria, Muscle/metabolism , Muscle Proteins/metabolism , Oxidation-Reduction , Pentose Phosphate Pathway , Phosphorylation , Proteolysis , Purine Nucleotides/metabolism , Random Allocation , Stress, Physiological , Young Adult
10.
Mov Disord ; 34(1): 114-123, 2019 01.
Article in English | MEDLINE | ID: mdl-30311259

ABSTRACT

BACKGROUND: Restless legs syndrome is a sensorimotor neurological disorder of the limbs that impairs quality of life and disturbs sleep. However, there has been progress in understanding the disease involving the dopaminergic system as well as iron metabolism. The exact pathophysiological mechanisms of restless legs syndrome remain elusive. We tried to elucidate the underlying mechanisms in iron metabolism in restless legs syndrome subjects on a systemic, cellular, and mitochondrial level. METHODS: We conducted a study prospectively recruiting 168 restless legs syndrome patients and 119 age-matched healthy controls focusing on iron metabolism using human monocytes as surrogates. RESULTS: Evaluation of systemic iron metabolism parameters in the circulation showed no significant difference between patients and controls. We observed a significant reduction in mRNA levels of heme oxygenase 1 and mitochondrial iron genes like mitoferrin 1 and 2 in monocytes isolated from restless legs syndrome patients, indicating mitochondrial iron deficiency. Interestingly, we also observed reduced expression of iron regulatory protein 2 along with impaired activity of mitochondrial aconitase and reduced mitochondrial superoxide formation in restless legs syndrome subjects. Along this line, patients had reduced mitochondrial respiratory capacity that improved in restless legs syndrome subjects under treatment with dopaminergic drugs compared with untreated patients. CONCLUSIONS: Our data suggest that restless legs syndrome is linked to mitochondrial iron deficiency and associated impairment of mitochondrial function. This is partly corrected by treatment with dopaminergic drugs compared with untreated patients, which may be linked to an effect of dopamine on cellular iron homeostasis. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Dopamine Agents/therapeutic use , Dopamine Agonists/therapeutic use , Homeostasis/drug effects , Mitochondria/drug effects , Restless Legs Syndrome/drug therapy , Anemia, Iron-Deficiency/drug therapy , Female , Humans , Male , Mitochondria/metabolism , Quality of Life
11.
BMC Musculoskelet Disord ; 19(1): 388, 2018 Oct 30.
Article in English | MEDLINE | ID: mdl-30376863

ABSTRACT

BACKGROUND: Myofascial trigger points (MTrPs) are hyperirritable areas in the fascia of the affected muscle, possibly related to mitochondrial impairment. They can result in pain and hypoxic areas within the muscle. This pilot study established a minimally invasive biopsy technique to obtain high-quality MTrP tissue samples to evaluate mitochondrial function via high-resolution respirometry. Secondary objectives included the feasibility and safety of the biopsy procedure. METHODS: Twenty healthy males participated in this study, 10 with a diagnosis of myofascial pain in the musculus (m.) trapezius MTrP (TTP group) and 10 with a diagnosis of myofascial pain in the m. gluteus medius (GTP group). Each participant had 2 muscle biopsies taken in one session. The affected muscle was biopsied followed by a biopsy from the m. vastus lateralis to be used as a control. Measurements of oxygen consumption were carried out using high-resolution respirometry. RESULTS: Mitochondrial respiration was highest in the GTP group compared to the TTP group and the control muscle whereas no differences were observed between the GTP and the control muscle. When normalizing respiration to an internal reference state, there were no differences between muscle groups. None of the participants had hematomas or reported surgical complications. Patient-reported pain was minimal for all 3 groups. All participants reported a low procedural burden. CONCLUSIONS: This pilot study used a safe and minimally invasive technique for obtaining biopsies from MTrPs suitable for high-resolution respirometry analysis of mitochondrial function. The results suggest that there are no qualitative differences in mitochondrial function of MTrPs of the trapezius and gluteus medius muscles compared to the vastus lateralis control muscle, implying that alterations of mitochondrial function do not appear to have a role in the development of MTrPs. TRIAL REGISTRATION: Registered as No. 20131128-850 at the Coordinating Center for Clinical Studies of the Medical University of Innsbruck, trial registration date: 28th November 2013 and retrospectively registered on 11th of October 2018 at ClinicalTrials.gov with the ID NCT03704311 .


Subject(s)
Mitochondria/physiology , Myofascial Pain Syndromes/diagnosis , Myofascial Pain Syndromes/metabolism , Oxygen Consumption/physiology , Superficial Back Muscles/metabolism , Superficial Back Muscles/pathology , Adult , Biopsy, Needle/methods , Buttocks , Cohort Studies , Humans , Male , Middle Aged , Pilot Projects , Prospective Studies , Young Adult
12.
Int J Mol Sci ; 19(7)2018 Jul 21.
Article in English | MEDLINE | ID: mdl-30037119

ABSTRACT

The idea of using metabolic aberrations as targets for diagnosis or therapeutic intervention has recently gained increasing interest. In a previous study, our group discovered intriguing differences in the oxidative mitochondrial respiration capacity of benign and prostate cancer (PCa) cells. In particular, we found that PCa cells had a higher total respiratory activity than benign cells. Moreover, PCa cells showed a substantial shift towards succinate-supported mitochondrial respiration compared to benign cells, indicating a re-programming of respiratory control. This study aimed to investigate the role of succinate and its main plasma membrane transporter NaDC3 (sodium-dependent dicarboxylate transporter member 3) in PCa cells and to determine whether targeting succinate metabolism can be potentially used to inhibit PCa cell growth. Using high-resolution respirometry analysis, we observed that ROUTINE respiration in viable cells and succinate-supported respiration in permeabilized cells was higher in cells lacking the tumor suppressor phosphatase and tensin-homolog deleted on chromosome 10 (PTEN), which is frequently lost in PCa. In addition, loss of PTEN was associated with increased intracellular succinate accumulation and higher expression of NaDC3. However, siRNA-mediated knockdown of NaDC3 only moderately influenced succinate metabolism and did not affect PCa cell growth. By contrast, mersalyl acid-a broad acting inhibitor of dicarboxylic acid carriers-strongly interfered with intracellular succinate levels and resulted in reduced numbers of PCa cells. These findings suggest that blocking NaDC3 alone is insufficient to intervene with altered succinate metabolism associated with PCa. In conclusion, our data provide evidence that loss of PTEN is associated with increased succinate accumulation and enhanced succinate-supported respiration, which cannot be overcome by inhibiting the succinate transporter NaDC3 alone.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Succinic Acid/metabolism , Cell Line, Tumor , Humans , Male , Oxidative Phosphorylation , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Respiration
13.
J Sports Sci Med ; 17(3): 339-347, 2018 09.
Article in English | MEDLINE | ID: mdl-30116106

ABSTRACT

This study aimed to investigate and compare the effects of repeated-sprint (RSH) and sprint interval training in hypoxia (SIH) on sea level running and cycling performance, and to elucidate potential common or divergent adaptations of muscle perfusion and -oxygenation as well as mitochondrial respiration of blood cells. Eleven team-sport athletes performed either RSH (3x5x10s, 20s and 5min recovery between repetitions and sets) or SIH (4x30s, 5min recovery) cycling training for 3weeks (3 times/week) at a simulated altitude of 2,200m. Before and three days after the training period, a Wingate and a repeated cycling sprint test (5x6s, 20s recovery) were performed with a 30min resting period between the tests. Four to five days after the training, participants performed a repeated running sprint test (RSA, 6x17m back and forth, 20s recovery) and a Yo-Yo intermittent recovery test (YYIR2) with 1 hour active recovery between tests. The order of the tests as well as the duration of the resting periods remained the same before and after the training period. During the cycling tests near-infrared spectroscopy was performed on the vastus lateralis. In four participants, mitochondrial respiration of peripheral blood mononuclear cells (PBMC) and platelets was measured before and after training. YYIR2 running distance increased by +96.7 ± 145.6 m after RSH and by +100.0 ± 51.6 m after SIH (p = 0.034, eta² = 0.449). RSA mean running time improved by -0.138 ± 0.14s and -0.107 ± 0.08s after RSH and SIH respectively (p = 0.012, eta² = 0.564). RSH compared to SIH improved re-oxygenation during repeated sprinting. Improvements in repeated cycling were associated with improvements in re-oxygenation (r = 0.707, p <0.05). Mitochondrial electron transfer capacity normalized per PBMC count was decreased in RSH only. This study showed that cycling RSH and SIH training improves sea-level running performance. Our preliminary results suggest that RSH and SIH training results in different patterns of muscular oxygen extraction and PBMC mitochondrial respiration, without effect on platelets respiration.


Subject(s)
Athletic Performance/physiology , High-Intensity Interval Training , Hypoxia , Mitochondria/physiology , Oxygen Consumption , Adult , Altitude , Bicycling/physiology , Electron Transport , Humans , Leukocytes, Mononuclear , Pilot Projects , Running/physiology , Time Factors , Young Adult
15.
Basic Res Cardiol ; 111(6): 69, 2016 11.
Article in English | MEDLINE | ID: mdl-27743118

ABSTRACT

In this meeting report, particularly addressing the topic of protection of the cardiovascular system from ischemia/reperfusion injury, highlights are presented that relate to conditioning strategies of the heart with respect to molecular mechanisms and outcome in patients' cohorts, the influence of co-morbidities and medications, as well as the contribution of innate immune reactions in cardioprotection. Moreover, developmental or systems biology approaches bear great potential in systematically uncovering unexpected components involved in ischemia-reperfusion injury or heart regeneration. Based on the characterization of particular platelet integrins, mitochondrial redox-linked proteins, or lipid-diol compounds in cardiovascular diseases, their targeting by newly developed theranostics and technologies opens new avenues for diagnosis and therapy of myocardial infarction to improve the patients' outcome.


Subject(s)
Cardiology/trends , Cardiovascular Diseases , Theranostic Nanomedicine/trends , Animals , Cardiology/methods , Humans
16.
Anal Biochem ; 446: 64-8, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24161612

ABSTRACT

The number of studies on mitochondrial function is growing as a result of the recognition of the pivotal role of an intact mitochondrial function in numerous diseases. Measurements of oxygen consumption by the mitochondria in human skeletal muscle are used in many studies. There are several advantages of studying mitochondrial respiration in permeabilized fibers (Pfi), but the method requires a manual procedure of mechanical separation of the fiber bundles in the biopsy and chemical permeabilization of the cell membrane. This is time-consuming and subject to interpersonal variability. An alternative is to use a semiautomatic tool for preparation of a homogenate of the muscle biopsy. We investigated whether the PBI shredder is useful in preparing a muscle homogenate for measurements of mitochondrial respiratory capacity. The homogenate is compared with the Pfi preparation. Maximal respiratory capacity was significantly reduced in the homogenate compared with the Pfi from human skeletal muscle. A marked cytochrome c response was observed in the homogenate, which was not the case with the Pfi, indicating that the outer mitochondrial membrane was not intact. The mitochondria in the homogenate were more uncoupled compared with the Pfi. Manual permeabilization is an advantageous technique for preparing human skeletal muscle biopsies for respirometry.


Subject(s)
Cytological Techniques/methods , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Animals , Cell Respiration , Female , Humans , Male , Mice , Middle Aged , Mitochondria, Muscle/metabolism , Permeability
17.
J Exp Biol ; 217(Pt 16): 2947-55, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24902742

ABSTRACT

Northern elephant seals (Mirounga angustirostris) are extreme, hypoxia-adapted endotherms that rely largely on aerobic metabolism during extended breath-hold dives in near-freezing water temperatures. While many aspects of their physiology have been characterized to account for these remarkable feats, the contribution of adaptations in the aerobic powerhouses of muscle cells, the mitochondria, are unknown. In the present study, the ontogeny and comparative physiology of elephant seal muscle mitochondrial respiratory function was investigated under a variety of substrate conditions and respiratory states. Intact mitochondrial networks were studied by high-resolution respirometry in saponin-permeabilized fiber bundles obtained from primary swimming muscles of pup, juvenile and adult seals, and compared with fibers from adult human vastus lateralis. Results indicate that seal muscle maintains a high capacity for fatty acid oxidation despite a progressive decrease in total respiratory capacity as animals mature from pups to adults. This is explained by a progressive increase in phosphorylation control and fatty acid utilization over pyruvate in adult seals compared with humans and seal pups. Interestingly, despite higher indices of oxidative phosphorylation efficiency, juvenile and adult seals also exhibit a ~50% greater capacity for respiratory 'leak' compared with humans and seal pups. The ontogeny of this phenotype suggests it is an adaptation of muscle to the prolonged breath-hold exercise and highly variable ambient temperatures experienced by mature elephant seals. These studies highlight the remarkable plasticity of mammalian mitochondria to meet the demands for both efficient ATP production and endothermy in a cold, oxygen-limited environment.


Subject(s)
Diving , Mitochondria, Muscle/physiology , Muscle, Skeletal/physiology , Seals, Earless/physiology , Adaptation, Physiological , Adult , Animals , Cell Respiration , Fatty Acids/metabolism , Humans , Male , Oxidation-Reduction , Phosphorylation , Young Adult
18.
Mol Metab ; 86: 101966, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38876266

ABSTRACT

BACKGROUND: Bioenergetic remodeling of core energy metabolism is essential to the initiation, survival, and progression of cancer cells through exergonic supply of adenosine triphosphate (ATP) and metabolic intermediates, as well as control of redox homeostasis. Mitochondria are evolutionarily conserved organelles that mediate cell survival by conferring energetic plasticity and adaptive potential. Mitochondrial ATP synthesis is coupled to the oxidation of a variety of substrates generated through diverse metabolic pathways. As such, inhibition of the mitochondrial bioenergetic system by restricting metabolite availability, direct inhibition of the respiratory Complexes, altering organelle structure, or coupling efficiency may restrict carcinogenic potential and cancer progression. SCOPE OF REVIEW: Here, we review the role of bioenergetics as the principal conductor of energetic functions and carcinogenesis while highlighting the therapeutic potential of targeting mitochondrial functions. MAJOR CONCLUSIONS: Mitochondrial bioenergetics significantly contribute to cancer initiation and survival. As a result, therapies designed to limit oxidative efficiency may reduce tumor burden and enhance the efficacy of currently available antineoplastic agents.

19.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167033, 2024 03.
Article in English | MEDLINE | ID: mdl-38280294

ABSTRACT

Mitochondrial disorders are hallmarked by the dysfunction of oxidative phosphorylation (OXPHOS) yet are highly heterogeneous at the clinical and genetic levels. Striking tissue-specific pathological manifestations are a poorly understood feature of these conditions, even if the disease-causing genes are ubiquitously expressed. To investigate the functional basis of this phenomenon, we analyzed several OXPHOS-related bioenergetic parameters, including oxygen consumption rates, electron transfer system (ETS)-related coenzyme Q (mtCoQ) redox state and production of reactive oxygen species (ROS) in mouse brain and liver mitochondria fueled by different substrates. In addition, we determined how these functional parameters are affected by ETS impairment in a tissue-specific manner using pathologically relevant mouse models lacking either Ndufs4 or Ttc19, leading to Complex I (CI) or Complex III (CIII) deficiency, respectively. Detailed OXPHOS analysis revealed striking differences between brain and liver mitochondria in the capacity of the different metabolic substrates to fuel the ETS, reduce the ETS-related mtCoQ, and to induce ROS production. In addition, ETS deficiency due to either CI or CIII dysfunction had a much greater impact on the intrinsic bioenergetic parameters of brain compared with liver mitochondria. These findings are discussed in terms of the still rather mysterious tissue-specific manifestations of mitochondrial disease.


Subject(s)
Mitochondria, Liver , Mitochondrial Diseases , Animals , Mice , Mitochondria, Liver/metabolism , Reactive Oxygen Species/metabolism , Energy Metabolism , Brain/metabolism , Mitochondrial Diseases/metabolism , Electron Transport Complex I/metabolism
20.
Redox Biol ; 71: 103037, 2024 May.
Article in English | MEDLINE | ID: mdl-38401291

ABSTRACT

Mitochondrial respiration extends beyond ATP generation, with the organelle participating in many cellular and physiological processes. Parallel changes in components of the mitochondrial electron transfer system with respiration render it an appropriate hub for coordinating cellular adaption to changes in oxygen levels. How changes in respiration under functional hypoxia (i.e., when intracellular O2 levels limit mitochondrial respiration) are relayed by the electron transfer system to impact mitochondrial adaption and remodeling after hypoxic exposure remains poorly defined. This is largely due to challenges integrating findings under controlled and defined O2 levels in studies connecting functions of isolated mitochondria to humans during physical exercise. Here we present experiments under conditions of hypoxia in isolated mitochondria, myotubes and exercising humans. Performing steady-state respirometry with isolated mitochondria we found that oxygen limitation of respiration reduced electron flow and oxidative phosphorylation, lowered the mitochondrial membrane potential difference, and decreased mitochondrial calcium influx. Similarly, in myotubes under functional hypoxia mitochondrial calcium uptake decreased in response to sarcoplasmic reticulum calcium release for contraction. In both myotubes and human skeletal muscle this blunted mitochondrial adaptive responses and remodeling upon contractions. Our results suggest that by regulating calcium uptake the mitochondrial electron transfer system is a hub for coordinating cellular adaption under functional hypoxia.


Subject(s)
Calcium , Oxygen Consumption , Humans , Calcium/metabolism , Oxygen Consumption/physiology , Cell Respiration , Hypoxia/metabolism , Muscle, Skeletal/metabolism , Oxygen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL