1
|
Sharma S, Shamjetsabam ND, Chauhan K, Yashavarddhan M, Gautam P, Prakash P, Choudhary P, Chhabra SS, Acharya R, Kalra SK, Gupta A, Jain S, Ganguly NK, Rana R. Quantitative tissue analysis reveals AK2, COL1A1, and PLG protein signatures: targeted therapeutics for meningioma. Int J Surg 2024; 110:7434-7446. [PMID: 39288025 PMCID: PMC11634153 DOI: 10.1097/js9.0000000000002054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Meningioma is the most prevalent primary intracranial brain tumor and accounts for one-third of all CNS tumors. Meningioma is known to be the most common yet life-threatening brain tumor with a higher recurrence rate. Globally, there is an increase in the healthcare burden due to meningioma and hence in its research. The present clinical approach includes surgical resection, chemotherapy, and radiation therapies to which the malignancy does not seem to respond efficiently. Targeted therapies and molecular markers provide elite patient treatment and care for individuals suffering from meningiomas as compared to conventional measures. Although there is proteomic data on meningioma the knowledge of potential biomarkers differentiating the grades is scarce. To identify the best set of biomarkers, validation of reported markers in large and independent sample cohorts in the future is necessary. METHODS A total of 12 samples, 3 each of control (which made pool 1) meningioma grade I (which made 2 sets: pool 2 and pool 3), and meningioma grade II (which made pool 4) were taken for LC-MS/MS. After this, the expression of three proteins was checked by immunocytochemistry, flow cytometry, and western blotting. RESULTS Protein expression was analyzed using various techniques like mass spectrometry, immunocytochemistry, flow cytometry, and western blotting. Mass spectrometry is the most commonly used standard and reliable technique for identifying and quantifying protein expression. We got three highly upregulated proteins namely AK2, COL1A1, and PLG using this technique. The biomarker potential of these proteins was further checked by ICC, western blotting, and flow cytometry. Three important proteins were found to be upregulated namely, AK2 (Adenylate kinase 2), COL1A1 (collagen 1A1), and PLG (plasminogen). The order of increased protein expression was control < MG grade I < MG grade II according to mass spectrometry and western blotting. In immunocytochemistry, we found that COL1A1 expression increases significantly with grades in comparison to control. Similarly, AK2 and PLG also showed little increase but not as much as COL1A1. In flow cytometry, PLG showed higher upregulation in grades than control. While AK2 and COL1A1 showed little increase in expression in grades than control. All techniques, especially mass spectrometry and western blotting, presented higher expression of these proteins in grades as compared to control. CONCLUSIONS In the quest to find a suitable therapeutic marker, this study incorporates quantitative labeling and detection followed by flow cytometry, immunocytochemistry, and western blotting for early diagnosis and treatment of meningioma. The article further explores the efficacy of some proteins namely AK2, COL1A1, and PLG to be the targeted molecules.
Collapse
Affiliation(s)
- Swati Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital
| | | | - Kirti Chauhan
- Department of Biotechnology and Research, Sir Ganga Ram Hospital
| | | | - Poonam Gautam
- Central Proteomics Facility, ICMR-National Institute of Pathology
| | - Prem Prakash
- Department of Molecular Medicine, Jamia Hamdard Deemed to be University
| | - Priyanka Choudhary
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Bathinda, Punjab, India
| | | | - Rajesh Acharya
- Department of Neurosurgery, Sir Ganga Ram Hospital, New Delhi
| | - Samir K. Kalra
- Department of Neurosurgery, Sir Ganga Ram Hospital, New Delhi
| | - Anshul Gupta
- Department of Neurosurgery, Sir Ganga Ram Hospital, New Delhi
| | - Sunila Jain
- Department of Histopathology, Sir Ganga Ram Hospital
| | | | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital
| |
Collapse
|
2
|
Watson WD, Arvidsson PM, Miller JJJ, Lewis AJ, Rider OJ. A Mitochondrial Basis for Heart Failure Progression. Cardiovasc Drugs Ther 2024; 38:1161-1171. [PMID: 38878138 PMCID: PMC11680631 DOI: 10.1007/s10557-024-07582-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 12/29/2024]
Abstract
In health, the human heart is able to match ATP supply and demand perfectly. It requires 6 kg of ATP per day to satisfy demands of external work (mechanical force generation) and internal work (ion movements and basal metabolism). The heart is able to link supply with demand via direct responses to ADP and AMP concentrations but calcium concentrations within myocytes play a key role, signalling both inotropy, chronotropy and matched increases in ATP production. Calcium/calmodulin-dependent protein kinase (CaMKII) is a key adapter to increased workload, facilitating a greater and more rapid calcium concentration change. In the failing heart, this is dysfunctional and ATP supply is impaired. This review aims to examine the mechanisms and pathologies that link increased energy demand to this disrupted situation. We examine the roles of calcium loading, oxidative stress, mitochondrial structural abnormalities and damage-associated molecular patterns.
Collapse
Affiliation(s)
- William D Watson
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK.
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK.
| | - Per M Arvidsson
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Clinical Physiology, Skåne University Hospital, Lund, Sweden
| | - Jack J J Miller
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Andrew J Lewis
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Galenkamp NS, Zernia S, Van Oppen YB, van den Noort M, Milias-Argeitis A, Maglia G. Allostery can convert binding free energies into concerted domain motions in enzymes. Nat Commun 2024; 15:10109. [PMID: 39572546 PMCID: PMC11582565 DOI: 10.1038/s41467-024-54421-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
Enzymatic mechanisms are typically inferred from structural data. However, understanding enzymes require unravelling the intricate dynamic interplay between dynamics, conformational substates, and multiple protein structures. Here, we use single-molecule nanopore analysis to investigate the catalytic conformational changes of adenylate kinase (AK), an enzyme that catalyzes the interconversion of various adenosine phosphates (ATP, ADP, and AMP). Kinetic analysis validated by hidden Markov models unravels the details of domain motions during catalysis. Our findings reveal that allosteric interactions between ligands and cofactor enable converting binding energies into directional conformational changes of the two catalytic domains of AK. These coordinated motions emerged to control the exact sequence of ligand binding and the affinity for the three different substrates, thereby guiding the reactants along the reaction coordinates. Interestingly, we find that about 10% of enzymes show altered allosteric regulation and ligand affinities, indicating that a subset of enzymes folds in alternative catalytically active forms. Since molecules or proteins might be able to selectively stabilize one of the folds, this observation suggests an evolutionary path for allostery in enzymes. In AK, this complex catalytic framework has likely emerged to prevent futile ATP/ADP hydrolysis and to regulate the enzyme for different energy needs of the cell.
Collapse
Affiliation(s)
- Nicole Stéphanie Galenkamp
- Chemical Biology I, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Sarah Zernia
- Chemical Biology I, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Yulan B Van Oppen
- Molecular Systems Biology, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Marco van den Noort
- Chemical Biology I, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Andreas Milias-Argeitis
- Molecular Systems Biology, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Giovanni Maglia
- Chemical Biology I, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
4
|
Paraskevaidis I, Kourek C, Farmakis D, Tsougos E. Heart Failure: A Deficiency of Energy-A Path Yet to Discover and Walk. Biomedicines 2024; 12:2589. [PMID: 39595155 PMCID: PMC11592498 DOI: 10.3390/biomedicines12112589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Heart failure is a complex syndrome and our understanding and therapeutic approach relies mostly on its phenotypic presentation. Notably, the heart is characterized as the most energy-consuming organ, being both a producer and consumer, in order to satisfy multiple cardiac functions: ion exchange, electromechanical coordination, excitation-contraction coupling, etc. By obtaining further knowledge of the cardiac energy field, we can probably better characterize the basic pathophysiological events occurring in heart disease patients and understand the metabolic substance changes, the relationship between the alteration of energy production/consumption, and hence energetic deficiency not only in the heart as a whole but in every single cardiac territory, which will hopefully provide us with the opportunity to uncover the beginning of the heart failure process. In this respect, using (a) newer imaging techniques, (b) biomedicine, (c) nanotechnology, and (d) artificial intelligence, we can gain a deeper understanding of this complex syndrome. This, in turn, can lead to earlier and more effective therapeutic approaches, ultimately improving human health. To date, the scientific community has not given sufficient attention to the energetic starvation model. In our view, this review aims to encourage scientists and the medical community to conduct studies for a better understanding and treatment of this syndrome.
Collapse
Affiliation(s)
- Ioannis Paraskevaidis
- 6th Department of Cardiology, Hygeia Hospital, 151 23 Athens, Greece; (I.P.); (E.T.)
| | - Christos Kourek
- Department of Cardiology, 417 Army Share Fund Hospital of Athens (NIMTS), 115 21 Athens, Greece;
| | - Dimitrios Farmakis
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistiran University of Athens, 124 62 Athens, Greece
| | - Elias Tsougos
- 6th Department of Cardiology, Hygeia Hospital, 151 23 Athens, Greece; (I.P.); (E.T.)
| |
Collapse
|
5
|
Takeuchi A, Matsuoka S. A simulation study on the role of mitochondria-sarcoplasmic reticulum Ca 2+ interaction in cardiomyocyte energetics during exercise. J Physiol 2024. [PMID: 39387569 DOI: 10.1113/jp286054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/15/2024] [Indexed: 10/15/2024] Open
Abstract
Previous studies demonstrated that the mitochondrial Ca2+ uniporter MCU and the Na+-Ca2+ exchanger NCLX exist in proximity to the sarcoplasmic reticulum (SR) ryanodine receptor RyR and the Ca2+ pump SERCA, respectively, creating a mitochondria-SR Ca2+ interaction. However, the physiological relevance of the mitochondria-SR Ca2+ interaction has remained unsolved. Furthermore, although mitochondrial Ca2+ has been proposed to be an important factor regulating mitochondrial energy metabolism, by activating NADH-producing dehydrogenases, the contribution of the Ca2+-dependent regulatory mechanisms to cellular functions under physiological conditions has been controversial. In this study, we constructed a new integrated model of human ventricular myocyte with excitation-contraction-energetics coupling and investigated systematically the contribution of mitochondria-SR Ca2+ interaction, especially focusing on cardiac energetics during dynamic workload transitions in exercise. Simulation analyses revealed that the spatial coupling of mitochondria and SR, particularly via mitochondrial Ca2+ uniport activity-RyR, was the primary determinant of mitochondrial Ca2+ concentration, and that the Ca2+-dependent regulatory mechanism facilitated mitochondrial NADH recovery during exercise and contributed to the stability of NADH in the workload transition by about 40%, while oxygen consumption rate and cytoplasmic ATP level were not influenced. We concluded that the mitochondria-SR Ca2+ interaction, created via the uneven distribution of Ca2+ handling proteins, optimizes the contribution of the mitochondrial Ca2+-dependent regulatory mechanism to stabilizing NADH during exercise. KEY POINTS: The mitochondrial Ca2+ uniporter protein MCU and the Na+-Ca2+ exchanger protein NCLX are reported to exist in proximity to the sarcoplasmic reticulum (SR) ryanodine receptor RyR and the Ca2+ pump SERCA, respectively, creating a mitochondria-SR Ca2+ interaction in cardiomyocytes. Mitochondrial Ca2+ (Ca2+ mit) has been proposed to be an important factor regulating mitochondrial energy metabolism, by activating NADH-producing dehydrogenases. Here we constructed an integrated model of a human ventricular myocyte with excitation-contraction-energetics coupling and investigated the role of the mitochondria-SR Ca2+ interaction in cardiac energetics during exercise. Simulation analyses revealed that the spatial coupling particularly via mitochondrial Ca2+ uniport activity-RyR is the primary determinant of Ca2+ mit concentration, and that the activation of NADH-producing dehydrogenases by Ca2+ mit contributes to NADH stability during exercise. The mitochondria-SR Ca2+ interaction optimizes the contribution of Ca2+ mit to the activation of NADH-producing dehydrogenases.
Collapse
Affiliation(s)
- Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences and Life Science Innovation Center, University of Fukui, Fukui, Japan
| | - Satoshi Matsuoka
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences and Life Science Innovation Center, University of Fukui, Fukui, Japan
| |
Collapse
|
6
|
Pichot V, Corbier C, Chouchou F. The contribution of granger causality analysis to our understanding of cardiovascular homeostasis: from cardiovascular and respiratory interactions to central autonomic network control. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1315316. [PMID: 39175608 PMCID: PMC11338816 DOI: 10.3389/fnetp.2024.1315316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 07/18/2024] [Indexed: 08/24/2024]
Abstract
Homeostatic regulation plays a fundamental role in maintenance of multicellular life. At different scales and in different biological systems, this principle allows a better understanding of biological organization. Consequently, a growing interest in studying cause-effect relations between physiological systems has emerged, such as in the fields of cardiovascular and cardiorespiratory regulations. For this, mathematical approaches such as Granger causality (GC) were applied to the field of cardiovascular physiology in the last 20 years, overcoming the limitations of previous approaches and offering new perspectives in understanding cardiac, vascular and respiratory homeostatic interactions. In clinical practice, continuous recording of clinical data of hospitalized patients or by telemetry has opened new applicability for these approaches with potential early diagnostic and prognostic information. In this review, we describe a theoretical background of approaches based on linear GC in time and frequency domains applied to detect couplings between time series of RR intervals, blood pressure and respiration. Interestingly, these tools help in understanding the contribution of homeostatic negative feedback and the anticipatory feedforward mechanisms in homeostatic cardiovascular and cardiorespiratory controls. We also describe experimental and clinical results based on these mathematical tools, consolidating previous experimental and clinical evidence on the coupling in cardiovascular and cardiorespiratory studies. Finally, we propose perspectives allowing to complete the understanding of these interactions between cardiovascular and cardiorespiratory systems, as well as the interplay between brain and cardiac, and vascular and respiratory systems, offering a high integrative view of cardiovascular and cardiorespiratory homeostatic regulation.
Collapse
Affiliation(s)
- Vincent Pichot
- Department of Clinical and Exercise Physiology, SAINBIOSE, Inserm U1059, Saint-Etienne Jean Monnet University, CHU Saint-Etienne, Saint-Etienne, France
| | - Christophe Corbier
- LASPI EA3059, Saint-Etienne Jean Monnet University, Roanne Technology University Institute, Roanne, France
| | - Florian Chouchou
- IRISSE Laboratory EA4075, University of La Réunion, UFR Science de ’Homme et de l’Environnement, Le Tampon, France
| |
Collapse
|
7
|
Gronda E, Palazzuoli A, Iacoviello M, Benevenuto M, Gabrielli D, Arduini A. Renal Oxygen Demand and Nephron Function: Is Glucose a Friend or Foe? Int J Mol Sci 2023; 24:9957. [PMID: 37373108 DOI: 10.3390/ijms24129957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The kidneys and heart work together to balance the body's circulation, and although their physiology is based on strict inter dependence, their performance fulfills different aims. While the heart can rapidly increase its own oxygen consumption to comply with the wide changes in metabolic demand linked to body function, the kidneys physiology are primarily designed to maintain a stable metabolic rate and have a limited capacity to cope with any steep increase in renal metabolism. In the kidneys, glomerular population filters a large amount of blood and the tubular system has been programmed to reabsorb 99% of filtrate by reabsorbing sodium together with other filtered substances, including all glucose molecules. Glucose reabsorption involves the sodium-glucose cotransporters SGLT2 and SGLT1 on the apical membrane in the proximal tubular section; it also enhances bicarbonate formation so as to preserve the acid-base balance. The complex work of reabsorption in the kidney is the main factor in renal oxygen consumption; analysis of the renal glucose transport in disease states provides a better understanding of the renal physiology changes that occur when clinical conditions alter the neurohormonal response leading to an increase in glomerular filtration pressure. In this circumstance, glomerular hyperfiltration occurs, imposing a higher metabolic demand on kidney physiology and causing progressive renal impairment. Albumin urination is the warning signal of renal engagement over exertion and most frequently heralds heart failure development, regardless of disease etiology. The review analyzes the mechanisms linked to renal oxygen consumption, focusing on sodium-glucose management.
Collapse
Affiliation(s)
- Edoardo Gronda
- Medicine and Medicine Sub-Specialties Department, Cardio Renal Program, U.O.C. Nephrology, Dialysis and Adult Renal Transplant Program, IRCCS Ca' Granda Foundation, Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, S. Maria alle Scotte Hospital University of Siena, 53100 Siena, Italy
| | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy
| | - Manuela Benevenuto
- Unità Operativa Complessa Cardiologia-UTIC-Emodinamica, PO Giuseppe Mazzini, 64100 Teramo, Italy
| | - Domenico Gabrielli
- Unità Operativa Complessa Cardiologia-UTIC, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | | |
Collapse
|
8
|
[Mechano-energetic defects in heart failure]. Herz 2023; 48:123-133. [PMID: 36700949 DOI: 10.1007/s00059-022-05161-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/27/2023]
Abstract
Heart failure is characterized by defects in excitation-contraction coupling, energetic deficit and oxidative stress. The energy for cardiac contraction and relaxation is provided in mitochondria, whose function is tightly regulated by excitation-contraction coupling in cardiac myocytes. In heart failure with reduced ejection fraction (HFrEF), alterations in the ion balance in cardiac myocytes impair mitochondrial Ca2+ uptake, which is required for activation of the Krebs cycle, causing an energetic deficit and oxidative stress in mitochondria. Recent clinical studies suggest that in heart failure with preserved ejection fraction (HFpEF), in stark contrast to HFrEF, hypercontractility often occurs as an attempt to compensate for a pathological increase in systemic and pulmonary vascular resistance. This hypercontractility increases cardiac energy and oxygen demands at rest and reduces the contractile, diastolic and coronary reserves, preventing an adequate increase in cardiac output during exercise. Moreover, increased contractility causes long-term maladaptive remodeling processes due to oxidative stress and redox-sensitive prohypertrophic signaling pathways. As overweight and diabetes, particularly in the interplay with hemodynamic stress, are important risk factors for the development of HFpEF, interventions targeting metabolism in particular could ameliorate the development and progression of HFpEF.
Collapse
|
9
|
Balakina-Vikulova NA, Katsnelson LB. Integrative Mathematical Model of Electrical, Metabolic and Mechanical Processes in Human Cardiomyocytes. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022070122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
10
|
Germanova E, Khmil N, Pavlik L, Mikheeva I, Mironova G, Lukyanova L. The Role of Mitochondrial Enzymes, Succinate-Coupled Signaling Pathways and Mitochondrial Ultrastructure in the Formation of Urgent Adaptation to Acute Hypoxia in the Myocardium. Int J Mol Sci 2022; 23:ijms232214248. [PMID: 36430733 PMCID: PMC9696391 DOI: 10.3390/ijms232214248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
The effect of a single one-hour exposure to three modes of hypobaric hypoxia (HBH) differed in the content of O2 in inhaled air (FiO2-14%, 10%, 8%) in the development of mitochondrial-dependent adaptive processes in the myocardium was studied in vivo. The following parameters have been examined: (a) an urgent reaction of catalytic subunits of mitochondrial enzymes (NDUFV2, SDHA, Cyt b, COX2, ATP5A) in the myocardium as an indicator of the state of the respiratory chain electron transport function; (b) an urgent activation of signaling pathways dependent on GPR91, HIF-1α and VEGF, allowing us to assess their role in the formation of urgent mechanisms of adaptation to hypoxia in the myocardium; (c) changes in the ultrastructure of three subpopulations of myocardial mitochondria under these conditions. The studies were conducted on two rat phenotypes: rats with low resistance (LR) and high resistance (HR) to hypoxia. The adaptive and compensatory role of the mitochondrial complex II (MC II) in maintaining the electron transport and energy function of the myocardium in a wide range of reduced O2 concentrations in the initial period of hypoxic exposure has been established. The features of urgent reciprocal regulatory interaction of NAD- and FAD-dependent oxidation pathways in myocardial mitochondria under these conditions have been revealed. The data indicating the participation of GPR91, HIF-1a and VEGF in this process have been obtained. The ultrastructure of the mitochondrial subpopulations in the myocardium of LR and HR rats differed in normoxic conditions and reacted differently to hypoxia of varying severity. The parameters studied together are highly informative indicators of the quality of cardiac activity and metabolic biomarkers of urgent adaptation in various hypoxic conditions.
Collapse
Affiliation(s)
- Elita Germanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
| | - Natalya Khmil
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Lyubov Pavlik
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Irina Mikheeva
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Galina Mironova
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
- Correspondence: (G.M.); (L.L.)
| | - Ludmila Lukyanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
- Correspondence: (G.M.); (L.L.)
| |
Collapse
|
11
|
Santiago A, Butakoff C, Eguzkitza B, Gray RA, May-Newman K, Pathmanathan P, Vu V, Vázquez M. Design and execution of a verification, validation, and uncertainty quantification plan for a numerical model of left ventricular flow after LVAD implantation. PLoS Comput Biol 2022; 18:e1010141. [PMID: 35696442 PMCID: PMC9232142 DOI: 10.1371/journal.pcbi.1010141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/24/2022] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Background
Left ventricular assist devices (LVADs) are implantable pumps that act as a life support therapy for patients with severe heart failure. Despite improving the survival rate, LVAD therapy can carry major complications. Particularly, the flow distortion introduced by the LVAD in the left ventricle (LV) may induce thrombus formation. While previous works have used numerical models to study the impact of multiple variables in the intra-LV stagnation regions, a comprehensive validation analysis has never been executed. The main goal of this work is to present a model of the LV-LVAD system and to design and follow a verification, validation and uncertainty quantification (VVUQ) plan based on the ASME V&V40 and V&V20 standards to ensure credible predictions.
Methods
The experiment used to validate the simulation is the SDSU cardiac simulator, a bench mock-up of the cardiovascular system that allows mimicking multiple operation conditions for the heart-LVAD system. The numerical model is based on Alya, the BSC’s in-house platform for numerical modelling. Alya solves the Navier-Stokes equation with an Arbitrary Lagrangian-Eulerian (ALE) formulation in a deformable ventricle and includes pressure-driven valves, a 0D Windkessel model for the arterial output and a LVAD boundary condition modeled through a dynamic pressure-flow performance curve. The designed VVUQ plan involves: (a) a risk analysis and the associated credibility goals; (b) a verification stage to ensure correctness in the numerical solution procedure; (c) a sensitivity analysis to quantify the impact of the inputs on the four quantities of interest (QoIs) (average aortic root flow Q A o a v g, maximum aortic root flow Q A o m a x, average LVAD flow Q V A D a v g, and maximum LVAD flow Q V A D m a x); (d) an uncertainty quantification using six validation experiments that include extreme operating conditions.
Results
Numerical code verification tests ensured correctness of the solution procedure and numerical calculation verification showed a grid convergence index (GCI)95% <3.3%. The total Sobol indices obtained during the sensitivity analysis demonstrated that the ejection fraction, the heart rate, and the pump performance curve coefficients are the most impactful inputs for the analysed QoIs. The Minkowski norm is used as validation metric for the uncertainty quantification. It shows that the midpoint cases have more accurate results when compared to the extreme cases. The total computational cost of the simulations was above 100 [core-years] executed in around three weeks time span in Marenostrum IV supercomputer.
Conclusions
This work details a novel numerical model for the LV-LVAD system, that is supported by the design and execution of a VVUQ plan created following recognised international standards. We present a methodology demonstrating that stringent VVUQ according to ASME standards is feasible but computationally expensive.
Collapse
Affiliation(s)
- Alfonso Santiago
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- ELEM biotech, Barcelona, Spain
| | | | | | - Richard A. Gray
- US Food and Drug Administration (FDA), Silver Spring, Maryland, United States of America
| | - Karen May-Newman
- Department of Mechanical Engineering, San Diego State University (SDSU), San Diego, California, United States of America
| | - Pras Pathmanathan
- US Food and Drug Administration (FDA), Silver Spring, Maryland, United States of America
| | - Vi Vu
- Department of Mechanical Engineering, San Diego State University (SDSU), San Diego, California, United States of America
| | - Mariano Vázquez
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- ELEM biotech, Barcelona, Spain
- * E-mail:
| |
Collapse
|
12
|
Cadour F, Thuny F, Sourdon J. New Insights in Early Detection of Anticancer Drug-Related Cardiotoxicity Using Perfusion and Metabolic Imaging. Front Cardiovasc Med 2022; 9:813883. [PMID: 35198613 PMCID: PMC8858802 DOI: 10.3389/fcvm.2022.813883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/06/2022] [Indexed: 12/16/2022] Open
Abstract
Cardio-oncology requires a good knowledge of the cardiotoxicity of anticancer drugs, their mechanisms, and their diagnosis for better management. Anthracyclines, anti-vascular endothelial growth factor (VEGF), alkylating agents, antimetabolites, anti-human epidermal growth factor receptor (HER), and receptor tyrosine kinase inhibitors (RTKi) are therapeutics whose cardiotoxicity involves several mechanisms at the cellular and subcellular levels. Current guidelines for anticancer drugs cardiotoxicity are essentially based on monitoring left ventricle ejection fraction (LVEF). However, knowledge of microvascular and metabolic dysfunction allows for better imaging assessment before overt LVEF impairment. Early detection of anticancer drug-related cardiotoxicity would therefore advance the prevention and patient care. In this review, we provide a comprehensive overview of the cardiotoxic effects of anticancer drugs and describe myocardial perfusion, metabolic, and mitochondrial function imaging approaches to detect them before over LVEF impairment.
Collapse
Affiliation(s)
- Farah Cadour
- Aix-Marseille Université, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
| | - Franck Thuny
- Aix-Marseille University, University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), Inserm 1263, Inrae 1260, Marseille, France
| | - Joevin Sourdon
- Aix-Marseille Université, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
- *Correspondence: Joevin Sourdon
| |
Collapse
|
13
|
Zhang X, Tomar N, Kandel SM, Audi SH, Cowley AW, Dash RK. Substrate- and Calcium-Dependent Differential Regulation of Mitochondrial Oxidative Phosphorylation and Energy Production in the Heart and Kidney. Cells 2021; 11:131. [PMID: 35011693 PMCID: PMC8750792 DOI: 10.3390/cells11010131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dehydrogenases are differentially stimulated by Ca2+. Ca2+ has also diverse regulatory effects on mitochondrial transporters and other enzymes. However, the consequences of these regulatory effects on mitochondrial oxidative phosphorylation (OxPhos) and ATP production, and the dependencies of these consequences on respiratory substrates, have not been investigated between the kidney and heart despite the fact that kidney energy requirements are second only to those of the heart. Our objective was, therefore, to elucidate these relationships in isolated mitochondria from the kidney outer medulla (OM) and heart. ADP-induced mitochondrial respiration was measured at different CaCl2 concentrations in the presence of various respiratory substrates, including pyruvate + malate (PM), glutamate + malate (GM), alpha-ketoglutarate + malate (AM), palmitoyl-carnitine + malate (PCM), and succinate + rotenone (SUC + ROT). The results showed that, in both heart and OM mitochondria, and for most complex I substrates, Ca2+ effects are biphasic: small increases in Ca2+ concentration stimulated, while large increases inhibited mitochondrial respiration. Furthermore, significant differences in substrate- and Ca2+-dependent O2 utilization towards ATP production between heart and OM mitochondria were observed. With PM and PCM substrates, Ca2+ showed more prominent stimulatory effects in OM than in heart mitochondria, while with GM and AM substrates, Ca2+ had similar biphasic regulatory effects in both OM and heart mitochondria. In contrast, with complex II substrate SUC + ROT, only inhibitory effects on mitochondrial respiration was observed in both the heart and the OM. We conclude that the regulatory effects of Ca2+ on mitochondrial OxPhos and ATP synthesis are biphasic, substrate-dependent, and tissue-specific.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Sunil M. Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Said H. Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee, WI 53223, USA;
| | - Allen W. Cowley
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
14
|
Bertero E, Nickel A, Kohlhaas M, Hohl M, Sequeira V, Brune C, Schwemmlein J, Abeßer M, Schuh K, Kutschka I, Carlein C, Münker K, Atighetchi S, Müller A, Kazakov A, Kappl R, von der Malsburg K, van der Laan M, Schiuma AF, Böhm M, Laufs U, Hoth M, Rehling P, Kuhn M, Dudek J, von der Malsburg A, Prates Roma L, Maack C. Loss of Mitochondrial Ca 2+ Uniporter Limits Inotropic Reserve and Provides Trigger and Substrate for Arrhythmias in Barth Syndrome Cardiomyopathy. Circulation 2021; 144:1694-1713. [PMID: 34648376 DOI: 10.1161/circulationaha.121.053755] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Barth syndrome (BTHS) is caused by mutations of the gene encoding tafazzin, which catalyzes maturation of mitochondrial cardiolipin and often manifests with systolic dysfunction during early infancy. Beyond the first months of life, BTHS cardiomyopathy typically transitions to a phenotype of diastolic dysfunction with preserved ejection fraction, blunted contractile reserve during exercise, and arrhythmic vulnerability. Previous studies traced BTHS cardiomyopathy to mitochondrial formation of reactive oxygen species (ROS). Because mitochondrial function and ROS formation are regulated by excitation-contraction coupling, integrated analysis of mechano-energetic coupling is required to delineate the pathomechanisms of BTHS cardiomyopathy. METHODS We analyzed cardiac function and structure in a mouse model with global knockdown of tafazzin (Taz-KD) compared with wild-type littermates. Respiratory chain assembly and function, ROS emission, and Ca2+ uptake were determined in isolated mitochondria. Excitation-contraction coupling was integrated with mitochondrial redox state, ROS, and Ca2+ uptake in isolated, unloaded or preloaded cardiac myocytes, and cardiac hemodynamics analyzed in vivo. RESULTS Taz-KD mice develop heart failure with preserved ejection fraction (>50%) and age-dependent progression of diastolic dysfunction in the absence of fibrosis. Increased myofilament Ca2+ affinity and slowed cross-bridge cycling caused diastolic dysfunction, in part, compensated by accelerated diastolic Ca2+ decay through preactivated sarcoplasmic reticulum Ca2+-ATPase. Taz deficiency provoked heart-specific loss of mitochondrial Ca2+ uniporter protein that prevented Ca2+-induced activation of the Krebs cycle during β-adrenergic stimulation, oxidizing pyridine nucleotides and triggering arrhythmias in cardiac myocytes. In vivo, Taz-KD mice displayed prolonged QRS duration as a substrate for arrhythmias, and a lack of inotropic response to β-adrenergic stimulation. Cellular arrhythmias and QRS prolongation, but not the defective inotropic reserve, were restored by inhibiting Ca2+ export through the mitochondrial Na+/Ca2+ exchanger. All alterations occurred in the absence of excess mitochondrial ROS in vitro or in vivo. CONCLUSIONS Downregulation of mitochondrial Ca2+ uniporter, increased myofilament Ca2+ affinity, and preactivated sarcoplasmic reticulum Ca2+-ATPase provoke mechano-energetic uncoupling that explains diastolic dysfunction and the lack of inotropic reserve in BTHS cardiomyopathy. Furthermore, defective mitochondrial Ca2+ uptake provides a trigger and a substrate for ventricular arrhythmias. These insights can guide the ongoing search for a cure of this orphaned disease.
Collapse
Affiliation(s)
- Edoardo Bertero
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Now with Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy (E.B.)
| | - Alexander Nickel
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Michael Kohlhaas
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Mathias Hohl
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Vasco Sequeira
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Carolin Brune
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Julia Schwemmlein
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Marco Abeßer
- Institute of Physiology, University of Würzburg, Germany (M.A., K.S., M. Kuhn)
| | - Kai Schuh
- Institute of Physiology, University of Würzburg, Germany (M.A., K.S., M. Kuhn)
| | - Ilona Kutschka
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Christopher Carlein
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Kai Münker
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Sarah Atighetchi
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Andreas Müller
- Clinic for Radiology (A.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Andrey Kazakov
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Reinhard Kappl
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Karina von der Malsburg
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling, PZMS, Faculty of Medicine (K.v.d.M., M.v.d.L., A.v.d.M.), Saarland University, Homburg/Saar, Germany
| | - Martin van der Laan
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling, PZMS, Faculty of Medicine (K.v.d.M., M.v.d.L., A.v.d.M.), Saarland University, Homburg/Saar, Germany
| | - Anna-Florentine Schiuma
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.)
| | - Michael Böhm
- Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany
| | - Ulrich Laufs
- Now with Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (U.L.)
| | - Markus Hoth
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, Georg-August University, Göttingen, Germany (P.R., J.D.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (P.R.).,Max-Planck Institute for Biophysical Chemistry, Göttingen, Germany (P.R.)
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Germany (M.A., K.S., M. Kuhn)
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Department of Cellular Biochemistry, Georg-August University, Göttingen, Germany (P.R., J.D.)
| | - Alexander von der Malsburg
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling, PZMS, Faculty of Medicine (K.v.d.M., M.v.d.L., A.v.d.M.), Saarland University, Homburg/Saar, Germany
| | - Leticia Prates Roma
- Department for Biophysics, ZHMB, CIPMM (C.C., R.K., M. Hoth, L.P.R.), Saarland University, Homburg/Saar, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic, Würzburg, Germany (E.B., A.N., M. Kohlhaas, V.S., J.S., I.K., K.M., S.A., A.-F.S., J.D., C.M.).,Clinic for Internal Medicine III (M. Hohl, C.B., K.M., S.A., A.K., M.B., C.M.), Saarland University Clinic, Homburg/Saar, Germany.,Department for Internal Medicine 1, University Clinic Würzburg, Germany (C.M.)
| |
Collapse
|
15
|
Tokarska-Schlattner M, Kay L, Perret P, Isola R, Attia S, Lamarche F, Tellier C, Cottet-Rousselle C, Uneisi A, Hininger-Favier I, Foretz M, Dubouchaud H, Ghezzi C, Zuppinger C, Viollet B, Schlattner U. Role of Cardiac AMP-Activated Protein Kinase in a Non-pathological Setting: Evidence From Cardiomyocyte-Specific, Inducible AMP-Activated Protein Kinase α1α2-Knockout Mice. Front Cell Dev Biol 2021; 9:731015. [PMID: 34733845 PMCID: PMC8558539 DOI: 10.3389/fcell.2021.731015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/24/2021] [Indexed: 12/25/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a key regulator of energy homeostasis under conditions of energy stress. Though heart is one of the most energy requiring organs and depends on a perfect match of energy supply with high and fluctuating energy demand to maintain its contractile performance, the role of AMPK in this organ is still not entirely clear, in particular in a non-pathological setting. In this work, we characterized cardiomyocyte-specific, inducible AMPKα1 and α2 knockout mice (KO), where KO was induced at the age of 8 weeks, and assessed their phenotype under physiological conditions. In the heart of KO mice, both AMPKα isoforms were strongly reduced and thus deleted in a large part of cardiomyocytes already 2 weeks after tamoxifen administration, persisting during the entire study period. AMPK KO had no effect on heart function at baseline, but alterations were observed under increased workload induced by dobutamine stress, consistent with lower endurance exercise capacity observed in AMPK KO mice. AMPKα deletion also induced a decrease in basal metabolic rate (oxygen uptake, energy expenditure) together with a trend to lower locomotor activity of AMPK KO mice 12 months after tamoxifen administration. Loss of AMPK resulted in multiple alterations of cardiac mitochondria: reduced respiration with complex I substrates as measured in isolated mitochondria, reduced activity of complexes I and IV, and a shift in mitochondrial cristae morphology from lamellar to mixed lamellar-tubular. A strong tendency to diminished ATP and glycogen level was observed in older animals, 1 year after tamoxifen administration. Our study suggests important roles of cardiac AMPK at increased cardiac workload, potentially limiting exercise performance. This is at least partially due to impaired mitochondrial function and bioenergetics which degrades with age.
Collapse
Affiliation(s)
- Malgorzata Tokarska-Schlattner
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Laurence Kay
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Pascale Perret
- Inserm U1039, Radiopharmaceutiques Biocliniques, Faculté de Médecine, University of Grenoble Alpes, Grenoble, France
| | - Raffaella Isola
- Department of Biomedical Sciences, Division of Cytomorphology, University of Cagliari, Cagliari, Italy
| | - Stéphane Attia
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Frédéric Lamarche
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Cindy Tellier
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Cécile Cottet-Rousselle
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Amjad Uneisi
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Isabelle Hininger-Favier
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Marc Foretz
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Hervé Dubouchaud
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Catherine Ghezzi
- Inserm U1039, Radiopharmaceutiques Biocliniques, Faculté de Médecine, University of Grenoble Alpes, Grenoble, France
| | - Christian Zuppinger
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Benoit Viollet
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Uwe Schlattner
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
16
|
Abstract
The design of the energy metabolism system in striated muscle remains a major area of investigation. Here, we review our current understanding and emerging hypotheses regarding the metabolic support of muscle contraction. Maintenance of ATP free energy, so called energy homeostasis, via mitochondrial oxidative phosphorylation is critical to sustained contractile activity, and this major design criterion is the focus of this review. Cell volume invested in mitochondria reduces the space available for generating contractile force, and this spatial balance between mitochondria acontractile elements to meet the varying sustained power demands across muscle types is another important design criterion. This is accomplished with remarkably similar mass-specific mitochondrial protein composition across muscle types, implying that it is the organization of mitochondria within the muscle cell that is critical to supporting sustained muscle function. Beyond the production of ATP, ubiquitous distribution of ATPases throughout the muscle requires rapid distribution of potential energy across these large cells. Distribution of potential energy has long been thought to occur primarily through facilitated metabolite diffusion, but recent analysis has questioned the importance of this process under normal physiological conditions. Recent structural and functional studies have supported the hypothesis that the mitochondrial reticulum provides a rapid energy distribution system via the conduction of the mitochondrial membrane potential to maintain metabolic homeostasis during contractile activity. We extensively review this aspect of the energy metabolism design contrasting it with metabolite diffusion models and how mitochondrial structure can play a role in the delivery of energy in the striated muscle.
Collapse
Affiliation(s)
- Brian Glancy
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| | - Robert S Balaban
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| |
Collapse
|
17
|
Examination of gender differences in patients with takotsubo syndrome according to left ventricular biopsy: two case reports. J Med Case Rep 2021; 15:281. [PMID: 34016184 PMCID: PMC8139097 DOI: 10.1186/s13256-021-02856-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/08/2021] [Indexed: 01/27/2023] Open
Abstract
Background Takotsubo syndrome is a stress-induced disease that makes up 23% of acute coronary syndrome cases. However, its onset mechanism remains unclear. Although females are overwhelmingly affected, males end up having more cardiac complications. Case presentation We examined the differences in stress responses in the myocardium between sexes in patients with takotsubo syndrome. We biopsied samples from an over 70-year-old Japanese male and an over 80-year-old Japanese female. Tissues from the left ventricle apex in the acute phase and the apical ballooning-type were examined using histopathology and deoxyribonucleic acid (DNA) microarray analysis. Our data showed that left ventricular ejection fractions were 38% and 56%, and peak creatinine kinase concentrations during hospitalization were 629U/L and 361U/L, for the male and female patient, respectively. The pulmonary capillary wedge pressure was 26mmHg and 11mmHg for the male and female patient, respectively. Negative T did not return to normal in the male subject after 6months. Histopathology results indicated that contraction band necrosis and lymphocyte infiltration were more common in the male subject. Conclusions We noticed that possible differences may exist between male and female patients using pathological examination and some DNA analyses. In particular, it may help treat acute severity in males. We will elucidate the mechanism of takotsubo syndrome development by increasing the number of samples to support the reliability of the data in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s13256-021-02856-9.
Collapse
|
18
|
Abstract
The heart has the highest energy demands per gram of any organ in the body and energy metabolism fuels normal contractile function. Metabolic inflexibility and impairment of myocardial energetics occur with several common cardiac diseases, including ischemia and heart failure. This review explores several decades of innovation in cardiac magnetic resonance spectroscopy modalities and their use to noninvasively identify and quantify metabolic derangements in the normal, failing, and diseased heart. The implications of this noninvasive modality for predicting significant clinical outcomes and guiding future investigation and therapies to improve patient care are discussed.
Collapse
|
19
|
Bilous T, Kretsu N, Koloskova O. Markers of myocardial dysfunction in neonates with sepsis of various gestational age. ACTA MEDICA INTERNATIONAL 2021. [DOI: 10.4103/amit.amit_53_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
20
|
The Most Useful Method To Evaluate The Volume Status Of Critical Patients In The Emergency And Intensive Care Units: Point Of Care Ultrasound. JOURNAL OF CONTEMPORARY MEDICINE 2020. [DOI: 10.16899/jcm.728902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
21
|
Piquereau J, Veksler V, Novotova M, Ventura-Clapier R. Energetic Interactions Between Subcellular Organelles in Striated Muscles. Front Cell Dev Biol 2020; 8:581045. [PMID: 33134298 PMCID: PMC7561670 DOI: 10.3389/fcell.2020.581045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/15/2020] [Indexed: 01/12/2023] Open
Abstract
Adult striated muscle cells present highly organized structure with densely packed intracellular organelles and a very sparse cytosol accounting for only few percent of cell volume. These cells have a high and fluctuating energy demand that, in continuously working oxidative muscles, is fulfilled mainly by oxidative metabolism. ATP produced by mitochondria should be directed to the main energy consumers, ATPases of the excitation-contraction system; at the same time, ADP near ATPases should rapidly be eliminated. This is achieved by phosphotransfer kinases, the most important being creatine kinase (CK). Specific CK isoenzymes are located in mitochondria and in close proximity to ATPases, forming efficient energy shuttle between these structures. In addition to phosphotransfer kinases, ATP/ADP can be directly channeled between mitochondria co-localized with ATPases in a process called “direct adenine nucleotide channeling, DANC.” This process is highly plastic so that inactivation of the CK system increases the participation of DANC to energy supply owing to the rearrangement of cell structure. The machinery for DANC is built during postnatal development in parallel with the increase in mitochondrial mass, organization, and complexification of the cell structure. Disorganization of cell architecture remodels the mitochondrial network and decreases the efficacy of DANC, showing that this process is intimately linked to cardiomyocyte structure. Accordingly, in heart failure, disorganization of the cell structure along with decrease in mitochondrial mass reduces the efficacy of DANC and together with alteration of the CK shuttle participates in energetic deficiency contributing to contractile failure.
Collapse
Affiliation(s)
- Jérôme Piquereau
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France
| | - Vladimir Veksler
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France
| | - Marta Novotova
- Department of Cellular Cardiology, Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | |
Collapse
|
22
|
Hydrogen peroxide diffusion and scavenging shapes mitochondrial network instability and failure by sensitizing ROS-induced ROS release. Sci Rep 2020; 10:15758. [PMID: 32978406 PMCID: PMC7519669 DOI: 10.1038/s41598-020-71308-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
The mitochondrial network of cardiac cells is finely tuned for ATP delivery to sites of energy demand; however, emergent phenomena, such as mitochondrial transmembrane potential oscillations or propagating waves of depolarization have been observed under metabolic stress. While regenerative signaling by reactive oxygen species (ROS)-induced ROS release (RIRR) has been suggested as a potential trigger, it is unknown how it could lead to widespread responses. Here, we present a novel computational model of RIRR transmission that explains the mechanisms of this phenomenon. The results reveal that superoxide mediates neighbor-neighbor activation of energy-dissipating ion channels, while hydrogen peroxide distributes oxidative stress to sensitize the network to mitochondrial criticality. The findings demonstrate the feasibility of RIRR as a synchronizing factor across the dimensions of the adult heart cell and illustrate how a cascade of failures at the organellar level can scale to impact cell and organ level functions of the heart.
Collapse
|
23
|
Primed Left Ventricle Heart Perfusion Creates Physiological Aortic Pressure in Porcine Hearts. ASAIO J 2020; 66:55-63. [PMID: 30893130 DOI: 10.1097/mat.0000000000000947] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This article presents a primed left ventricle heart perfusion method to generate physiologic aortic pressure (AoP) and perform functional assessment. Isolated hearts of male Yorkshire pigs were used to study the hemodynamic behaviors of AoPs generated in the primed left ventricle heart perfusion (n = 6) and conventional (zero-loaded left ventricle) Langendorff perfusion (n = 6). The measurement results show that left ventricular pressure generated in the primed left ventricle heart perfusion is a determinant of physiologic AoP (i.e. systolic and diastolic pressures within physiologic range). The aortic pulse pressure (systolic pressure = 124.5 ± 1.7 mm Hg, diastolic pressure = 87.8 ± 0.9 mm Hg, aortic pulse pressure = 36.7 ± 2.6 mm Hg) from the primed left ventricle heart perfusion represents close match with the in vivo physiologic data. The volume in the left ventricle remains constant throughout the primed left ventricle heart perfusion, which allows us to perform isovolumetric left ventricular pressure measurement in ex vivo heart perfusion (EVHP). Left ventricular contractility measurements (maximum and minimum rates of left ventricular pressure change) were derived for cardiac assessment. In summary, the proposed primed left ventricle heart perfusion method is able to create physiologic AoP and enables left ventricular functional assessment in EVHP in porcine hearts.
Collapse
|
24
|
Peterzan MA, Lewis AJM, Neubauer S, Rider OJ. Non-invasive investigation of myocardial energetics in cardiac disease using 31P magnetic resonance spectroscopy. Cardiovasc Diagn Ther 2020; 10:625-635. [PMID: 32695642 DOI: 10.21037/cdt-20-275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiac metabolism and function are intrinsically linked. High-energy phosphates occupy a central and obligate position in cardiac metabolism, coupling oxygen and substrate fuel delivery to the myocardium with external work. This insight underlies the widespread clinical use of ischaemia testing. However, other deficits in high-energy phosphate metabolism (not secondary to supply-demand mismatch of oxygen and substrate fuels) may also be documented, and are of particular interest when found in the context of structural heart disease. This review introduces the scope of deficits in high-energy phosphate metabolism that may be observed in the myocardium, how to assess for them, and how they might be interpreted.
Collapse
Affiliation(s)
- Mark A Peterzan
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J M Lewis
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Oliver J Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Zuurbier CJ, Bertrand L, Beauloye CR, Andreadou I, Ruiz‐Meana M, Jespersen NR, Kula‐Alwar D, Prag HA, Eric Botker H, Dambrova M, Montessuit C, Kaambre T, Liepinsh E, Brookes PS, Krieg T. Cardiac metabolism as a driver and therapeutic target of myocardial infarction. J Cell Mol Med 2020; 24:5937-5954. [PMID: 32384583 PMCID: PMC7294140 DOI: 10.1111/jcmm.15180] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/13/2020] [Accepted: 03/08/2020] [Indexed: 12/11/2022] Open
Abstract
Reducing infarct size during a cardiac ischaemic-reperfusion episode is still of paramount importance, because the extension of myocardial necrosis is an important risk factor for developing heart failure. Cardiac ischaemia-reperfusion injury (IRI) is in principle a metabolic pathology as it is caused by abruptly halted metabolism during the ischaemic episode and exacerbated by sudden restart of specific metabolic pathways at reperfusion. It should therefore not come as a surprise that therapy directed at metabolic pathways can modulate IRI. Here, we summarize the current knowledge of important metabolic pathways as therapeutic targets to combat cardiac IRI. Activating metabolic pathways such as glycolysis (eg AMPK activators), glucose oxidation (activating pyruvate dehydrogenase complex), ketone oxidation (increasing ketone plasma levels), hexosamine biosynthesis pathway (O-GlcNAcylation; administration of glucosamine/glutamine) and deacetylation (activating sirtuins 1 or 3; administration of NAD+ -boosting compounds) all seem to hold promise to reduce acute IRI. In contrast, some metabolic pathways may offer protection through diminished activity. These pathways comprise the malate-aspartate shuttle (in need of novel specific reversible inhibitors), mitochondrial oxygen consumption, fatty acid oxidation (CD36 inhibitors, malonyl-CoA decarboxylase inhibitors) and mitochondrial succinate metabolism (malonate). Additionally, protecting the cristae structure of the mitochondria during IR, by maintaining the association of hexokinase II or creatine kinase with mitochondria, or inhibiting destabilization of FO F1 -ATPase dimers, prevents mitochondrial damage and thereby reduces cardiac IRI. Currently, the most promising and druggable metabolic therapy against cardiac IRI seems to be the singular or combined targeting of glycolysis, O-GlcNAcylation and metabolism of ketones, fatty acids and succinate.
Collapse
Affiliation(s)
- Coert J. Zuurbier
- Department of AnesthesiologyLaboratory of Experimental Intensive Care and AnesthesiologyAmsterdam Infection & ImmunityAmsterdam Cardiovascular SciencesAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Luc Bertrand
- Institut de Recherche Expérimentale et CliniquePole of Cardiovascular ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Christoph R. Beauloye
- Institut de Recherche Expérimentale et CliniquePole of Cardiovascular ResearchUniversité catholique de LouvainBrusselsBelgium
- Cliniques Universitaires Saint‐LucBrusselsBelgium
| | - Ioanna Andreadou
- Laboratory of PharmacologyFaculty of PharmacyNational and Kapodistrian University of AthensAthensGreece
| | - Marisol Ruiz‐Meana
- Department of CardiologyHospital Universitari Vall d’HebronVall d’Hebron Institut de Recerca (VHIR)CIBER‐CVUniversitat Autonoma de Barcelona and Centro de Investigación Biomédica en Red‐CVMadridSpain
| | | | | | - Hiran A. Prag
- Department of MedicineUniversity of CambridgeCambridgeUK
| | - Hans Eric Botker
- Department of CardiologyAarhus University HospitalAarhus NDenmark
| | - Maija Dambrova
- Pharmaceutical PharmacologyLatvian Institute of Organic SynthesisRigaLatvia
| | - Christophe Montessuit
- Department of Pathology and ImmunologyUniversity of Geneva School of MedicineGenevaSwitzerland
| | - Tuuli Kaambre
- Laboratory of Chemical BiologyNational Institute of Chemical Physics and BiophysicsTallinnEstonia
| | - Edgars Liepinsh
- Pharmaceutical PharmacologyLatvian Institute of Organic SynthesisRigaLatvia
| | - Paul S. Brookes
- Department of AnesthesiologyUniversity of Rochester Medical CenterRochesterNYUSA
| | - Thomas Krieg
- Department of MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
26
|
Peterzan MA, Clarke WT, Lygate CA, Lake HA, Lau JYC, Miller JJ, Johnson E, Rayner JJ, Hundertmark MJ, Sayeed R, Petrou M, Krasopoulos G, Srivastava V, Neubauer S, Rodgers CT, Rider OJ. Cardiac Energetics in Patients With Aortic Stenosis and Preserved Versus Reduced Ejection Fraction. Circulation 2020; 141:1971-1985. [PMID: 32438845 PMCID: PMC7294745 DOI: 10.1161/circulationaha.119.043450] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Supplemental Digital Content is available in the text. Why some but not all patients with severe aortic stenosis (SevAS) develop otherwise unexplained reduced systolic function is unclear. We investigate the hypothesis that reduced creatine kinase (CK) capacity and flux is associated with this transition.
Collapse
Affiliation(s)
- Mark A Peterzan
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine (M.A.P., J.Y.C.L., J.J.M., J.J.R., M.J.H., S.N., O.J.R.), University of Oxford, United Kingdom
| | - William T Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences (W.T.C.), University of Oxford, United Kingdom
| | | | - Hannah A Lake
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (H.A.L.), University of Oxford, United Kingdom
| | - Justin Y C Lau
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine (M.A.P., J.Y.C.L., J.J.M., J.J.R., M.J.H., S.N., O.J.R.), University of Oxford, United Kingdom
| | - Jack J Miller
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine (M.A.P., J.Y.C.L., J.J.M., J.J.R., M.J.H., S.N., O.J.R.), University of Oxford, United Kingdom
| | - Errin Johnson
- Dunn School of Pathology (E.J.), University of Oxford, United Kingdom
| | - Jennifer J Rayner
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine (M.A.P., J.Y.C.L., J.J.M., J.J.R., M.J.H., S.N., O.J.R.), University of Oxford, United Kingdom
| | - Moritz J Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine (M.A.P., J.Y.C.L., J.J.M., J.J.R., M.J.H., S.N., O.J.R.), University of Oxford, United Kingdom
| | - Rana Sayeed
- Department of Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, United Kingdom (R.S., G.K., V.S.)
| | - Mario Petrou
- Department of Cardiothoracic Surgery, Royal Brompton and Harefield National Health Service Foundation Trust, London, United Kingdom (M.P.)
| | - George Krasopoulos
- Department of Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, United Kingdom (R.S., G.K., V.S.)
| | - Vivek Srivastava
- Department of Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, United Kingdom (R.S., G.K., V.S.)
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine (M.A.P., J.Y.C.L., J.J.M., J.J.R., M.J.H., S.N., O.J.R.), University of Oxford, United Kingdom
| | | | - Oliver J Rider
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine (M.A.P., J.Y.C.L., J.J.M., J.J.R., M.J.H., S.N., O.J.R.), University of Oxford, United Kingdom
| |
Collapse
|
27
|
Klepinin A, Zhang S, Klepinina L, Rebane-Klemm E, Terzic A, Kaambre T, Dzeja P. Adenylate Kinase and Metabolic Signaling in Cancer Cells. Front Oncol 2020; 10:660. [PMID: 32509571 PMCID: PMC7248387 DOI: 10.3389/fonc.2020.00660] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/08/2020] [Indexed: 12/23/2022] Open
Abstract
A hallmark of cancer cells is the ability to rewire their bioenergetics and metabolic signaling circuits to fuel their uncontrolled proliferation and metastasis. Adenylate kinase (AK) is the critical enzyme in the metabolic monitoring of cellular adenine nucleotide homeostasis. It also directs AK→ AMP→ AMPK signaling controlling cell cycle and proliferation, and ATP energy transfer from mitochondria to distribute energy among cellular processes. The significance of AK isoform network in the regulation of a variety of cellular processes, which include cell differentiation and motility, is rapidly growing. Adenylate kinase 2 (AK2) isoform, localized in intermembrane and intra-cristae space, is vital for mitochondria nucleotide exchange and ATP export. AK2 deficiency disrupts cell energetics, causes severe human diseases, and is embryonically lethal in mice, signifying the importance of catalyzed phosphotransfer in cellular energetics. Suppression of AK phosphotransfer and AMP generation in cancer cells and consequently signaling through AMPK could be an important factor in the initiation of cancerous transformation, unleashing uncontrolled cell cycle and growth. Evidence also builds up that shift in AK isoforms is used later by cancer cells for rewiring energy metabolism to support their high proliferation activity and tumor progression. As cell motility is an energy-consuming process, positioning of AK isoforms to increased energy consumption sites could be an essential factor to incline cancer cells to metastases. In this review, we summarize recent advances in studies of the significance of AK isoforms involved in cancer cell metabolism, metabolic signaling, metastatic potential, and a therapeutic target.
Collapse
Affiliation(s)
- Aleksandr Klepinin
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Song Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ljudmila Klepinina
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Egle Rebane-Klemm
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Petras Dzeja
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
28
|
Takeuchi A, Matsuoka S. Integration of mitochondrial energetics in heart with mathematical modelling. J Physiol 2020; 598:1443-1457. [DOI: 10.1113/jp276817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/23/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ayako Takeuchi
- Department of Integrative and Systems PhysiologyFaculty of Medical Sciencesand Life Science Innovation CenterUniversity of Fukui Fukui 910‐1193 Japan
| | - Satoshi Matsuoka
- Department of Integrative and Systems PhysiologyFaculty of Medical Sciencesand Life Science Innovation CenterUniversity of Fukui Fukui 910‐1193 Japan
| |
Collapse
|
29
|
Kuznetsov AV, Javadov S, Grimm M, Margreiter R, Ausserlechner MJ, Hagenbuchner J. Crosstalk between Mitochondria and Cytoskeleton in Cardiac Cells. Cells 2020; 9:cells9010222. [PMID: 31963121 PMCID: PMC7017221 DOI: 10.3390/cells9010222] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/28/2022] Open
Abstract
Elucidation of the mitochondrial regulatory mechanisms for the understanding of muscle bioenergetics and the role of mitochondria is a fundamental problem in cellular physiology and pathophysiology. The cytoskeleton (microtubules, intermediate filaments, microfilaments) plays a central role in the maintenance of mitochondrial shape, location, and motility. In addition, numerous interactions between cytoskeletal proteins and mitochondria can actively participate in the regulation of mitochondrial respiration and oxidative phosphorylation. In cardiac and skeletal muscles, mitochondrial positions are tightly fixed, providing their regular arrangement and numerous interactions with other cellular structures such as sarcoplasmic reticulum and cytoskeleton. This can involve association of cytoskeletal proteins with voltage-dependent anion channel (VDAC), thereby, governing the permeability of the outer mitochondrial membrane (OMM) to metabolites, and regulating cell energy metabolism. Cardiomyocytes and myocardial fibers demonstrate regular arrangement of tubulin beta-II isoform entirely co-localized with mitochondria, in contrast to other isoforms of tubulin. This observation suggests the participation of tubulin beta-II in the regulation of OMM permeability through interaction with VDAC. The OMM permeability is also regulated by the specific isoform of cytolinker protein plectin. This review summarizes and discusses previous studies on the role of cytoskeletal proteins in the regulation of energy metabolism and mitochondrial function, adenosine triphosphate (ATP) production, and energy transfer.
Collapse
Affiliation(s)
- Andrey V. Kuznetsov
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, 6020 Innsbruck, Austria;
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria;
- Correspondence: (A.V.K.); (J.H.); Tel.: +43-512-504-27815 (A.V.K.); +43-512-504-81578 (J.H.)
| | - Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, USA;
| | - Michael Grimm
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, 6020 Innsbruck, Austria;
| | - Raimund Margreiter
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | | | - Judith Hagenbuchner
- Department of Paediatrics II, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (A.V.K.); (J.H.); Tel.: +43-512-504-27815 (A.V.K.); +43-512-504-81578 (J.H.)
| |
Collapse
|
30
|
Garlid AO, Schaffer CT, Kim J, Bhatt H, Guevara-Gonzalez V, Ping P. TAZ encodes tafazzin, a transacylase essential for cardiolipin formation and central to the etiology of Barth syndrome. Gene 2019; 726:144148. [PMID: 31647997 DOI: 10.1016/j.gene.2019.144148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/12/2019] [Accepted: 09/27/2019] [Indexed: 12/31/2022]
Abstract
Tafazzin, which is encoded by the TAZ gene, catalyzes transacylation to form mature cardiolipin and shows preference for the transfer of a linoleic acid (LA) group from phosphatidylcholine (PC) to monolysocardiolipin (MLCL) with influence from mitochondrial membrane curvature. The protein contains domains and motifs involved in targeting, anchoring, and an active site for transacylase activity. Tafazzin activity affects many aspects of mitochondrial structure and function, including that of the electron transport chain, fission-fusion, as well as apoptotic signaling. TAZ mutations are implicated in Barth syndrome, an underdiagnosed and devastating disease that primarily affects male pediatric patients with a broad spectrum of disease pathologies that impact the cardiovascular, neuromuscular, metabolic, and hematologic systems.
Collapse
Affiliation(s)
- Anders O Garlid
- Cardiovascular Data Science Training Program at UCLA, University of California at Los Angeles, CA 90095, USA; Department of Physiology, University of California at Los Angeles, CA 90095, USA.
| | - Calvin T Schaffer
- Cardiovascular Data Science Training Program at UCLA, University of California at Los Angeles, CA 90095, USA; Department of Physiology, University of California at Los Angeles, CA 90095, USA
| | - Jaewoo Kim
- Cardiovascular Data Science Training Program at UCLA, University of California at Los Angeles, CA 90095, USA; Department of Physiology, University of California at Los Angeles, CA 90095, USA
| | - Hirsh Bhatt
- Cardiovascular Data Science Training Program at UCLA, University of California at Los Angeles, CA 90095, USA; Department of Physiology, University of California at Los Angeles, CA 90095, USA
| | - Vladimir Guevara-Gonzalez
- Cardiovascular Data Science Training Program at UCLA, University of California at Los Angeles, CA 90095, USA; Department of Mathematics, University of California at Los Angeles, CA 90095, USA
| | - Peipei Ping
- Cardiovascular Data Science Training Program at UCLA, University of California at Los Angeles, CA 90095, USA; Department of Physiology, University of California at Los Angeles, CA 90095, USA; Department of Medicine/Cardiology, University of California at Los Angeles, CA 90095, USA; Department of Bioinformatics, University of California at Los Angeles, CA 90095, USA; Scalable Analytics Institute (ScAi), University of California at Los Angeles, CA 90095, USA.
| |
Collapse
|
31
|
Creatine nanoliposome reverts the HPA-induced damage in complex II–III activity of the rats’ cerebral cortex. Mol Biol Rep 2019; 46:5897-5908. [DOI: 10.1007/s11033-019-05023-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/06/2019] [Indexed: 12/12/2022]
|
32
|
Abdou RH, Basha WA, Khalil WF. Subacute Toxicity of Nerium oleander Ethanolic Extract in Mice. Toxicol Res 2019; 35:233-239. [PMID: 31341552 PMCID: PMC6629440 DOI: 10.5487/tr.2019.35.3.233] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/20/2018] [Accepted: 12/22/2018] [Indexed: 01/04/2023] Open
Abstract
Nerium oleander (N. oleander) is a well-known poisonous shrub that is frequently grown in gardens and public areas and contains numerous toxic compounds. The major toxic components are the cardiac glycosides oleandrin and neriin. The aim of our study was to evaluate the toxic effects of an ethanolic N. oleander leaf extract on haematological, cardiac, inflammatory, and serum biochemical parameters, as well as histopathological changes in the heart. N. oleander extract was orally administered for 14 and 30 consecutive days at doses of 100 and 200 mg of dried extract/kg of body weight in 0.5 mL of saline. The results showed significant increases in mean corpuscular volume, white blood cell counts, platelet counts, interleukins (IL-1 and IL-6), tumour necrosis factor alpha, C reactive protein, alanine aminotransferase, lactate dehydrogenase, creatine kinase and creatine kinase MB, especially at high doses. Marked pathological changes were perceived in the heart tissue. Thus, it can be concluded that exposure to N. oleander leaf extract adversely affects the heart and liver.
Collapse
Affiliation(s)
- Rania H Abdou
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Walaa A Basha
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Waleed F Khalil
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
33
|
Mado K, Chekulayev V, Shevchuk I, Puurand M, Tepp K, Kaambre T. On the role of tubulin, plectin, desmin, and vimentin in the regulation of mitochondrial energy fluxes in muscle cells. Am J Physiol Cell Physiol 2019; 316:C657-C667. [PMID: 30811221 DOI: 10.1152/ajpcell.00303.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitochondria perform a central role in life and death of the eukaryotic cell. They are major players in the generation of macroergic compounds and function as integrated signaling pathways, including the regulation of Ca2+ signals and apoptosis. A growing amount of evidence is demonstrating that mitochondria of muscle cells use cytoskeletal proteins (both microtubules and intermediate filaments) not only for their movement and proper cellular positioning, but also to maintain their biogenesis, morphology, function, and regulation of energy fluxes through the outer mitochondrial membrane (MOM). Here we consider the known literature data concerning the role of tubulin, plectin, desmin and vimentin in bioenergetic function of mitochondria in striated muscle cells, as well as in controlling the permeability of MOM for adenine nucleotides (ADNs). This is of great interest since dysfunctionality of these cytoskeletal proteins has been shown to result in severe myopathy associated with pronounced mitochondrial dysfunction. Further efforts are needed to uncover the pathways by which the cytoskeleton supports the functional capacity of mitochondria and transport of ADN(s) across the MOM (through voltage-dependent anion channel).
Collapse
Affiliation(s)
- Kati Mado
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Vladimir Chekulayev
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Igor Shevchuk
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Marju Puurand
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Kersti Tepp
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| |
Collapse
|
34
|
Ventura-Clapier R, Piquereau J, Veksler V, Garnier A. Estrogens, Estrogen Receptors Effects on Cardiac and Skeletal Muscle Mitochondria. Front Endocrinol (Lausanne) 2019; 10:557. [PMID: 31474941 PMCID: PMC6702264 DOI: 10.3389/fendo.2019.00557] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/29/2019] [Indexed: 12/27/2022] Open
Abstract
Mitochondria are unique organelles present in almost all cell types. They are involved not only in the supply of energy to the host cell, but also in multiple biochemical and biological processes like calcium homeostasis, production, and regulation of reactive oxygen species (ROS), pH control, or cell death. The importance of mitochondria in cell biology and pathology is increasingly recognized. Being maternally inherited, mitochondria exhibit a tissue-specificity, because most of the mitochondrial proteins are encoded by the nuclear genome. This renders them exquisitely well-adapted to the physiology of the host cell. It is thus not surprising that mitochondria show a sexual dimorphism and that they are also prone to the influence of sex chromosomes and sex hormones. Estrogens affect mitochondria through multiple processes involving membrane and nuclear estrogen receptors (ERs) as well as more direct effects. Moreover, estrogen receptors have been identified within mitochondria. The effects of estrogens on mitochondria comprise protein content and specific activity of mitochondrial proteins, phospholipid content of membranes, oxidant and anti-oxidant capacities, oxidative phosphorylation, and calcium retention capacities. Herein we will briefly review the life cycle and functions of mitochondria, the importance of estrogen receptors and the effects of estrogens on heart and skeletal muscle mitochondria.
Collapse
|
35
|
Involvement of the phosphoryl transfer network on cardiac energetic metabolism during Staphylococcus aureus infection and its association to disease pathophysiology. Microb Pathog 2018; 126:318-322. [PMID: 30439401 DOI: 10.1016/j.micpath.2018.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/29/2018] [Accepted: 11/06/2018] [Indexed: 11/21/2022]
Abstract
Evidences have suggested that the phosphoryl transfer network by the enzymatic activities of creatine kinase (CK), adenylate kinase (AK), pyruvate kinase (PK), and lactate dehydrogenase (LDH), shows new perspectives to understand some disturbances in the energy metabolism during bacterial infections. Thus, the aim of this study was to evaluate whether Staphylococcus aureus infection in mice could alter serum and cardiac activities of these enzymes and their association to disease pathophysiology. For that, we measured total leukocytes, lymphocytes and neutrophils (just 48 h of infection) that were lower in infected animals after 48 and 72 h in infected mice compared with negative control, while total protein and globulin plasma levels were higher after 72 h of infection. The serum CK activity was higher in infected animals 48 and 72 h post-infection compared to the control group, as well as observed for mitochondrial cardiac CK activity. The serum PK activity was higher in infected animals after 72 h of infection compared to the control group, and lower in the cardiac tissue. The cardiac AK activity was lower in infected animals 48 h and 72 h post-infection compared to the control group, while serum and cardiac LDH activities were higher. Based on these evidences, it is possible to conclude that the stimulation of CK activity exerts a key role as an attempt to maintain the bioenergetic homeostasis by the production of phosphocreatine to avoid a rapid fall on the concentrations of total adenosine triphosphate. In summary, the phosphoryl transfer network can be considered a pathway involved in the improvement on tissue and cellular energy homeostasis of S. aureus-infected mice.
Collapse
|
36
|
Intracellular Energy-Transfer Networks and High-Resolution Respirometry: A Convenient Approach for Studying Their Function. Int J Mol Sci 2018; 19:ijms19102933. [PMID: 30261663 PMCID: PMC6213097 DOI: 10.3390/ijms19102933] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
Compartmentalization of high-energy phosphate carriers between intracellular micro-compartments is a phenomenon that ensures efficient energy use. To connect these sites, creatine kinase (CK) and adenylate kinase (AK) energy-transfer networks, which are functionally coupled to oxidative phosphorylation (OXPHOS), could serve as important regulators of cellular energy fluxes. Here, we introduce how selective permeabilization of cellular outer membrane and high-resolution respirometry can be used to study functional coupling between CK or AK pathways and OXPHOS in different cells and tissues. Using the protocols presented here the ability of creatine or adenosine monophosphate to stimulate OXPHOS through CK and AK reactions, respectively, is easily observable and quantifiable. Additionally, functional coupling between hexokinase and mitochondria can be investigated by monitoring the effect of glucose on respiration. Taken together, high-resolution respirometry in combination with permeabilization is a convenient approach for investigating energy-transfer networks in small quantities of cells and tissues in health and in pathology.
Collapse
|
37
|
Jaguezeski AM, Baldissera MD, Rhoden LA, Gomes TMA, Mendes RE, Bottari NB, Morsch VM, Schetinger MRC, Stefani LM, Giongo JL, Vaucher RA, Da Silva AS. Listeria monocytogenes impairs enzymes of the phosphotransfer network and alters antioxidant/oxidant status in cattle brain structures. Microb Pathog 2018; 124:284-290. [PMID: 30142467 DOI: 10.1016/j.micpath.2018.08.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/29/2018] [Accepted: 08/20/2018] [Indexed: 01/24/2023]
Abstract
Several evidences have suggested the involvement of enzymes belonging to the phosphotransfer network, formed by creatine kinase (CK), pyruvate kinase (PK) and adenylate kinase (AK), as well the oxidative stress on the pathogenesis of infectious diseases associated with the central nervous system (CNS). Thus, the aim of this study was to evaluate whether listeriosis alters the brain energy metabolism and/or causes oxidative stress in different brain structures of cattle experimentally infected by Listeria monocytogenes. The cytosolic CK activity was inhibited in the cerebral cortex, cerebellum, brainstem and hippocampus of infected animals compared to uninfected animals, while the mitochondrial CK activity was increased. The PK activity was inhibited in all brain structures of infected animals, while the AK activity was unchanged. Na+, K+-ATPase activity decreased in the cerebral cortex, cerebellum and hippocampus of animals infected by L. monocytogenes. Regarding the oxidative strees variables, the cerebellum and brainstem of infected animals showed increased thiobarbituric acid reactive substances, while the catalase activity was inhibited. Glutathione S-transferarase was inhibited in the cerebral cortex and brainstem of infected animals, and it was increased in the cerebellum. L. monocytogenes was quantified in the liver (n = 5/5) and cerebral cortex (n = 4/5) of the infected cattle. Based on these evidences, the nucleocytoplasmic communication between CK isoenzymes was insufficient to avoid an impairment of cerebral bioenergetics. Moreover, the inhibition on brain PK activity caused an impairment in the communication between sites of ATP generation and ATP utilization. The lipid peroxidation and alteration on antioxidant status observed in some brain structures were also involved during the disease. In summary, these alterations contribute to disease pathogenesis linked to CNS during cattle listeriosis.
Collapse
Affiliation(s)
- Antonise M Jaguezeski
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina, Chapecó, Santa Catarina, Brazil
| | - Matheus D Baldissera
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Leandro A Rhoden
- Laboratory of Pathology, Instituto Federal Catarinense, Concórdia, Santa Catarina, Brazil
| | - Teane M A Gomes
- Laboratory of Pathology, Instituto Federal Catarinense, Concórdia, Santa Catarina, Brazil
| | - Ricardo E Mendes
- Laboratory of Pathology, Instituto Federal Catarinense, Concórdia, Santa Catarina, Brazil
| | - Nathieli B Bottari
- Graduate Program in Toxicological Biochemistry, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Vera M Morsch
- Graduate Program in Toxicological Biochemistry, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Maria Rosa C Schetinger
- Graduate Program in Toxicological Biochemistry, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Lenita M Stefani
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina, Chapecó, Santa Catarina, Brazil
| | - Janice L Giongo
- Pharmacy Laboratory, Faculdade Anhanguera, Pelotas, RS, Brazil
| | - Rodrigo A Vaucher
- Laboratory of Biochemistry Research and Molecular Biology of Microorganisms (LaPeBBiOM), Universidade Federal de Pelotas, RS, Brazil
| | - Aleksandro Schafer Da Silva
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina, Chapecó, Santa Catarina, Brazil; Graduate Program in Toxicological Biochemistry, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
38
|
Abstract
Isoforms of creatine kinase (CK) generate and use phosphocreatine, a concentrated and highly diffusible cellular "high energy" intermediate, for the main purpose of energy buffering and transfer in order to maintain cellular energy homeostasis. The mitochondrial CK isoform (mtCK) localizes to the mitochondrial intermembrane and cristae space, where it assembles into peripherally membrane-bound, large cuboidal homooctamers. These are part of proteolipid complexes wherein mtCK directly interacts with cardiolipin and other anionic phospholipids, as well as with the VDAC channel in the outer membrane. This leads to a stabilization and cross-linking of inner and outer mitochondrial membrane, forming so-called contact sites. Also the adenine nucleotide translocator of the inner membrane can be recruited into these proteolipid complexes, probably mediated by cardiolipin. The complexes have functions mainly in energy transfer to the cytosol and stimulation of oxidative phosphorylation, but also in restraining formation of reactive oxygen species and apoptosis. In vitro evidence indicates a putative role of mtCK in mitochondrial phospholipid distribution, and most recently a role in thermogenesis has been proposed. This review summarizes the essential structural and functional data of these mtCK complexes and describes in more detail the more recent advances in phospholipid interaction, thermogenesis, cancer and evolution of mtCK.
Collapse
|
39
|
Piquereau J, Ventura-Clapier R. Maturation of Cardiac Energy Metabolism During Perinatal Development. Front Physiol 2018; 9:959. [PMID: 30072919 PMCID: PMC6060230 DOI: 10.3389/fphys.2018.00959] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/29/2018] [Indexed: 12/26/2022] Open
Abstract
As one of the highest energy consumer organ in mammals, the heart has to be provided with a high amount of energy as soon as its first beats in utero. During the development of this organ, energy is produced within the cardiac muscle cell depending on substrate availability, oxygen pressure and cardiac workload that drastically change at birth. Thus, energy metabolism relying essentially on carbohydrates in fetal heart is very different from the adult one and birth is the trigger of a profound maturation which ensures the transition to a highly oxidative metabolism depending on lipid utilization. To face the substantial increase in cardiac workload resulting from the growth of the organism during the postnatal period, the heart not only develops its capacity for energy production but also undergoes a hypertrophic growth to adapt its contractile capacity to its new function. This leads to a profound cytoarchitectural remodeling of the cardiomyocyte which becomes a highly compartmentalized structure. As a consequence, within the mature cardiac muscle, energy transfer between energy producing and consuming compartments requires organized energy transfer systems that are established in the early postnatal life. This review aims at describing the major rearrangements of energy metabolism during the perinatal development.
Collapse
Affiliation(s)
- Jérôme Piquereau
- Signalling and Cardiovascular Pathophysiology - UMR-S 1180, Université Paris-Sud, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Châtenay-Malabry, France
| | - Renée Ventura-Clapier
- Signalling and Cardiovascular Pathophysiology - UMR-S 1180, Université Paris-Sud, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
40
|
Tatekoshi Y, Tanno M, Kouzu H, Abe K, Miki T, Kuno A, Yano T, Ishikawa S, Ohwada W, Sato T, Niinuma T, Suzuki H, Miura T. Translational regulation by miR-301b upregulates AMP deaminase in diabetic hearts. J Mol Cell Cardiol 2018; 119:138-146. [PMID: 29733818 DOI: 10.1016/j.yjmcc.2018.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
AMP deaminase (AMPD) plays a crucial role in adenine nucleotide metabolism. Recently we found that upregulated AMPD activity is associated with ATP depletion and contractile dysfunction under the condition of pressure overloading in the heart of a rat model of type 2 diabetes mellitus (T2DM), OLETF. Here we examined the mechanism of AMPD upregulation by T2DM. The protein level of 90-kDa full-length AMPD3 was increased in whole myocardial lysates by 55% in OLETF compared to those in LETO, a non-diabetic control. In contrast, the mRNA levels of AMPD3 in the myocardium were similar in OLETF and LETO. AMPD3 was comparably ubiquitinated in OLETF and LETO, and its degradation ex vivo was more sensitive to MG-132, a proteasome inhibitor, in OLETF than in LETO. MicroRNA array analysis revealed downregulation (>50%) of 57 microRNAs in OLETF compared to those in LETO, among which miR-301b was predicted to interact with the 3'UTR of AMPD3 mRNA. AMPD3 protein level was significantly increased by a miR-301b inhibitor and was decreased by a miR-301b mimetic in H9c2 cells. A luciferase reporter assay confirmed binding of miR-301b to the 3'UTR of AMPD3 mRNA. Transfection of neonatal rat cardiomyocytes with a miR-301b inhibitor increased 90-kDa AMPD3 and reduced ATP level. The results indicate that translational regulation by miR-301b mediates upregulated expression of cardiac AMPD3 protein in OLETF, which potentially reduces the adenine nucleotide pool at the time of increased work load. The miR-301b-AMPD3 axis may be a novel therapeutic target for intervening enegy metabolism in diabetic hearts.
Collapse
Affiliation(s)
- Yuki Tatekoshi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaya Tanno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hidemichi Kouzu
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koki Abe
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Miki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Satoko Ishikawa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Wataru Ohwada
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takeshi Niinuma
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| |
Collapse
|
41
|
Role of superoxide ion formation in hypothermia/rewarming induced contractile dysfunction in cardiomyocytes. Cryobiology 2018; 81:57-64. [PMID: 29458041 DOI: 10.1016/j.cryobiol.2018.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 01/24/2023]
Abstract
Rewarming following accidental hypothermia is associated with circulatory collapse due primarily to impaired cardiac contractile (systolic) function. Previously, we found that reduced myofilament Ca2+ sensitivity underlies hypothermia/rewarming (H/R)-induced cardiac contractile dysfunction. This reduced Ca2+ sensitivity is associated with troponin I (cTnI) phosphorylation. We hypothesize that H/R induces reactive oxygen species (ROS) formation in cardiomyocytes, which leads to cTnI phosphorylation and reduced myofilament Ca2+ sensitivity. To test this hypothesis, we exposed isolated rat cardiomyocytes to a 2-h period of severe hypothermia (15 °C) followed by rewarming (35 °C) with and without antioxidant (TEMPOL) treatment. Simultaneous measurements of cytosolic Ca2+ ([Ca2+]cyto) and contractile (sarcomere shortening) responses indicated that H/R-induced contractile dysfunction and reduced Ca2+ sensitivity was prevented in cardiomyocytes treated with TEMPOL. In addition, TEMPOL treatment blunted H/R-induced cTnI phosphorylation. These results support our overall hypothesis and suggest that H/R disrupts excitation-contraction coupling of the myocardium through a cascade of event triggered by excessive ROS formation during hypothermia. Antioxidant treatment may improve successful rescue of accidental hypothermia victims.
Collapse
|
42
|
Zou R, Shi W, Tao J, Li H, Lin X, Yang S, Hua P. SIRT5 and post-translational protein modifications: A potential therapeutic target for myocardial ischemia-reperfusion injury with regard to mitochondrial dynamics and oxidative metabolism. Eur J Pharmacol 2018; 818:410-418. [DOI: 10.1016/j.ejphar.2017.11.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 10/23/2017] [Accepted: 11/01/2017] [Indexed: 11/27/2022]
|
43
|
Chaui-Berlinck JG, Monteiro LHA. Frank-Starling mechanism and short-term adjustment of cardiac flow. ACTA ACUST UNITED AC 2017; 220:4391-4398. [PMID: 28912258 DOI: 10.1242/jeb.167106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/11/2017] [Indexed: 11/20/2022]
Abstract
The Frank-Starling law of the heart is a filling-force mechanism (FFm), a positive relationship between the distension of a ventricular chamber and its force of ejection, and such a mechanism is found across all the studied vertebrate lineages. The functioning of the cardiovascular system is usually described by means of the cardiac and vascular functions, the former related to the contractility of the heart and the latter related to the afterload imposed on the ventricle. The crossing of these functions is the so-called 'operation point', and the FFm is supposed to play a stabilizing role for the short-term variations in the working of the system. In the present study, we analyze whether the FFm is truly responsible for such a stability within two different settings: one-ventricle and two-ventricle hearts. To approach the query, we linearized the region around an arbitrary operation point and put forward a dynamical system of differential equations to describe the relationship among volumes in face of blood flows governed by pressure differences between compartments. Our results show that the FFm is not necessary to give stability to an operation point. Thus, which forces selected and maintained such a mechanism in all vertebrates? The present results indicate three different and complementary roles for the FFm: (1) it decreases the demands of a central controlling system over the circulatory system; (2) it smooths out perturbations in volumes; and (3) it guarantees faster transitions between operation points, i.e. it allows for rapid changes in cardiac output.
Collapse
Affiliation(s)
- José Guilherme Chaui-Berlinck
- Energetics and Theoretical Physiology Laboratory, Department of Physiology, Biosciences Institute, University of São Paulo, Rua do Matão, 101, CEP 05508-090, São Paulo, Brazil
| | - Luiz Henrique Alves Monteiro
- Escola de Engenharia da Universidade Presbiteriana Mackenzie, Rua da Consolação, 930 - Consolação, São Paulo - SP/Brasil, CEP 01302-907, Brazil.,Escola Politécnica da Universidade de São Paulo, Department of Telecommunications and Control, Av. Prof. Luciano Gualberto, 380 - CEP 05508-010 - São Paulo - SP/Brasil, Brazil
| |
Collapse
|
44
|
Peterzan MA, Lygate CA, Neubauer S, Rider OJ. Metabolic remodeling in hypertrophied and failing myocardium: a review. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646030 DOI: 10.1152/ajpheart.00731.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The energy starvation hypothesis proposes that maladaptive metabolic remodeling antedates, initiates, and maintains adverse contractile dysfunction in heart failure (HF). Better understanding of the cardiac metabolic phenotype and metabolic signaling could help identify the role metabolic remodeling plays within HF and the conditions known to transition toward HF, including "pathological" hypertrophy. In this review, we discuss metabolic phenotype and metabolic signaling in the contexts of pathological hypertrophy and HF. We discuss the significance of alterations in energy supply (substrate utilization, oxidative capacity, and phosphotransfer) and energy sensing using observations from human and animal disease models and models of manipulated energy supply/sensing. We aim to provide ways of thinking about metabolic remodeling that center around metabolic flexibility, capacity (reserve), and efficiency rather than around particular substrate preferences or transcriptomic profiles. We show that maladaptive metabolic remodeling takes multiple forms across multiple energy-handling domains. We suggest that lack of metabolic flexibility and reserve (substrate, oxidative, and phosphotransfer) represents a final common denominator ultimately compromising efficiency and contractile reserve in stressful contexts.
Collapse
Affiliation(s)
- Mark A Peterzan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Oliver J Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
45
|
Neuroprotective Effect of Creatine and Pyruvate on Enzyme Activities of Phosphoryl Transfer Network and Oxidative Stress Alterations Caused by Leucine Administration in Wistar Rats. Neurotox Res 2017; 32:575-584. [DOI: 10.1007/s12640-017-9762-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 05/22/2017] [Accepted: 05/26/2017] [Indexed: 01/25/2023]
|
46
|
Gupta A, Houston B. A comprehensive review of the bioenergetics of fatty acid and glucose metabolism in the healthy and failing heart in nondiabetic condition. Heart Fail Rev 2017; 22:825-842. [DOI: 10.1007/s10741-017-9623-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Kohlhaas M, Nickel AG, Bergem S, Casadei B, Laufs U, Maack C. Endogenous nitric oxide formation in cardiac myocytes does not control respiration during β-adrenergic stimulation. J Physiol 2017; 595:3781-3798. [PMID: 28229450 DOI: 10.1113/jp273750] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/16/2017] [Indexed: 01/05/2023] Open
Abstract
KEY POINTS In the heart, endothelial nitric oxide (NO) controls oxygen consumption in the working heart through paracrine mechanisms. While cardiac myocytes contain several isoforms of NO synthases, it is unclear whether these can control respiration in an intracrine fashion. A long-standing controversy is whether a NOS exists within mitochondria. By combining fluorescence technologies with electrical field stimulation or the patch-clamp technique in beating cardiac myocytes, we identified a neuronal NO synthase (nNOS) as the most relevant source of intracellular NO during β-adrenergic stimulation, while no evidence for a mitochondria-located NOS was obtained. The amounts of NO produced by non-mitochondrial nNOS were insufficient to regulate respiration during β-adrenergic stimulation, arguing against intracrine control of respiration by NO within cardiac myocytes. ABSTRACT Endothelial nitric oxide (NO) controls cardiac oxygen (O2 ) consumption in a paracrine way by slowing respiration at the mitochondrial electron transport chain. While NO synthases (NOSs) are also expressed in cardiac myocytes, it is unclear whether they control respiration in an intracrine way. Furthermore, the existence of a mitochondrial NOS is controversial. Here, by combining fluorescence imaging with electrical field stimulation, the patch-clamp method and knock-out technology, we determined the sources and consequences of intracellular NO formation during workload transitions in isolated murine and guinea pig cardiac myocytes and mitochondria. Using 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF) as a fluorescent NO-sensor that locates to the cytosol and mitochondria, we observed that NO increased by ∼12% within 3 min of β-adrenergic stimulation in beating cardiac myocytes. This NO stems from neuronal NOS (nNOS), but not endothelial (eNOS). After patch clamp-mediated dialysis of cytosolic DAF, the remaining NO signals (mostly mitochondrial) were blocked by nNOS deletion, but not by inhibiting the mitochondrial Ca2+ uniporter with Ru360. While in isolated mitochondria exogenous NO inhibited respiration and reduced the NAD(P)H redox state, pyridine nucleotide redox states were unaffected by pharmacological or genetic disruption of endogenous nNOS or eNOS during workload transitions in cardiac myoctyes. We conclude that under physiological conditions, nNOS is the most relevant source for NO in cardiac myocytes, but this nNOS is not located in mitochondria and does not control respiration. Therefore, cardiac O2 consumption is controlled by endothelial NO in a paracrine, but not intracrine, fashion.
Collapse
Affiliation(s)
- Michael Kohlhaas
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421, Homburg, Germany
| | - Alexander G Nickel
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421, Homburg, Germany
| | - Stefanie Bergem
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421, Homburg, Germany
| | - Barbara Casadei
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Ulrich Laufs
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421, Homburg, Germany
| | - Christoph Maack
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421, Homburg, Germany
| |
Collapse
|
48
|
Baldissera MD, Souza CF, Santos RC, Stefani LM, Moreira KLS, da Veiga ML, da Rocha MIU, Baldisserotto B. Pseudomonas aeruginosa strain PA01 impairs enzymes of the phosphotransfer network in the gills of Rhamdia quelen. Vet Microbiol 2017; 201:121-125. [DOI: 10.1016/j.vetmic.2017.01.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 11/16/2022]
|
49
|
Schniertshauer D, Müller S, Mayr T, Sonntag T, Gebhard D, Bergemann J. Accelerated Regeneration of ATP Level after Irradiation in Human Skin Fibroblasts by Coenzyme Q10. Photochem Photobiol 2016; 92:488-94. [PMID: 26946184 DOI: 10.1111/php.12583] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 01/15/2016] [Accepted: 02/02/2016] [Indexed: 12/29/2022]
Abstract
Human skin is exposed to a number of harmful agents of which the ultraviolet (UV) component of solar radiation is most important. UV-induced damages include direct DNA lesions as well as oxidative damage in DNA, proteins and lipids caused by reactive oxygen species (ROS). Being the main site of ROS generation in the cell, mitochondria are particularly affected by photostress. The resulting mitochondrial dysfunction may have negative effects on many essential cellular processes. To counteract these effects, coenzyme Q10 (CoQ10 ) is used as a potent therapeutic in a number of diseases. We analyzed the mitochondrial respiration profile, the mitochondrial membrane potential and cellular ATP level in skin fibroblasts after irradiation. We observed an accelerated regeneration of cellular ATP level, a decrease in mitochondrial dysfunction as well as a preservation of the mitochondrial membrane potential after irradiation in human skin fibroblasts by treatment with CoQ10 . We conclude that the faster regeneration of the ATP level was achieved by a preservation of mitochondrial function by the addition of CoQ10 and that the protective effect of CoQ10 is primarily mediated via its antioxidative function. We suggest also that it might be further dependent on a stimulation of DNA repair enzymes by CoQ10 .
Collapse
Affiliation(s)
- Daniel Schniertshauer
- Department of Life Sciences, Albstadt-Sigmaringen University of Applied Sciences, Sigmaringen, Germany
| | - Sonja Müller
- Department of Life Sciences, Albstadt-Sigmaringen University of Applied Sciences, Sigmaringen, Germany
| | - Tobias Mayr
- Department of Life Sciences, Albstadt-Sigmaringen University of Applied Sciences, Sigmaringen, Germany
| | - Tanja Sonntag
- Department of Life Sciences, Albstadt-Sigmaringen University of Applied Sciences, Sigmaringen, Germany
| | - Daniel Gebhard
- Department of Life Sciences, Albstadt-Sigmaringen University of Applied Sciences, Sigmaringen, Germany
| | - Jörg Bergemann
- Department of Life Sciences, Albstadt-Sigmaringen University of Applied Sciences, Sigmaringen, Germany
| |
Collapse
|
50
|
Relationship between pathological findings and enzymes of the energy metabolism in liver of rats infected by Trypanosoma evansi. Parasitol Int 2015; 64:547-52. [DOI: 10.1016/j.parint.2015.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/01/2015] [Accepted: 07/30/2015] [Indexed: 12/20/2022]
|