1
|
Qin X, Jin L, Gong H, Zheng Q. Electro-metabolic coupling in atrial fibrillation: A deeper understanding of the metabolic driver. Biomed Pharmacother 2024; 180:117536. [PMID: 39378681 DOI: 10.1016/j.biopha.2024.117536] [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: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024] Open
Abstract
Atrial fibrillation (AF), the most common sustained heart rhythm abnormality, disrupts the normal link between electrical activity and atrial muscle contraction; this disruption is termed "excitation-contraction uncoupling". It weakens atrial contractions and contributes to the development and persistence of AF. In addition to electrical dysfunction, AF is increasingly recognized as a metabolic disorder. Metabolic remodeling may reportedly precede electrophysiological, contractile, and structural changes in AF. Both clinical observations and experimental studies have underscored the critical importance of metabolic homeostasis, and its disturbance is considered a key initial factor in the development of AF. Research in this field has progressed, and a consensus has emerged that metabolic status (energy flux) and electrophysiological signaling (ion flux) are interactively regulated, highlighting the concept of "electro-metabolic coupling." Their uncoupling or decompensation constitutes a common pathological basis of AF. Despite growing recognition of the importance of metabolic balance, the role of electro-metabolic coupling in AF remains unclear. Thus, this review aimed to discuss 1) a comprehensive understanding of electro-metabolic alterations post-AF, 2) the pivotal role of metabolic homeostasis in AF pathogenesis, and 3) the mutual regulation of electro-metabolic signaling, along with potential therapeutic strategies targeting these imbalances.
Collapse
Affiliation(s)
- Xinghua Qin
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| | - Lingyan Jin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Haoyu Gong
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| |
Collapse
|
2
|
Kedia S, Awal NM, Seddon J, Marder E. Sulfonylurea receptor coupled conductances alter the performace of two central pattern generating circuits in Cancer borealis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602760. [PMID: 39026863 PMCID: PMC11257524 DOI: 10.1101/2024.07.09.602760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Neuronal activity and energy supply must maintain a fine balance for neuronal fitness. Various channels of communication between the two could impact network output in different ways. Sulfonylurea receptors (SURs) are a modification of ATP-binding cassette proteins (ABCs) that confer ATP-dependent gating on their associated ion channels. They are widely expressed and link metabolic states directly to neuronal activity. The role they play varies in different circuits, both enabling bursting and inhibiting activity in pathological conditions. The crab, Cancer borealis, has central patterns generators (CPGs) that fire in rhythmic bursts nearly constantly and it is unknown how energy availability influences these networks. The pyloric network of the stomatogastric ganglion (STG) and cardiac ganglion (GC) control rhythmic contractions of the foregut and heart respectively. Pharmacological manipulation of SURs results in opposite effects in the two CPGs. Neuronal firing completely stops in the STG when SUR-associated channels are open, and firing increases when the channels are closed. This results from a decrease in the excitability of pyloric dilator (PD) neurons, which are a part of the pacemaker kernel. The neurons of the CG, paradoxically, increase firing within bursts when SUR-associated channels are opened, and bursting slows when SUR-associated channels are closed. The channel permeability and sensitivities analyses present novel SUR-conductance biophysics, which nevertheless change activity in ways reminiscent of the predominantly studied mammalian receptor/channels. We suggest that SUR-associated conductances allow different neurons to respond to energy states in different ways through a common mechanism.
Collapse
Affiliation(s)
- Sonal Kedia
- Biology Department and Volen Center, Brandeis University, Waltham, MA 02454
| | - Naziru M Awal
- Biology Department and Volen Center, Brandeis University, Waltham, MA 02454
| | - Jackie Seddon
- Biology Department and Volen Center, Brandeis University, Waltham, MA 02454
| | - Eve Marder
- Biology Department and Volen Center, Brandeis University, Waltham, MA 02454
| |
Collapse
|
3
|
Harold KM, Matsuzaki S, Pranay A, Loveland BL, Batushansky A, Mendez Garcia MF, Eyster C, Stavrakis S, Chiao YA, Kinter M, Humphries KM. Loss of Cardiac PFKFB2 Drives Metabolic, Functional, and Electrophysiological Remodeling in the Heart. J Am Heart Assoc 2024; 13:e033676. [PMID: 38533937 PMCID: PMC11179765 DOI: 10.1161/jaha.123.033676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2) is a critical glycolytic regulator responsible for upregulation of glycolysis in response to insulin and adrenergic signaling. PFKFB2, the cardiac isoform of PFK-2, is degraded in the heart in the absence of insulin signaling, contributing to diabetes-induced cardiac metabolic inflexibility. However, previous studies have not examined how the loss of PFKFB2 affects global cardiac metabolism and function. METHODS AND RESULTS To address this, we have generated a mouse model with a cardiomyocyte-specific knockout of PFKFB2 (cKO). Using 9-month-old cKO and control mice, we characterized the impacts of PFKFB2 on cardiac metabolism, function, and electrophysiology. cKO mice have a shortened life span of 9 months. Metabolically, cKO mice are characterized by increased glycolytic enzyme abundance and pyruvate dehydrogenase activity, as well as decreased mitochondrial abundance and beta oxidation, suggesting a shift toward glucose metabolism. This was supported by a decrease in the ratio of palmitoyl carnitine to pyruvate-dependent mitochondrial respiration in cKO relative to control animals. Metabolomic, proteomic, and Western blot data support the activation of ancillary glucose metabolism, including pentose phosphate and hexosamine biosynthesis pathways. Physiologically, cKO animals exhibited impaired systolic function and left ventricular dilation, represented by reduced fractional shortening and increased left ventricular internal diameter, respectively. This was accompanied by electrophysiological alterations including increased QT interval and other metrics of delayed ventricular conduction. CONCLUSIONS Loss of PFKFB2 results in metabolic remodeling marked by cardiac ancillary pathway activation. This could delineate an underpinning of pathologic changes to mechanical and electrical function in the heart.
Collapse
Affiliation(s)
- Kylene M. Harold
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
- Department of Biochemistry and Molecular PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Brooke L. Loveland
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
- Ilse Katz Institute for Nanoscale Science & TechnologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Maria F. Mendez Garcia
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Craig Eyster
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Stavros Stavrakis
- Department of Medicine, Section of Cardiovascular MedicineUniversity of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
- Department of Biochemistry and Molecular PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Kenneth M. Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research FoundationOklahoma CityOKUSA
- Department of Biochemistry and Molecular PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| |
Collapse
|
4
|
Kembro JM, Flesia AG, Acosta-Rodríguez VA, Takahashi JS, Nieto PS. Dietary restriction modulates ultradian rhythms and autocorrelation properties in mice behavior. Commun Biol 2024; 7:303. [PMID: 38461321 PMCID: PMC10925031 DOI: 10.1038/s42003-024-05991-3] [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: 06/22/2023] [Accepted: 02/28/2024] [Indexed: 03/11/2024] Open
Abstract
Animal behavior emerges from integration of many processes with different spatial and temporal scales. Dynamical behavioral patterns, including daily and ultradian rhythms and the dynamical microstructure of behavior (i.e., autocorrelations properties), can be differentially affected by external cues. Identifying these patterns is important for understanding how organisms adapt to their environment, yet unbiased methods to quantify dynamical changes over multiple temporal scales are lacking. Herein, we combine a wavelet approach with Detrended Fluctuation Analysis to identify behavioral patterns and evaluate changes over 42-days in mice subjected to different dietary restriction paradigms. We show that feeding restriction alters dynamical patterns: not only are daily rhythms modulated but also the presence, phase and/or strength of ~12h-rhythms, as well as the nature of autocorrelation properties of feed-intake and wheel running behaviors. These results highlight the underlying complexity of behavioral architecture and offer insights into the multi-scale impact of feeding habits on physiology.
Collapse
Affiliation(s)
- Jackelyn Melissa Kembro
- Universidad Nacional de Córdoba (UNC), Facultad de Ciencias Exactas, Físicas y Naturales, Instituto de Ciencia y Tecnología de los Alimentos (ICTA) and Departamento de Química, Cátedra de Química Biológica, Córdoba, Córdoba, X5000HUA, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- UNC, Córdoba, Córdoba, X5000HUA, Argentina
| | - Ana Georgina Flesia
- Universidad Nacional de Córdoba, Facultad de Matemática, Astronomía, Física y Computación, Córdoba, Córdoba, X5000HUA, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones y Estudios de Matemática (CIEM, CONICET-UNC), Córdoba, Córdoba, X5000HUA, Argentina
| | - Victoria América Acosta-Rodríguez
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9111, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9111, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9111, USA
| | - Paula Sofía Nieto
- Universidad Nacional de Córdoba, Facultad de Matemática, Astronomía, Física y Computación, Córdoba, Córdoba, X5000HUA, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Física Enrique Gaviola (IFEG, CONICET-UNC), Universidad Nacional de Córdoba, Córdoba, Córdoba, X5000HUA, Argentina.
| |
Collapse
|
5
|
Harold KM, Matsuzaki S, Pranay A, Loveland BL, Batushansky A, Mendez Garcia MF, Eyster C, Stavrakis S, Chiao YA, Kinter M, Humphries KM. Loss of cardiac PFKFB2 drives Metabolic, Functional, and Electrophysiological Remodeling in the Heart. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568379. [PMID: 38045353 PMCID: PMC10690253 DOI: 10.1101/2023.11.22.568379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2) is a critical glycolytic regulator responsible for upregulation of glycolysis in response to insulin and adrenergic signaling. PFKFB2, the cardiac isoform of PFK-2, is degraded in the heart in the absence of insulin signaling, contributing to diabetes-induced cardiac metabolic inflexibility. However, previous studies have not examined how the loss of PFKFB2 affects global cardiac metabolism and function. Methods To address this, we have generated a mouse model with a cardiomyocyte-specific knockout of PFKFB2 (cKO). Using 9-month-old cKO and control (CON) mice, we characterized impacts of PFKFB2 on cardiac metabolism, function, and electrophysiology. Results cKO mice have a shortened lifespan of 9 months. Metabolically, cKO mice are characterized by increased glycolytic enzyme abundance and pyruvate dehydrogenase (PDH) activity, as well as decreased mitochondrial abundance and beta oxidation, suggesting a shift toward glucose metabolism. This was supported by a decrease in the ratio of palmitoyl carnitine to pyruvate-dependent mitochondrial respiration in cKO relative to CON animals. Metabolomic, proteomic, and western blot data support the activation of ancillary glucose metabolism, including pentose phosphate and hexosamine biosynthesis pathways. Physiologically, cKO animals exhibited impaired systolic function and left ventricular (LV) dilation, represented by reduced fractional shortening and increased LV internal diameter, respectively. This was accompanied by electrophysiological alterations including increased QT interval and other metrics of delayed ventricular conduction. Conclusions Loss of PFKFB2 results in metabolic remodeling marked by cardiac ancillary pathway activation. This could delineate an underpinning of pathologic changes to mechanical and electrical function in the heart. Clinical Perspective What is New?: We have generated a novel cardiomyocyte-specific knockout model of PFKFB2, the cardiac isoform of the primary glycolytic regulator Phosphofructokinase-2 (cKO).The cKO model demonstrates that loss of cardiac PFKFB2 drives metabolic reprogramming and shunting of glucose metabolites to ancillary metabolic pathways.The loss of cardiac PFKFB2 promotes electrophysiological and functional remodeling in the cKO heart.What are the Clinical Implications?: PFKFB2 is degraded in the absence of insulin signaling, making its loss particularly relevant to diabetes and the pathophysiology of diabetic cardiomyopathy.Changes which we observe in the cKO model are consistent with those often observed in diabetes and heart failure of other etiologies.Defining PFKFB2 loss as a driver of cardiac pathogenesis identifies it as a target for future investigation and potential therapeutic intervention.
Collapse
|
6
|
Ghosheh M, Ehrlich A, Ioannidis K, Ayyash M, Goldfracht I, Cohen M, Fischer A, Mintz Y, Gepstein L, Nahmias Y. Electro-metabolic coupling in multi-chambered vascularized human cardiac organoids. Nat Biomed Eng 2023; 7:1493-1513. [PMID: 37550423 DOI: 10.1038/s41551-023-01071-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 06/27/2023] [Indexed: 08/09/2023]
Abstract
The study of cardiac physiology is hindered by physiological differences between humans and small-animal models. Here we report the generation of multi-chambered self-paced vascularized human cardiac organoids formed under anisotropic stress and their applicability to the study of cardiac arrhythmia. Sensors embedded in the cardiac organoids enabled the simultaneous measurement of oxygen uptake, extracellular field potentials and cardiac contraction at resolutions higher than 10 Hz. This microphysiological system revealed 1 Hz cardiac respiratory cycles that are coupled to the electrical rather than the mechanical activity of cardiomyocytes. This electro-mitochondrial coupling was driven by mitochondrial calcium oscillations driving respiration cycles. Pharmaceutical or genetic inhibition of this coupling results in arrhythmogenic behaviour. We show that the chemotherapeutic mitoxantrone induces arrhythmia through disruption of this pathway, a process that can be partially reversed by the co-administration of metformin. Our microphysiological cardiac systems may further facilitate the study of the mitochondrial dynamics of cardiac rhythms and advance our understanding of human cardiac physiology.
Collapse
Affiliation(s)
- Mohammad Ghosheh
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avner Ehrlich
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Tissue Dynamics, LTD, Jerusalem, Israel
| | - Konstantinos Ioannidis
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Tissue Dynamics, LTD, Jerusalem, Israel
| | - Muneef Ayyash
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Idit Goldfracht
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
| | - Merav Cohen
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amit Fischer
- Department of Biological Chemistry, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yoav Mintz
- Department of General Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lior Gepstein
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
- Cardiology Department, Rambam Health Care Campus, Haifa, Israel
| | - Yaakov Nahmias
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Tissue Dynamics, LTD, Jerusalem, Israel.
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
7
|
Hoces D, Miguens Blanco J, Hernández-López RA. A synthetic biology approach to engineering circuits in immune cells. Immunol Rev 2023; 320:120-137. [PMID: 37464881 DOI: 10.1111/imr.13244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
A synthetic circuit in a biological system involves the designed assembly of genetic elements, biomolecules, or cells to create a defined function. These circuits are central in synthetic biology, enabling the reprogramming of cellular behavior and the engineering of cells with customized responses. In cancer therapeutics, engineering T cells with circuits have the potential to overcome the challenges of current approaches, for example, by allowing specific recognition and killing of cancer cells. Recent advances also facilitate engineering integrated circuits for the controlled release of therapeutic molecules at specified locations, for example, in a solid tumor. In this review, we discuss recent strategies and applications of synthetic receptor circuits aimed at enhancing immune cell functions for cancer immunotherapy. We begin by introducing the concept of circuits in networks at the molecular and cellular scales and provide an analysis of the development and implementation of several synthetic circuits in T cells that have the goal to overcome current challenges in cancer immunotherapy. These include specific targeting of cancer cells, increased T-cell proliferation, and persistence in the tumor microenvironment. By harnessing the power of synthetic biology, and the characteristics of certain circuit architectures, it is now possible to engineer a new generation of immune cells that recognize cancer cells, while minimizing off-target toxicities. We specifically discuss T-cell circuits for antigen density sensing. These circuits allow targeting of solid tumors that share antigens with normal tissues. Additionally, we explore designs for synthetic circuits that could control T-cell differentiation or T-cell fate as well as the concept of synthetic multicellular circuits that leverage cellular communication and division of labor to achieve improved therapeutic efficacy. As our understanding of cell biology expands and novel tools for genome, protein, and cell engineering are developed, we anticipate further innovative approaches to emerge in the design and engineering of circuits in immune cells.
Collapse
Affiliation(s)
- Daniel Hoces
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Jesús Miguens Blanco
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Rogelio A Hernández-López
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford, California, USA
- Chan-Zuckerberg Biohub-San Francisco, San Francisco, California, USA
| |
Collapse
|
8
|
Xiong LI, Garfinkel A. Are physiological oscillations physiological? J Physiol 2023. [PMID: 37622389 DOI: 10.1113/jp285015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
Despite widespread and striking examples of physiological oscillations, their functional role is often unclear. Even glycolysis, the paradigm example of oscillatory biochemistry, has seen questions about its oscillatory function. Here, we take a systems approach to argue that oscillations play critical physiological roles, such as enabling systems to avoid desensitization, to avoid chronically high and therefore toxic levels of chemicals, and to become more resistant to noise. Oscillation also enables complex physiological systems to reconcile incompatible conditions such as oxidation and reduction, by cycling between them, and to synchronize the oscillations of many small units into one large effect. In pancreatic β-cells, glycolytic oscillations synchronize with calcium and mitochondrial oscillations to drive pulsatile insulin release, critical for liver regulation of glucose. In addition, oscillation can keep biological time, essential for embryonic development in promoting cell diversity and pattern formation. The functional importance of oscillatory processes requires a re-thinking of the traditional doctrine of homeostasis, holding that physiological quantities are maintained at constant equilibrium values, a view that has largely failed in the clinic. A more dynamic approach will initiate a paradigm shift in our view of health and disease. A deeper look into the mechanisms that create, sustain and abolish oscillatory processes requires the language of nonlinear dynamics, well beyond the linearization techniques of equilibrium control theory. Nonlinear dynamics enables us to identify oscillatory ('pacemaking') mechanisms at the cellular, tissue and system levels.
Collapse
Affiliation(s)
- Lingyun Ivy Xiong
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Alan Garfinkel
- Departments of Medicine (Cardiology) and Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| |
Collapse
|
9
|
Kurz FT, Aon MA, Schlemmer HP, Jende JME, O'Rourke B, Armoundas AA. Fractal dynamics of individual mitochondrial oscillators measure local inter-mitochondrial coupling. Biophys J 2023; 122:1459-1469. [PMID: 36905121 PMCID: PMC10147834 DOI: 10.1016/j.bpj.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/29/2022] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Mitochondrial inner membrane potentials in cardiomyocytes may oscillate in cycles of depolarization/repolarization when the mitochondrial network is exposed to metabolic or oxidative stress. The frequencies of such oscillations are dynamically changing while clusters of weakly coupled mitochondrial oscillators adjust to a common phase and frequency. Across the cardiac myocyte, the averaged signal of the mitochondrial population follows self-similar or fractal dynamics; however, fractal properties of individual mitochondrial oscillators have not yet been examined. We show that the largest synchronously oscillating cluster exhibits a fractal dimension, D, that is indicative of self-similar behavior with D=1.27±0.11, in contrast to the remaining network mitochondria whose fractal dimension is close to that of Brownian noise, D=1.58±0.10. We further demonstrate that fractal behavior is correlated with local coupling mechanisms, whereas it is only weakly linked to measures of functional connections between mitochondria. Our findings suggest that individual mitochondrial fractal dimensions may serve as a simple measure of local mitochondrial coupling.
Collapse
Affiliation(s)
- Felix T Kurz
- German Cancer Research Center, Division of Radiology, Heidelberg, Germany; Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, Massachusetts.
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | | | - Johann M E Jende
- Heidelberg University Hospital, Department of Neuroradiology, Heidelberg, Germany
| | - Brian O'Rourke
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, Maryland
| | - Antonis A Armoundas
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, Massachusetts; Broad Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
10
|
Chakravarty S, Hong CI, Csikász-Nagy A. Systematic analysis of negative and positive feedback loops for robustness and temperature compensation in circadian rhythms. NPJ Syst Biol Appl 2023; 9:5. [PMID: 36774353 PMCID: PMC9922291 DOI: 10.1038/s41540-023-00268-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/30/2023] [Indexed: 02/13/2023] Open
Abstract
Temperature compensation and robustness to biological noise are two key characteristics of the circadian clock. These features allow the circadian pacemaker to maintain a steady oscillation in a wide range of environmental conditions. The presence of a time-delayed negative feedback loop in the regulatory network generates autonomous circadian oscillations in eukaryotic systems. In comparison, the circadian clock of cyanobacteria is controlled by a strong positive feedback loop. Positive feedback loops with substrate depletion can also generate oscillations, inspiring other circadian clock models. What makes a circadian oscillatory network robust to extrinsic noise is unclear. We investigated four basic circadian oscillators with negative, positive, and combinations of positive and negative feedback loops to explore network features necessary for circadian clock resilience. We discovered that the negative feedback loop system performs the best in compensating temperature changes. We also show that a positive feedback loop can reduce extrinsic noise in periods of circadian oscillators, while intrinsic noise is reduced by negative feedback loops.
Collapse
Affiliation(s)
- Suchana Chakravarty
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Christian I Hong
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Attila Csikász-Nagy
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
| |
Collapse
|
11
|
Kumpost V, Hilbert L, Mikut R. Noise facilitates entrainment of a population of uncoupled limit cycle oscillators. J R Soc Interface 2023; 20:20220781. [PMID: 36628527 PMCID: PMC9832296 DOI: 10.1098/rsif.2022.0781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Many biological oscillators share two properties: they are subject to stochastic fluctuations (noise) and they must reliably adjust their period to changing environmental conditions (entrainment). While noise seems to distort the ability of single oscillators to entrain, in populations of uncoupled oscillators noise allows population-level entrainment for a wider range of input amplitudes and periods. Here, we investigate how this effect depends on the noise intensity and the number of oscillators in the population. We have found that, if a population consists of a sufficient number of oscillators, increasing noise intensity leads to faster entrainment after a phase change of the input signal (jet lag) and increases sensitivity to low-amplitude input signals.
Collapse
Affiliation(s)
- Vojtech Kumpost
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- Institute of Biological and Chemical Systems—Biological Information Processing, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Lennart Hilbert
- Institute of Biological and Chemical Systems—Biological Information Processing, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- Department of Systems Biology and Bioinformatics, Zoological Institute, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
12
|
Khmelinskii I, Makarov VI. IR exciton activation mechanism of ethanol oxidation by human alcohol dehydrogenase (ADH) 1A enzyme. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2022.114371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
13
|
Li RJ, Xu JJ, Zhang ZH, Chen MW, Liu SX, Yang C, Li YL, Luo P, Liu YJ, Tang R, Shan ZG. Rhein ameliorates transverse aortic constriction-induced cardiac hypertrophy via regulating STAT3 and p38 MAPK signaling pathways. Front Pharmacol 2022; 13:940574. [PMID: 36091816 PMCID: PMC9459036 DOI: 10.3389/fphar.2022.940574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/07/2022] [Indexed: 11/28/2022] Open
Abstract
The progression from compensatory hypertrophy to heart failure is difficult to reverse, in part due to extracellular matrix fibrosis and continuous activation of abnormal signaling pathways. Although the anthraquinone rhein has been examined for its many biological properties, it is not clear whether it has therapeutic value in the treatment of cardiac hypertrophy and heart failure. In this study, we report for the first time that rhein can ameliorate transverse aortic constriction (TAC)-induced cardiac hypertrophy and other cardiac damage in vivo and in vitro. In addition, rhein can reduce cardiac hypertrophy by attenuating atrial natriuretic peptide, brain natriuretic peptide, and β-MHC expression; cardiac fibrosis; and ERK phosphorylation and transport into the nucleus. Furthermore, the inhibitory effect of rhein on myocardial hypertrophy was similar to that of specific inhibitors of STAT3 and ERK signaling. In addition, rhein at therapeutic doses had no significant adverse effects or toxicity on liver and kidney function. We conclude that rhein reduces TAC-induced cardiac hypertrophy via targeted inhibition of the molecular function of ERK and downregulates STAT3 and p38 MAPK signaling. Therefore, rhein might be a novel and effective agent for treating cardiac hypertrophy and other cardiovascular diseases.
Collapse
Affiliation(s)
- Run-Jing Li
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jia-Jia Xu
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zheng-Hao Zhang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Min-Wei Chen
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shi-Xiao Liu
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Cui Yang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yan-Ling Li
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ping Luo
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yi-Jiang Liu
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rong Tang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Rong Tang, ; Zhong-Gui Shan,
| | - Zhong-Gui Shan
- Department of Cardiac Surgery, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Rong Tang, ; Zhong-Gui Shan,
| |
Collapse
|
14
|
Reilly L, Munawar S, Zhang J, Crone WC, Eckhardt LL. Challenges and innovation: Disease modeling using human-induced pluripotent stem cell-derived cardiomyocytes. Front Cardiovasc Med 2022; 9:966094. [PMID: 36035948 PMCID: PMC9411865 DOI: 10.3389/fcvm.2022.966094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
Disease modeling using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has both challenges and promise. While patient-derived iPSC-CMs provide a unique opportunity for disease modeling with isogenic cells, the challenge is that these cells still demonstrate distinct properties which make it functionally less akin to adult cardiomyocytes. In response to this challenge, numerous innovations in differentiation and modification of hiPSC-CMs and culture techniques have been developed. Here, we provide a focused commentary on hiPSC-CMs for use in disease modeling, the progress made in generating electrically and metabolically mature hiPSC-CMs and enabling investigative platforms. The solutions are bringing us closer to the promise of modeling heart disease using human cells in vitro.
Collapse
Affiliation(s)
- Louise Reilly
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Saba Munawar
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Jianhua Zhang
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Wendy C. Crone
- Department of Engineering Physics, College of Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Lee L. Eckhardt
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States,*Correspondence: Lee L. Eckhardt
| |
Collapse
|
15
|
Khmelinskii I, Makarov VI. Theoretical approaches used in the modelling of reversible and irreversible mitochondrial swelling in vitro. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 172:15-23. [PMID: 35447196 DOI: 10.1016/j.pbiomolbio.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/25/2022] [Accepted: 04/11/2022] [Indexed: 06/14/2023]
Abstract
Existing theoretical approaches were considered that allow modelling of mitochondrial swelling (MS) dynamics. Simple phenomenological kinetic models were reviewed. Simple and extended biophysical and bioenergetic models that ignore mechanical properties of inner mitochondrial membrane (IMM), and similar models that include these mechanical properties were also reviewed. Limitations of these models we considered, as regards correct modelling of MS dynamics. It was found that simple phenomenological kinetic models have significant limitations, due to dependence of the kinetic parameter values estimated by fitting of the experimental data on the experimental conditions. Additionally, such simple models provide no understanding of the detailed mechanisms behind the MS dynamics, nor of the dynamics of various system parameters during MS. Thus, biophysical and bioenergetic models ignoring IMM mechanical properties can't be used to model the transition between reversible and irreversible MS. However, simple and extended biophysical models that include IMM mechanical properties allow modelling the transition to irreversible swelling. These latter models are still limited due to significantly simplified description of biochemistry, compared to those of bioenergetic models. Finally, a strategy of model development is proposed, towards correct interpretation of the mitochondrial life cycle, including the effects of MS dynamics.
Collapse
Affiliation(s)
- Igor Khmelinskii
- Universidade do Algarve, FCT, DQB and CEOT, 8005-139, Faro, Portugal
| | - Vladimir I Makarov
- University of Puerto Rico, Rio Piedras Campus, PO Box 23343, San Juan, PR, 00931-3343, USA.
| |
Collapse
|
16
|
Khmelinskii I, Makarov V. Theoretical analysis of reversible and irreversible mitochondrial swelling in vivo. Biosystems 2022; 217:104679. [DOI: 10.1016/j.biosystems.2022.104679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/06/2022] [Indexed: 11/02/2022]
|
17
|
Khmelinskii I, Makarov VI. Photo-activation of mitochondrial ATP synthesis. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 228:112376. [PMID: 35121525 DOI: 10.1016/j.jphotobiol.2021.112376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
ATP production by mitochondria isolated from Saccharomyces cerevisiae cells was accelerated upon both direct and indirect mitochondrial photo-activation (MPA). The extent of direct MPA was dependent on the wavelength of excitation light. Direct MPA was created by light in cytochrome c spectral absorption bands (440, 520 and 550 nm), this light was absorbed producing electronically excited cytochrome c, and the excitation energy of the latter was used in the ATP production chain. The activity of cytochrome c was tested with 600 nm light, where cytochrome c does not absorb, and thus ATP production rate remained the same as in darkness. Note that ATP production rates were significantly larger under light at 550, 520 and 440 nm. Therefore, photo-activation of cytochrome c was the first step of MPA synthesis of ATP. Indirect MPA of ATP production also proceeded via electronically excited cytochrome c, by energy transfer from electronically excited Co/BN film to cytochrome c located in the inner mitochondrial membrane (IMM). Co/BN excitons were generated by photons absorbed by the Co/BN film, which was not in contact with the mitochondrial sample. Next, these excitons propagated along the Co/BN film to the part of the film that was in contact with the mitochondrial sample. There the exciton energy was transferred to cytochrome c located in the IMM, producing electronically excited cytochrome c. Thus, excited cytochrome c was generated in a way different from that of direct MPA. Next, the energy of excited cytochrome c was used in activated ATP synthesis, with virtually the same effect for 519 and 427 nm excitation. Thus, the first step of ATP synthesis in indirect MPA was the exciton energy transfer from Co/BN film to cytochrome c located in the IMM, producing an electronically excited cytochrome c molecule. A phenomenological mechanism of direct and indirect MPA was proposed, and the model parameters were obtained by fitting the model to the experimental data. However, more information is needed before the detailed mechanism of ATP synthesis activation by electronically excited cytochrome c could be understood. The present results support the earlier proposed hypothesis of indirect MPA of ATP production in vertebrate retina in daylight.
Collapse
Affiliation(s)
- Igor Khmelinskii
- Faculty of Science and Technology, Department of Chemistry and Pharmacy, and Center of Electronics, Optoelectronics, and Telecommunications, University of Algarve, Portugal
| | - Vladimir I Makarov
- Department of Physics, University of Puerto Rico, Rio Piedras Campus, San Juan, USA.
| |
Collapse
|
18
|
Khmelinskii I, Makarov VI. Mitochondrial ATP Synthesis Activated by Exciton Energy Transfer from Müller cell Intermediate Filaments. Chem Phys 2022. [DOI: 10.1016/j.chemphys.2022.111475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
19
|
Khmelinskii I, Makarov V. Reversible and irreversible mitochondrial swelling: Effects of variable mitochondrial activity. Biosystems 2021; 210:104559. [PMID: 34627969 DOI: 10.1016/j.biosystems.2021.104559] [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: 08/23/2021] [Revised: 10/03/2021] [Accepted: 10/03/2021] [Indexed: 10/20/2022]
Abstract
An extended biophysical model was obtained by upgrading the previously reported one (Khmelinskii and Makarov, 2021). The upgraded model accommodates variations of solute transport rates through the inner mitochondrial membrane (IMM) within the mitochondrial population, described by a Gaussian distribution. However, the model may be used for any functional form of the distribution. The dynamics of system parameters as predicted by the current model differed from that predicted by the previous model in the same initial conditions (Khmelinskii and Makarov, 2021). The amount of change varied from one parameter to the other, remaining in the 1-38% range. The upgraded model fitted the available experimental data with a better accuracy (R = 0.993) compared to the previous model (R = 0.978) using the same experimental data (Khmelinskii and Makarov, 2021). The fitting procedure also estimated the Gaussian distribution parameters. The new model requires much larger computational resources, but given its higher accuracy, it may be used for better analysis of experimental data and for better prediction of MS dynamics in different initial conditions. Note that activities of individual mitochondria in mitochondrial populations should vary within biological tissues. Thus, the currently upgraded model is a better tool for biological and bio-medical applications. We believe that this model is much better adapted to the analysis of MS dynamics in vivo.
Collapse
Affiliation(s)
- Igor Khmelinskii
- Universidade do Algarve, FCT, DQB and CEOT, 8005-139, Faro, Portugal
| | - Vladimir Makarov
- University of Puerto Rico, Rio Piedras Campus, PO Box 23343, San Juan, PR, 00931-3343, USA.
| |
Collapse
|
20
|
Khmelinskii I, Makarov V. Reversible and irreversible mitochondrial swelling in vitro. Biophys Chem 2021; 278:106668. [PMID: 34418677 DOI: 10.1016/j.bpc.2021.106668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 11/18/2022]
Abstract
Mitochondrial activity as regards ATP production strongly depends on mitochondrial swelling (MS) mode. Therefore, this work analyzes reversible and irreversible MS using a detailed biophysical model. The reported model includes mechanical properties of the inner mitochondrial membrane (IMM). The model describes MS dynamics for spherically symmetric, axisymmetric ellipsoidal and general ellipsoidal mitochondria. Mechanical stretching properties of the IMM were described by a second-rank rigidity tensor. The tensor components were estimated by fitting to the earlier reported results of in vitro experiments. The IMM rigidity constant of ca. 0.008 dyn/nm was obtained for linear deformations. The model also included membrane bending effects, which were small compared to those of membrane stretching. The model was also tested by simulation of the earlier reported experimental data and of the system dynamics at different initial conditions, predicting the system behavior. The transition criteria from reversible to irreversible swelling were determined and tested. The presently developed model is applicable directly to the analysis of in vitro experimental data, while additional improvements are necessary before it could be used to describe mitochondrial swelling in vivo. The reported theoretical model also provides an idea of physically consistent mechanism for the permeability transport pore (PTP) opening, which depends on the IMM stretching stress. In the current study, this idea is discussed briefly, but a detailed theoretical analysis of these ideas will be performed later. The currently developed model provides new understanding of the detailed MS mechanism and of the conditions for the transition between reversible and irreversible MS modes. On the other hand, the current model provides useful mathematical tools, that may be successfully used in mitochondrial biophysics research, and also in other applications, predicting the behavior of mitochondria in different conditions of the surrounding media in vitro or cellular cyto(sarco)plasm in vivo. These mathematical tools are based on real biophysical processes occurring in mitochondria. Thus, we note a significant progress in the theoretical approach, which may be used in real biological systems, compared to the earlier reported models. Significance of this study derives from inclusion of IMM mechanical properties, which directly impact the reversible and irreversible mitochondrial swelling dynamics. Reversible swelling corresponds to reversible IMM deformations, while irreversible swelling corresponds to irreversible deformations, with eventual membrane disruption. The IMM mechanical properties are directly dependent on the membrane biochemical composition and structure. The IMM deformationas are induced by osmotic pressure created by the ionic/neutral solute imbalance between the mitochondrial matrix media and the bulk solution in vitro, or cyto(sarco)plasm in vivo. The novelty of the reported model is in the biophysical mechanism detailing ionic and neutral solute transport for a large number of solutes, which were not taken into account in the earlier reported biophysical models of MS. Therefore, the reported model allows understanding response of mitochondria to the changes of initial concentration(s) of any of the solute(s) included in the model. Note that the values of all of the model parameters and kinetic constants have been estimated and the resulting complete model may be used for quantitative analysis of mitochondrial swelling dynamics in conditions of real in vitro experiments.
Collapse
Affiliation(s)
- Igor Khmelinskii
- Universidade do Algarve, FCT, DQB and CEOT, 8005-139 Faro, Portugal
| | - Vladimir Makarov
- University of Puerto Rico, Rio Piedras Campus, PO Box 23343, San Juan, PR 00931-3343, USA.
| |
Collapse
|
21
|
Energy transfer along Müller cell intermediate filaments isolated from porcine retina: II. Excitons at 2500 cm−1 produced by ADH1A upon hydrolysis of one ATP molecule. Chem Phys Lett 2021. [DOI: 10.1016/j.cplett.2021.138651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
MitoWave: Spatiotemporal analysis of mitochondrial membrane potential fluctuations during I/R. Biophys J 2021; 120:3261-3271. [PMID: 34297964 DOI: 10.1016/j.bpj.2021.05.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 03/26/2021] [Accepted: 05/06/2021] [Indexed: 11/21/2022] Open
Abstract
Mitochondria exhibit unstable inner membrane potentials (ΔΨm) when subjected to stress, such as during ischemia/reperfusion (I/R). Understanding the mechanism of ΔΨm instability involves characterizing and quantifying this phenomenon in an unbiased and reproducible manner. Here, we describe a simple analytical workflow called "MitoWave" that combines wavelet transform methods and image segmentation to unravel dynamic ΔΨm changes in the cardiac mitochondrial network during I/R. In vitro ischemia was affected by placing a glass coverslip on a monolayer of neonatal mouse ventricular myocytes for 1 h and removing the coverslip to allow for reperfusion, revealing complex oscillatory ΔΨm. MitoWave analysis was then used to identify individual mitochondrial clusters within the cells and track their intrinsic oscillation frequencies over the course of reperfusion. Responses segregated into five typical behaviors were quantified by MitoWave that were corroborated by visual inspection of the time series. Statistical analysis of the distribution of oscillating mitochondrial clusters during reperfusion showed significant differences between the five different outcomes. Features such as the time point of ΔΨm depolarization during I/R, area of mitochondrial clusters, and time-resolved frequency components during reperfusion were determined per cell and per mitochondrial cluster. Mitochondria from neonatal mouse ventricular myocytes subjected to I/R oscillate in the frequency range of 8.6-45 mHz, with a mean of 8.73 ± 4.35 mHz. Oscillating clusters had smaller areas ranging from 49.8 ± 1.2 μm2, whereas nonoscillating clusters had larger areas 66 ± 1.5 μm2. A negative correlation between frequency and mitochondrial cluster area was observed. We also observed that late ΔΨm loss during ischemia correlated with early ΔΨm stabilization after oscillation on reperfusion. Thus, MitoWave analysis provides a semiautomated method to quantify complex time-resolved mitochondrial behavior in an easy-to-follow workflow, enabling unbiased, reproducible quantitation of complex nonstationary cellular phenomena.
Collapse
|
23
|
Heltberg ML, Krishna S, Kadanoff LP, Jensen MH. A tale of two rhythms: Locked clocks and chaos in biology. Cell Syst 2021; 12:291-303. [PMID: 33887201 DOI: 10.1016/j.cels.2021.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/19/2021] [Accepted: 03/17/2021] [Indexed: 12/16/2022]
Abstract
The fundamental mechanisms that control and regulate biological organisms exhibit a surprising level of complexity. Oscillators are perhaps the simplest motifs that produce time-varying dynamics and are ubiquitous in biological systems. It is also known that such biological oscillators interact with each other-for instance, circadian oscillators affect the cell cycle, and somitogenesis clock proteins in adjacent cells affect each other in developing embryos. Therefore, it is vital to understand the effects that can emerge from non-linear interaction between oscillations. Here, we show how oscillations typically arise in biology and take the reader on a tour through the great variety in dynamics that can emerge even from a single pair of coupled oscillators. We explain how chaotic dynamics can emerge and outline the methods of detecting this in experimental time traces. Finally, we discuss the potential role of such complex dynamical features in biological systems.
Collapse
Affiliation(s)
- Mathias L Heltberg
- Niels Bohr Institute, University of Copenhagen, 2100 Copenhagen, Denmark; Laboratoire de Physique Théorique, Ecole Normale Supérieure, 75 231 Paris Cedex 05, France
| | - Sandeep Krishna
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences TIFR, GKVK Campus, Bellary Road, Bangalore 560065, India
| | - Leo P Kadanoff
- The James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Mogens H Jensen
- Niels Bohr Institute, University of Copenhagen, 2100 Copenhagen, Denmark; The James Franck Institute, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
24
|
Garbern JC, Lee RT. Mitochondria and metabolic transitions in cardiomyocytes: lessons from development for stem cell-derived cardiomyocytes. Stem Cell Res Ther 2021; 12:177. [PMID: 33712058 PMCID: PMC7953594 DOI: 10.1186/s13287-021-02252-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/28/2021] [Indexed: 12/13/2022] Open
Abstract
Current methods to differentiate cardiomyocytes from human pluripotent stem cells (PSCs) inadequately recapitulate complete development and result in PSC-derived cardiomyocytes (PSC-CMs) with an immature or fetal-like phenotype. Embryonic and fetal development are highly dynamic periods during which the developing embryo or fetus is exposed to changing nutrient, oxygen, and hormone levels until birth. It is becoming increasingly apparent that these metabolic changes initiate developmental processes to mature cardiomyocytes. Mitochondria are central to these changes, responding to these metabolic changes and transitioning from small, fragmented mitochondria to large organelles capable of producing enough ATP to support the contractile function of the heart. These changes in mitochondria may not simply be a response to cardiomyocyte maturation; the metabolic signals that occur throughout development may actually be central to the maturation process in cardiomyocytes. Here, we review methods to enhance maturation of PSC-CMs and highlight evidence from development indicating the key roles that mitochondria play during cardiomyocyte maturation. We evaluate metabolic transitions that occur during development and how these affect molecular nutrient sensors, discuss how regulation of nutrient sensing pathways affect mitochondrial dynamics and function, and explore how changes in mitochondrial function can affect metabolite production, the cell cycle, and epigenetics to influence maturation of cardiomyocytes.
Collapse
Affiliation(s)
- Jessica C Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA, 02138, USA
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA, 02138, USA.
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
| |
Collapse
|
25
|
Rowland Adams J, Stefanovska A. Modeling Cell Energy Metabolism as Weighted Networks of Non-autonomous Oscillators. Front Physiol 2021; 11:613183. [PMID: 33584336 PMCID: PMC7876325 DOI: 10.3389/fphys.2020.613183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022] Open
Abstract
Networks of oscillating processes are a common occurrence in living systems. This is as true as anywhere in the energy metabolism of individual cells. Exchanges of molecules and common regulation operate throughout the metabolic processes of glycolysis and oxidative phosphorylation, making the consideration of each of these as a network a natural step. Oscillations are similarly ubiquitous within these processes, and the frequencies of these oscillations are never truly constant. These features make this system an ideal example with which to discuss an alternative approach to modeling living systems, which focuses on their thermodynamically open, oscillating, non-linear and non-autonomous nature. We implement this approach in developing a model of non-autonomous Kuramoto oscillators in two all-to-all weighted networks coupled to one another, and themselves driven by non-autonomous oscillators. Each component represents a metabolic process, the networks acting as the glycolytic and oxidative phosphorylative processes, and the drivers as glucose and oxygen supply. We analyse the effect of these features on the synchronization dynamics within the model, and present a comparison between this model, experimental data on the glycolysis of HeLa cells, and a comparatively mainstream model of this experiment. In the former, we find that the introduction of oscillator networks significantly increases the proportion of the model's parameter space that features some form of synchronization, indicating a greater ability of the processes to resist external perturbations, a crucial behavior in biological settings. For the latter, we analyse the oscillations of the experiment, finding a characteristic frequency of 0.01–0.02 Hz. We further demonstrate that an output of the model comparable to the measurements of the experiment oscillates in a manner similar to the measured data, achieving this with fewer parameters and greater flexibility than the comparable model.
Collapse
|
26
|
Rogov AG, Goleva TN, Epremyan KK, Kireev II, Zvyagilskaya RA. Propagation of Mitochondria-Derived Reactive Oxygen Species within the Dipodascus magnusii Cells. Antioxidants (Basel) 2021; 10:antiox10010120. [PMID: 33467672 PMCID: PMC7830518 DOI: 10.3390/antiox10010120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/04/2022] Open
Abstract
Mitochondria are considered to be the main source of reactive oxygen species (ROS) in the cell. It was shown that in cardiac myocytes exposed to excessive oxidative stress, ROS-induced ROS release is triggered. However, cardiac myocytes have a network of densely packed organelles that do not move, which is not typical for the majority of eukaryotic cells. The purpose of this study was to trace the spatiotemporal development (propagation) of prooxidant-induced oxidative stress and its interplay with mitochondrial dynamics. We used Dipodascus magnusii yeast cells as a model, as they have advantages over other models, including a uniquely large size, mitochondria that are easy to visualize and freely moving, an ability to vigorously grow on well-defined low-cost substrates, and high responsibility. It was shown that prooxidant-induced oxidative stress was initiated in mitochondria, far preceding the appearance of generalized oxidative stress in the whole cell. For yeasts, these findings were obtained for the first time. Preincubation of yeast cells with SkQ1, a mitochondria-addressed antioxidant, substantially diminished production of mitochondrial ROS, while only slightly alleviating the generalized oxidative stress. This was expected, but had not yet been shown. Importantly, mitochondrial fragmentation was found to be primarily induced by mitochondrial ROS preceding the generalized oxidative stress development.
Collapse
Affiliation(s)
- Anton G. Rogov
- Bach Institute of Biochemistry, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences 33, bld. 2 Leninsky Ave., Moscow 119071, Russia; (A.G.R.); (T.N.G.); (K.K.E.)
| | - Tatiana N. Goleva
- Bach Institute of Biochemistry, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences 33, bld. 2 Leninsky Ave., Moscow 119071, Russia; (A.G.R.); (T.N.G.); (K.K.E.)
| | - Khoren K. Epremyan
- Bach Institute of Biochemistry, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences 33, bld. 2 Leninsky Ave., Moscow 119071, Russia; (A.G.R.); (T.N.G.); (K.K.E.)
| | - Igor I. Kireev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Vorobyevy Gory 1, Moscow 119992, Russia;
| | - Renata A. Zvyagilskaya
- Bach Institute of Biochemistry, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences 33, bld. 2 Leninsky Ave., Moscow 119071, Russia; (A.G.R.); (T.N.G.); (K.K.E.)
- Correspondence:
| |
Collapse
|
27
|
Mason FE, Pronto JRD, Alhussini K, Maack C, Voigt N. Cellular and mitochondrial mechanisms of atrial fibrillation. Basic Res Cardiol 2020; 115:72. [PMID: 33258071 PMCID: PMC7704501 DOI: 10.1007/s00395-020-00827-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/26/2020] [Indexed: 11/06/2022]
Abstract
The molecular mechanisms underlying atrial fibrillation (AF), the most common form of arrhythmia, are poorly understood and therefore target-specific treatment options remain an unmet clinical need. Excitation–contraction coupling in cardiac myocytes requires high amounts of adenosine triphosphate (ATP), which is replenished by oxidative phosphorylation in mitochondria. Calcium (Ca2+) is a key regulator of mitochondrial function by stimulating the Krebs cycle, which produces nicotinamide adenine dinucleotide for ATP production at the electron transport chain and nicotinamide adenine dinucleotide phosphate for the elimination of reactive oxygen species (ROS). While it is now well established that mitochondrial dysfunction plays an important role in the pathophysiology of heart failure, this has been less investigated in atrial myocytes in AF. Considering the high prevalence of AF, investigating the role of mitochondria in this disease may guide the path towards new therapeutic targets. In this review, we discuss the importance of mitochondrial Ca2+ handling in regulating ATP production and mitochondrial ROS emission and how alterations, particularly in these aspects of mitochondrial activity, may play a role in AF. In addition to describing research advances, we highlight areas in which further studies are required to elucidate the role of mitochondria in AF.
Collapse
Affiliation(s)
- Fleur E Mason
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery, University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center Würzburg, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany. .,Department of Internal Medicine I, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
28
|
Medina MÁ. Metabolic Reprogramming is a Hallmark of Metabolism Itself. Bioessays 2020; 42:e2000058. [PMID: 32939776 DOI: 10.1002/bies.202000058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/13/2020] [Indexed: 12/16/2022]
Abstract
The reprogramming of metabolism has been identified as one of the hallmarks of cancer. It is becoming more and more frequent to connect other diseases with metabolic reprogramming. This article aims to argue that metabolic reprogramming is not driven by disease but instead is the main hallmark of metabolism, based on its dynamic behavior that allows it to continuously adapt to changes in the internal and external conditions.
Collapse
Affiliation(s)
- Miguel Ángel Medina
- Andalucía Tech, Facultad de Ciencias, Departamento de Biología Molecular y Bioquímica, and IBIMA (Biomedical Research Institute of Málaga), Universidad de Málaga, Málaga, E-29071, Spain.,CIBER de Enfermedades Raras (CIBERER), Málaga, E-29071, Spain
| |
Collapse
|
29
|
Adameova A, Shah AK, Dhalla NS. Role of Oxidative Stress in the Genesis of Ventricular Arrhythmias. Int J Mol Sci 2020; 21:E4200. [PMID: 32545595 PMCID: PMC7349053 DOI: 10.3390/ijms21124200] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Ventricular arrhythmias, mainly lethal arrhythmias, such as ventricular tachycardia and fibrillation, may lead to sudden cardiac death. These are triggered as a result of cardiac injury due to chronic ischemia, acute myocardial infarction and various stressful conditions associated with increased levels of circulating catecholamines and angiotensin II. Several mechanisms have been proposed to underlie electrical instability of the heart promoting ventricular arrhythmias; however, oxidative stress which adversely affects ion homeostasis due to changes in the ion channel structure and function, seems to play a critical role in eliciting different types of ventricular arrhythmias. Prevention or mitigation of the severity of ventricular arrhythmias due to antioxidants has been indicated as the fundamental contribution in the field of preventive cardiology; however, novel interventions have to be developed for greater effectiveness and specificity in attenuating the adverse effects of oxidative stress. In this review, we have attempted to discuss proarrhythmic effects of oxidative stress differing in time and concentration dependence and highlight a molecular and cellular concept how it alters cardiac cell automaticity and conduction velocity sensitizing the probability of ventricular arrhythmias with resultant sudden cardiac death due to ischemic heart disease and other stressful situations. It is concluded that pharmacological approaches targeting multiple mechanisms besides oxidative stress might be more effective in the treatment of ventricular arrhythmias than current antiarrhythmic therapy.
Collapse
Affiliation(s)
- Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, and Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Anureet K. Shah
- Department of Kinesiology, Nutrition and Food Science, California State University, Los Angeles, CA 90032, USA;
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada;
| |
Collapse
|
30
|
Vetter L, Cortassa S, O'Rourke B, Armoundas AA, Bedja D, Jende JME, Bendszus M, Paolocci N, Sollot SJ, Aon MA, Kurz FT. Diabetes Increases the Vulnerability of the Cardiac Mitochondrial Network to Criticality. Front Physiol 2020; 11:175. [PMID: 32210835 PMCID: PMC7077512 DOI: 10.3389/fphys.2020.00175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/14/2020] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial criticality describes a state in which the mitochondrial cardiac network under intense oxidative stress becomes very sensitive to small perturbations, leading from local to cell-wide depolarization and synchronized oscillations that may escalate to the myocardial syncytium generating arrhythmias. Herein, we describe the occurrence of mitochondrial criticality in the chronic setting of a metabolic disorder, type 1 diabetes (T1DM), using a streptozotocin (STZ)-treated guinea pig (GP) animal model. Using wavelet analysis of mitochondrial networks from two-photon microscopy imaging of cardiac myocytes loaded with a fluorescent probe of the mitochondrial membrane potential, we show that cardiomyocytes from T1DM GPs are closer to criticality, making them more vulnerable to cell-wide mitochondrial oscillations as can be judged by the latency period to trigger oscillations after a laser flash perturbation, and their propensity to oscillate. Insulin treatment of T1DM GPs rescued cardiac myocytes to sham control levels of susceptibility, a protective condition that could also be attained with interventions leading to improvement of the cellular redox environment such as preincubation of diabetic cardiac myocytes with the lipid palmitate or a cell-permeable form of glutathione, in the presence of glucose.
Collapse
Affiliation(s)
- Larissa Vetter
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.,Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Antonis A Armoundas
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology Cambridge, MA, United States
| | - Djahida Bedja
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Johann M E Jende
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Steven J Sollot
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Felix T Kurz
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
31
|
Lu X, Thai PN, Lu S, Pu J, Bers DM. Intrafibrillar and perinuclear mitochondrial heterogeneity in adult cardiac myocytes. J Mol Cell Cardiol 2019; 136:72-84. [PMID: 31491377 DOI: 10.1016/j.yjmcc.2019.08.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/12/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022]
Abstract
Mitochondria are involved in multiple cellular functions, in addition to their core role in energy metabolism. Mitochondria localized in different cellular locations may have different morphology, Ca2+ handling and biochemical properties and may interact differently with other intracellular structures, causing functional specificity. However, most prior studies have utilized isolated mitochondria, removed from their intracellular environment. Mitochondria in cardiac ventricular myocytes are highly organized, with a majority squeezed between the myofilaments in longitudinal chains (intrafibrillar mitochondria, IFM). There is another population of perinuclear mitochondria (PNM) around and between the two nuclei typical in myocytes. Here, we take advantage of live myocyte imaging to test for quantitative morphological and functional differences between IFM and PNM with respect to calcium fluxes, membrane potential, sensitivity to oxidative stress, shape and dynamics. Our findings show higher mitochondrial Ca2+ uptake and oxidative stress sensitivity for IFM vs. PNM, which may relate to higher local energy demand supporting the contractile machinery. In contrast to IFM which are remarkably static, PNM are relatively mobile, appear to participate readily in fission/fusion dynamics and appear to play a central role in mitochondrial genesis and turnover. We conclude that while IFM may be physiologically tuned to support local myofilament energy demands, PNM may be more critical in mitochondrial turnover and regulation of nuclear function and import/export. Thus, important functional differences are present in intrafibrillar vs. perinuclear mitochondrial subpopulations.
Collapse
Affiliation(s)
- Xiyuan Lu
- Division of Cardiology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital School of Medicine, Shanghai Cancer Institute, Jiaotong University, Shanghai, China; Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | - Phung N Thai
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Shan Lu
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Jun Pu
- Division of Cardiology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital School of Medicine, Shanghai Cancer Institute, Jiaotong University, Shanghai, China
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| |
Collapse
|
32
|
Stožer A, Markovič R, Dolenšek J, Perc M, Marhl M, Slak Rupnik M, Gosak M. Heterogeneity and Delayed Activation as Hallmarks of Self-Organization and Criticality in Excitable Tissue. Front Physiol 2019; 10:869. [PMID: 31333504 PMCID: PMC6624746 DOI: 10.3389/fphys.2019.00869] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022] Open
Abstract
Self-organized critical dynamics is assumed to be an attractive mode of functioning for several real-life systems and entails an emergent activity in which the extent of observables follows a power-law distribution. The hallmarks of criticality have recently been observed in a plethora of biological systems, including beta cell populations within pancreatic islets of Langerhans. In the present study, we systematically explored the mechanisms that drive the critical and supercritical behavior in networks of coupled beta cells under different circumstances by means of experimental and computational approaches. Experimentally, we employed high-speed functional multicellular calcium imaging of fluorescently labeled acute mouse pancreas tissue slices to record calcium signals in a large number of beta cells simultaneously, and with a high spatiotemporal resolution. Our experimental results revealed that the cellular responses to stimulation with glucose are biphasic and glucose-dependent. Under physiological as well as under supraphysiological levels of stimulation, an initial activation phase was followed by a supercritical plateau phase with a high number of global intercellular calcium waves. However, the activation phase displayed fingerprints of critical behavior under lower stimulation levels, with a progressive recruitment of cells and a power-law distribution of calcium wave sizes. On the other hand, the activation phase provoked by pathophysiologically high glucose concentrations, differed considerably and was more rapid, less continuous, and supercritical. To gain a deeper insight into the experimentally observed complex dynamical patterns, we built up a phenomenological model of coupled excitable cells and explored empirically the model’s necessities that ensured a good overlap between computational and experimental results. It turned out that such a good agreement between experimental and computational findings was attained when both heterogeneous and stimulus-dependent time lags, variability in excitability levels, as well as a heterogeneous cell-cell coupling were included into the model. Most importantly, since our phenomenological approach involved only a few parameters, it naturally lends itself not only for determining key mechanisms of self-organized criticality at the tissue level, but also points out various features for comprehensive and realistic modeling of different excitable systems in nature.
Collapse
Affiliation(s)
- Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Faculty of Education, University of Maribor, Maribor, Slovenia.,Faculty of Energy Technology, University of Maribor, Krško, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Matjaž Perc
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Center for Applied Mathematics and Theoretical Physics, University of Maribor, Maribor, Slovenia.,Complexity Science Hub Vienna, Vienna, Austria
| | - Marko Marhl
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Faculty of Education, University of Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Institute of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.,Alma Mater Europaea - ECM, Maribor, Slovenia
| | - Marko Gosak
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| |
Collapse
|
33
|
Grisanti LA. Diabetes and Arrhythmias: Pathophysiology, Mechanisms and Therapeutic Outcomes. Front Physiol 2018; 9:1669. [PMID: 30534081 PMCID: PMC6275303 DOI: 10.3389/fphys.2018.01669] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
The prevalence of diabetes is rapidly increasing and closely associated with cardiovascular morbidity and mortality. While the major cardiovascular complication associated with diabetes is coronary artery disease, it is becoming increasingly apparent that diabetes impacts the electrical conduction system in the heart, resulting in atrial fibrillation, and ventricular arrhythmias. The relationship between diabetes and arrhythmias is complex and multifactorial including autonomic dysfunction, atrial and ventricular remodeling and molecular alterations. This review will provide a comprehensive overview of the link between diabetes and arrhythmias with insight into the common molecular mechanisms, structural alterations and therapeutic outcomes.
Collapse
Affiliation(s)
- Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
34
|
Kembro JM, Cortassa S, Lloyd D, Sollott SJ, Aon MA. Mitochondrial chaotic dynamics: Redox-energetic behavior at the edge of stability. Sci Rep 2018; 8:15422. [PMID: 30337561 PMCID: PMC6194025 DOI: 10.1038/s41598-018-33582-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/25/2018] [Indexed: 12/14/2022] Open
Abstract
Mitochondria serve multiple key cellular functions, including energy generation, redox balance, and regulation of apoptotic cell death, thus making a major impact on healthy and diseased states. Increasingly recognized is that biological network stability/instability can play critical roles in determining health and disease. We report for the first-time mitochondrial chaotic dynamics, characterizing the conditions leading from stability to chaos in this organelle. Using an experimentally validated computational model of mitochondrial function, we show that complex oscillatory dynamics in key metabolic variables, arising at the “edge” between fully functional and pathological behavior, sets the stage for chaos. Under these conditions, a mild, regular sinusoidal redox forcing perturbation triggers chaotic dynamics with main signature traits such as sensitivity to initial conditions, positive Lyapunov exponents, and strange attractors. At the “edge” mitochondrial chaos is exquisitely sensitive to the antioxidant capacity of matrix Mn superoxide dismutase as well as to the amplitude and frequency of the redox perturbation. These results have potential implications both for mitochondrial signaling determining health maintenance, and pathological transformation, including abnormal cardiac rhythms.
Collapse
Affiliation(s)
- Jackelyn M Kembro
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), and Instituto de Ciencia y Tecnología de los Alimentos, Cátedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Velez Sarsfield 1611, Córdoba, X5000HUA, Cordoba, Argentina
| | - Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH. 251 Bayview Boulevard, Baltimore, 21224, MD, USA
| | - David Lloyd
- School of Biosciences, Cardiff University, Main Building, Museum Avenue, Cardiff, CF10 3AT, Wales, UK
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH. 251 Bayview Boulevard, Baltimore, 21224, MD, USA
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH. 251 Bayview Boulevard, Baltimore, 21224, MD, USA.
| |
Collapse
|
35
|
Makarov VI, Khmelinskii I, Javadov S. Computational Modeling of In Vitro Swelling of Mitochondria: A Biophysical Approach. Molecules 2018; 23:molecules23040783. [PMID: 29597314 PMCID: PMC5901922 DOI: 10.3390/molecules23040783] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/12/2018] [Accepted: 03/27/2018] [Indexed: 11/17/2022] Open
Abstract
Swelling of mitochondria plays an important role in the pathogenesis of human diseases by stimulating mitochondria-mediated cell death through apoptosis, necrosis, and autophagy. Changes in the permeability of the inner mitochondrial membrane (IMM) of ions and other substances induce an increase in the colloid osmotic pressure, leading to matrix swelling. Modeling of mitochondrial swelling is important for simulation and prediction of in vivo events in the cell during oxidative and energy stress. In the present study, we developed a computational model that describes the mechanism of mitochondrial swelling based on osmosis, the rigidity of the IMM, and dynamics of ionic/neutral species. The model describes a new biophysical approach to swelling dynamics, where osmotic pressure created in the matrix is compensated for by the rigidity of the IMM, i.e., osmotic pressure induces membrane deformation, which compensates for the osmotic pressure effect. Thus, the effect is linear and reversible at small membrane deformations, allowing the membrane to restore its normal form. On the other hand, the membrane rigidity drops to zero at large deformations, and the swelling becomes irreversible. As a result, an increased number of dysfunctional mitochondria can activate mitophagy and initiate cell death. Numerical modeling analysis produced results that reasonably describe the experimental data reported earlier.
Collapse
Affiliation(s)
- Vladimir I Makarov
- Department of Physics, University of Puerto Rico, Rio Piedras Campus, San Juan, PR 00931-3343, USA.
| | - Igor Khmelinskii
- Faculty of Sciences and Technology, Department of Chemistry and Pharmacy, and Interdisciplinary Centre of Chemistry of Algarve, University of Algarve, 8005-139 Faro, Portugal.
| | - Sabzali Javadov
- Department of Physiology and Biophysics, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936-5067, USA.
| |
Collapse
|
36
|
Kurz FT, Aon MA, O'Rourke B, Armoundas AA. Assessing Spatiotemporal and Functional Organization of Mitochondrial Networks. Methods Mol Biol 2018; 1782:383-402. [PMID: 29851013 PMCID: PMC7003721 DOI: 10.1007/978-1-4939-7831-1_23] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
The functional and spatiotemporal organization of mitochondrial redox signaling networks can be studied in detail in cardiac myocytes and neurons by assessing the time-resolved signaling traits of their individual mitochondrial components. Perturbations of the mitochondrial network through oxidative stress can lead to coordinated, cluster-bound behavior in the form of synchronized limit-cycle oscillations of mitochondrial inner membrane potentials. These oscillations are facilitated by both structural coupling through changes in the local redox balance and signaling microdomains and functional coupling that is yet poorly understood. Thus, quantifiable measures of both coupling mechanisms, local dynamic mitochondrial coupling constants and functional clustering coefficients, are likely to offer valuable information on mitochondrial network organization. We provide step-by-step methodologies on how to acquire and assess these measures for inner membrane potential fluorescence fluctuations in laser-scanning two-photon microscope recordings of cardiac myocytes and neurons, that can be applied to other tissues as well.
Collapse
MESH Headings
- Animals
- Cluster Analysis
- Fluorescent Dyes/chemistry
- Guinea Pigs
- Image Processing, Computer-Assisted/instrumentation
- Image Processing, Computer-Assisted/methods
- Membrane Potential, Mitochondrial
- Microscopy, Confocal/instrumentation
- Microscopy, Confocal/methods
- Microscopy, Fluorescence/instrumentation
- Microscopy, Fluorescence/methods
- Mitochondria, Heart/metabolism
- Mitochondrial Dynamics
- Models, Biological
- Myocytes, Cardiac/cytology
- Neurons
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Software
Collapse
Affiliation(s)
- Felix T Kurz
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Antonis A Armoundas
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
37
|
Hung YP, Teragawa C, Kosaisawe N, Gillies TE, Pargett M, Minguet M, Distor K, Rocha-Gregg BL, Coloff JL, Keibler MA, Stephanopoulos G, Yellen G, Brugge JS, Albeck JG. Akt regulation of glycolysis mediates bioenergetic stability in epithelial cells. eLife 2017; 6:27293. [PMID: 29239720 PMCID: PMC5730373 DOI: 10.7554/elife.27293] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 12/05/2017] [Indexed: 12/26/2022] Open
Abstract
Cells use multiple feedback controls to regulate metabolism in response to nutrient and signaling inputs. However, feedback creates the potential for unstable network responses. We examined how concentrations of key metabolites and signaling pathways interact to maintain homeostasis in proliferating human cells, using fluorescent reporters for AMPK activity, Akt activity, and cytosolic NADH/NAD+ redox. Across various conditions, including glycolytic or mitochondrial inhibition or cell proliferation, we observed distinct patterns of AMPK activity, including both stable adaptation and highly dynamic behaviors such as periodic oscillations and irregular fluctuations that indicate a failure to reach a steady state. Fluctuations in AMPK activity, Akt activity, and cytosolic NADH/NAD+ redox state were temporally linked in individual cells adapting to metabolic perturbations. By monitoring single-cell dynamics in each of these contexts, we identified PI3K/Akt regulation of glycolysis as a multifaceted modulator of single-cell metabolic dynamics that is required to maintain metabolic stability in proliferating cells.
Collapse
Affiliation(s)
- Yin P Hung
- Department of Cell Biology, Harvard Medical School, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States.,Department of Pathology, Brigham and Women's Hospital, Boston, United States
| | - Carolyn Teragawa
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Nont Kosaisawe
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Taryn E Gillies
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Michael Pargett
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Marta Minguet
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Kevin Distor
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Briana L Rocha-Gregg
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| | - Jonathan L Coloff
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Mark A Keibler
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - John G Albeck
- Department of Molecular and Cellular Biology, University of California, Davis, United States
| |
Collapse
|
38
|
Gambardella J, Sorriento D, Ciccarelli M, Del Giudice C, Fiordelisi A, Napolitano L, Trimarco B, Iaccarino G, Santulli G. Functional Role of Mitochondria in Arrhythmogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:191-202. [PMID: 28551788 DOI: 10.1007/978-3-319-55330-6_10] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Growing evidence indicate that mitochondria play a functional role in arrhythmogenesis. We report here the molecular mechanisms underlying the action of these highly dynamic organelles in the regulation of cell metabolism, action potential and, overall, heart excitability. In particular, we examine the role of cardiac mitochondria in linking metabolism and cell excitability. The importance of the main mitochondrial channels is evaluated as well, including the recently identified calcium uniporter. Promises and pitfalls of potential therapeutic strategies targeting mitochondrial pathways are also assessed.
Collapse
Affiliation(s)
- Jessica Gambardella
- Columbia University Medical Center, New York, NY, USA.,Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Carmine Del Giudice
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Luigi Napolitano
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Gaetano Santulli
- Dept. of Biomedical Advanced Sciences, Federico II University, Naples, Italy. .,Columbia University Medical Center, New York Presbyterian Hospital-Manhattan, New York, NY, USA.
| |
Collapse
|
39
|
Javadov S, Chapa-Dubocq X, Makarov V. Different approaches to modeling analysis of mitochondrial swelling. Mitochondrion 2017; 38:58-70. [PMID: 28802667 DOI: 10.1016/j.mito.2017.08.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/21/2017] [Accepted: 08/08/2017] [Indexed: 12/11/2022]
Abstract
Mitochondria are critical players involved in both cell life and death through multiple pathways. Structural integrity, metabolism and function of mitochondria are regulated by matrix volume due to physiological changes of ion homeostasis in cellular cytoplasm and mitochondria. Ca2+ and K+ presumably play a critical role in physiological and pathological swelling of mitochondria when increased uptake (influx)/decreased release (efflux) of these ions enhances osmotic pressure accompanied by high water accumulation in the matrix. Changes in the matrix volume in the physiological range have a stimulatory effect on electron transfer chain and oxidative phosphorylation to satisfy metabolic requirements of the cell. However, excessive matrix swelling associated with the sustained opening of mitochondrial permeability transition pores (PTP) and other PTP-independent mechanisms compromises mitochondrial function and integrity leading to cell death. The mechanisms of transition from reversible (physiological) to irreversible (pathological) swelling of mitochondria remain unknown. Mitochondrial swelling is involved in the pathogenesis of many human diseases such as neurodegenerative and cardiovascular diseases. Therefore, modeling analysis of the swelling process is important for understanding the mechanisms of cell dysfunction. This review attempts to describe the role of mitochondrial swelling in cell life and death and the main mechanisms involved in the maintenance of ion homeostasis and swelling. The review also summarizes and discusses different kinetic models and approaches that can be useful for the development of new models for better simulation and prediction of in vivo mitochondrial swelling.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR, USA.
| | - Xavier Chapa-Dubocq
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR, USA
| | - Vladimir Makarov
- Department of Physics, Rio Piedras Campus, University of Puerto Rico, San Juan, PR, USA
| |
Collapse
|
40
|
Tomasek M, Misak A, Grman M, Tomaskova Z. Subconductance states of mitochondrial chloride channels: implication for functionally-coupled tetramers. FEBS Lett 2017. [PMID: 28640976 DOI: 10.1002/1873-3468.12721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recently, it has been discovered that isoforms of intracellular chloride channels (CLIC) are present in cardiac mitochondria. By reconstituting rat cardiac mitochondrial chloride channels into bilayer lipid membranes, we detected three equally separated subconductance states with conductance increment of 45 pS and < 2% occupancy. The observed rare events of channel decomposition into substates, accompanied by disrupted gating, provide an insight into channel quaternary structure. Our findings suggest that the observed channels work as four functionally coupled subunits with synchronized gating. We discuss the putative connection of channel activity from native mitochondria with the recombinant CLIC channels. However, conclusive evidence is needed to prove this connection.
Collapse
Affiliation(s)
| | - Anton Misak
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak
| | - Marian Grman
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak
| | - Zuzana Tomaskova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak
| |
Collapse
|
41
|
Kuznetsov AV, Javadov S, Saks V, Margreiter R, Grimm M. Synchronism in mitochondrial ROS flashes, membrane depolarization and calcium sparks in human carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:418-431. [PMID: 28279675 DOI: 10.1016/j.bbabio.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/09/2017] [Accepted: 03/02/2017] [Indexed: 02/02/2023]
Abstract
Mitochondria are major producers of reactive oxygen species (ROS) in many cells including cancer cells. However, complex interrelationships between mitochondrial ROS (mitoROS), mitochondrial membrane potential (ΔΨm) and Ca2+ are not completely understood. Using human carcinoma cells, we further highlight biphasic ROS dynamics: - gradual mitoROS increase followed by mitoROS flash. Also, we demonstrate heterogeneity in rates of mitoROS generation and flash initiation time. Comparing mitochondrial and near-extra-mitochondrial signals, we show that mechanisms of mitoROS flashes in single mitochondria, linked to mitochondrial permeability transition pore opening (ΔΨm collapse) and calcium sparks, may involve flash triggering by certain levels of external ROS released from the same mitochondria. In addition, mitochondria-mitochondria interactions can produce wave propagations of mitoROS flashes and ΔΨm collapses in cancer cells similar to phenomena of ROS-induced ROS release (RIRR). Our data suggest that in cancer cells RIRR, activation of mitoROS flashes and mitochondrial depolarization may involve participation of extramitochondrial-ROS produced either by individual mitochondria and/or by neighboring mitochondria. This could represent general mechanisms in ROS-ROS signaling with suggested role in both mitochondrial and cellular physiology and signaling.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Cardiac Surgery Laboratory, Department of Heart Surgery, Medical University of Innsbruck, Innsbruck A-6020, Austria.
| | - Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, USA
| | - Valdur Saks
- Laboratory of Fundamental and Applied Bioenergetics, INSERM U884, University Joseph Fourier, Grenoble, France
| | - Raimund Margreiter
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Grimm
- Cardiac Surgery Laboratory, Department of Heart Surgery, Medical University of Innsbruck, Innsbruck A-6020, Austria
| |
Collapse
|
42
|
Kurz FT, Aon MA, O'Rourke B, Armoundas AA. Functional Implications of Cardiac Mitochondria Clustering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:1-24. [PMID: 28551779 PMCID: PMC7003720 DOI: 10.1007/978-3-319-55330-6_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The spatio-temporal organization of mitochondria in cardiac myocytes facilitates myocyte-wide, cluster-bound, mitochondrial inner membrane potential oscillatory depolarizations, commonly triggered by metabolic or oxidative stressors. Local intermitochondrial coupling can be mediated by reactive oxygen species (ROS) that activate inner membrane pores to initiate a ROS-induced-ROS-release process that produces synchronized limit cycle oscillations of mitochondrial clusters within the whole mitochondrial network. The network's dynamic organization, structure and function can be assessed by quantifying dynamic local coupling constants and dynamic functional clustering coefficients, both providing information about the network's response to external stimuli. In addition to its special organization, the mitochondrial network of cardiac myocytes exhibits substrate-sensitive coupling constants and clustering coefficients. The myocyte's ability to form functional clusters of synchronously oscillating mitochondria is sensitive to conditions such as substrate availability (e.g., glucose, pyruvate, β-hydroxybutyrate), antioxidant status, respiratory chain activity, or history of oxidative challenge (e.g., ischemia-reperfusion). This underscores the relevance of quantitative methods to characterize the network's functional status as a way to assess the myocyte's resilience to pathological stressors.
Collapse
Affiliation(s)
- Felix T Kurz
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, MA, USA.
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Antonis A Armoundas
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
43
|
Lancaster G, Suprunenko YF, Jenkins K, Stefanovska A. Modelling chronotaxicity of cellular energy metabolism to facilitate the identification of altered metabolic states. Sci Rep 2016; 6:29584. [PMID: 27483987 PMCID: PMC4971499 DOI: 10.1038/srep29584] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/22/2016] [Indexed: 12/22/2022] Open
Abstract
Altered cellular energy metabolism is a hallmark of many diseases, one notable example being cancer. Here, we focus on the identification of the transition from healthy to abnormal metabolic states. To do this, we study the dynamics of energy production in a cell. Due to the thermodynamic openness of a living cell, the inability to instantaneously match fluctuating supply and demand in energy metabolism results in nonautonomous time-varying oscillatory dynamics. However, such oscillatory dynamics is often neglected and treated as stochastic. Based on experimental evidence of metabolic oscillations, we show that changes in metabolic state can be described robustly by alterations in the chronotaxicity of the corresponding metabolic oscillations, i.e. the ability of an oscillator to resist external perturbations. We also present a method for the identification of chronotaxicity, applicable to general oscillatory signals and, importantly, apply this to real experimental data. Evidence of chronotaxicity was found in glycolytic oscillations in real yeast cells, verifying that chronotaxicity could be used to study transitions between metabolic states.
Collapse
Affiliation(s)
- Gemma Lancaster
- Department of Physics, Lancaster University, Lancaster, LA1 4YB, UK
| | - Yevhen F Suprunenko
- Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Kirsten Jenkins
- Randall Division of Cell &Molecular Biophysics, King's College London, London, WC2R 2LS, UK
| | | |
Collapse
|
44
|
Alleman RJ, Tsang AM, Ryan TE, Patteson DJ, McClung JM, Spangenburg EE, Shaikh SR, Neufer PD, Brown DA. Exercise-induced protection against reperfusion arrhythmia involves stabilization of mitochondrial energetics. Am J Physiol Heart Circ Physiol 2016; 310:H1360-70. [PMID: 26945082 PMCID: PMC4888539 DOI: 10.1152/ajpheart.00858.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/26/2016] [Indexed: 11/22/2022]
Abstract
Mitochondria influence cardiac electrophysiology through energy- and redox-sensitive ion channels in the sarcolemma, with the collapse of energetics believed to be centrally involved in arrhythmogenesis. This study was conducted to determine if preservation of mitochondrial membrane potential (ΔΨm) contributes to the antiarrhythmic effect of exercise. We utilized perfused hearts, isolated myocytes, and isolated mitochondria exposed to metabolic challenge to determine the effects of exercise on cardiac mitochondria. Hearts from sedentary (Sed) and exercised (Ex; 10 days of treadmill running) Sprague-Dawley rats were perfused on a two-photon microscope stage for simultaneous measurement of ΔΨm and ECG. After ischemia-reperfusion, the collapse of ΔΨm was commensurate with the onset of arrhythmia. Exercise preserved ΔΨm and decreased the incidence of fibrillation/tachycardia (P < 0.05). Our findings in intact hearts were corroborated in isolated myocytes exposed to in vitro hypoxia-reoxygenation, with Ex rats demonstrating enhanced redox control and sustained ΔΨm during reoxygenation. Finally, we induced anoxia-reoxygenation in isolated mitochondria using high-resolution respirometry with simultaneous measurement of respiration and H2O2 Mitochondria from Ex rats sustained respiration with lower rates of H2O2 emission than Sed rats. Exercise helps sustain postischemic mitochondrial bioenergetics and redox homeostasis, which is associated with preserved ΔΨm and protection against reperfusion arrhythmia. The reduction of fatal ventricular arrhythmias through exercise-induced mitochondrial adaptations indicates that mitochondrial therapeutics may be an effective target for the treatment of heart disease.
Collapse
Affiliation(s)
- Rick J Alleman
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Alvin M Tsang
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Daniel J Patteson
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Espen E Spangenburg
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Saame Raza Shaikh
- East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - P Darrell Neufer
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| |
Collapse
|
45
|
Opacic D, van Bragt KA, Nasrallah HM, Schotten U, Verheule S. Atrial metabolism and tissue perfusion as determinants of electrical and structural remodelling in atrial fibrillation. Cardiovasc Res 2016; 109:527-41. [DOI: 10.1093/cvr/cvw007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022] Open
|
46
|
Kurz FT, Derungs T, Aon MA, O'Rourke B, Armoundas AA. Mitochondrial networks in cardiac myocytes reveal dynamic coupling behavior. Biophys J 2016; 108:1922-33. [PMID: 25902432 DOI: 10.1016/j.bpj.2015.01.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/13/2015] [Indexed: 01/20/2023] Open
Abstract
Oscillatory behavior of mitochondrial inner membrane potential (ΔΨm) is commonly observed in cells subjected to oxidative or metabolic stress. In cardiac myocytes, the activation of inner membrane pores by reactive oxygen species (ROS) is a major factor mediating intermitochondrial coupling, and ROS-induced ROS release has been shown to underlie propagated waves of ΔΨm depolarization as well as synchronized limit cycle oscillations of ΔΨm in the network. The functional impact of ΔΨm instability on cardiac electrophysiology, Ca(2+) handling, and even cell survival, is strongly affected by the extent of such intermitochondrial coupling. Here, we employ a recently developed wavelet-based analytical approach to examine how different substrates affect mitochondrial coupling in cardiac cells, and we also determine the oscillatory coupling properties of mitochondria in ventricular cells in intact perfused hearts. The results show that the frequency of ΔΨm oscillations varies inversely with the size of the oscillating mitochondrial cluster, and depends on the strength of local intermitochondrial coupling. Time-varying coupling constants could be quantitatively determined by applying a stochastic phase model based on extension of the well-known Kuramoto model for networks of coupled oscillators. Cluster size-frequency relationships varied with different substrates, as did mitochondrial coupling constants, which were significantly larger for glucose (7.78 × 10(-2) ± 0.98 × 10(-2) s(-1)) and pyruvate (7.49 × 10(-2) ± 1.65 × 10(-2) s(-1)) than lactate (4.83 × 10(-2) ± 1.25 × 10(-2) s(-1)) or β-hydroxybutyrate (4.11 × 10(-2) ± 0.62 × 10(-2) s(-1)). The findings indicate that mitochondrial spatiotemporal coupling and oscillatory behavior is influenced by substrate selection, perhaps through differing effects on ROS/redox balance. In particular, glucose-perfusion generates strong intermitochondrial coupling and temporal oscillatory stability. Pathological changes in specific catabolic pathways, which are known to occur during the progression of cardiovascular disease, could therefore contribute to altered sensitivity of the mitochondrial network to oxidative stress and emergent ΔΨm instability, ultimately scaling to produce organ level dysfunction.
Collapse
Affiliation(s)
- Felix T Kurz
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, Massachusetts; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Cardiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Derungs
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, Massachusetts; Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Miguel A Aon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Antonis A Armoundas
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, Massachusetts.
| |
Collapse
|
47
|
|
48
|
Goh KY, Qu J, Hong H, Liu T, Dell'Italia LJ, Wu Y, O'Rourke B, Zhou L. Impaired mitochondrial network excitability in failing guinea-pig cardiomyocytes. Cardiovasc Res 2015; 109:79-89. [PMID: 26433944 DOI: 10.1093/cvr/cvv230] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 09/25/2015] [Indexed: 01/11/2023] Open
Abstract
AIMS Studies in guinea-pig cardiomyocytes show that reactive oxygen species (ROS) produced by a few mitochondria can propagate to their neighbours, triggering synchronized, cell-wide network oscillations via an ROS-induced ROS release (RIRR) mechanism. How mitochondria in cardiomyocytes from failing hearts (HF) respond to local oxidative stress perturbations has not been investigated. Since mitochondrial ultrastructure is reportedly disrupted in HF, and propagation of ROS signals depends on mitochondrial network integrity, we hypothesized that the laser flash-induced RIRR is altered in HF. METHODS AND RESULTS To test the hypothesis, pressure-overload HF was induced in guinea pigs by ascending aortic constriction leading to left ventricular dilatation and decreased ejection fraction after 8 weeks. Isolated cardiomyocytes were studied with two-photon/confocal microscopy to determine their basal oxidative stress and propensity to undergo mitochondrial depolarization/oscillations in response to local laser flash stimulations. The expression of mitofusin proteins and mitochondrial network structure were also analysed. Results showed that HF cardiomyocytes had higher baseline ROS levels and less reduced glutathione, and were more prone to laser flash-induced mitochondrial depolarization. In contrast, the delay between the laser flash and synchronized cell-wide network oscillations was prolonged in HF myocytes compared with shams, and the spatial extent of coupling was diminished, suggesting dampened RIRR and ROS signal propagation. In addition, the expressions of mitofusin proteins in HF myocardium were down-regulated compared with these from sham-operated animals, and the mitochondrial network structure altered. CONCLUSION The disrupted inter-mitochondrial tethering and loss of structural organization may underlie decreased ROS-dependent mitochondrial coupling in HF.
Collapse
Affiliation(s)
- Kah Yong Goh
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jing Qu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Huixian Hong
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ting Liu
- Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Louis J Dell'Italia
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yong Wu
- Division of Molecular Medicine, Department of Anesthesiology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Lufang Zhou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
49
|
Nemutlu E, Gupta A, Zhang S, Viqar M, Holmuhamedov E, Terzic A, Jahangir A, Dzeja P. Decline of Phosphotransfer and Substrate Supply Metabolic Circuits Hinders ATP Cycling in Aging Myocardium. PLoS One 2015; 10:e0136556. [PMID: 26378442 PMCID: PMC4574965 DOI: 10.1371/journal.pone.0136556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/04/2015] [Indexed: 12/24/2022] Open
Abstract
Integration of mitochondria with cytosolic ATP-consuming/ATP-sensing and substrate supply processes is critical for muscle bioenergetics and electrical activity. Whether age-dependent muscle weakness and increased electrical instability depends on perturbations in cellular energetic circuits is unknown. To define energetic remodeling of aged atrial myocardium we tracked dynamics of ATP synthesis-utilization, substrate supply, and phosphotransfer circuits through adenylate kinase (AK), creatine kinase (CK), and glycolytic/glycogenolytic pathways using 18O stable isotope-based phosphometabolomic technology. Samples of intact atrial myocardium from adult and aged rats were subjected to 18O-labeling procedure at resting basal state, and analyzed using the 18O-assisted HPLC-GC/MS technique. Characteristics for aging atria were lower inorganic phosphate Pi[18O], γ-ATP[18O], β-ADP[18O], and creatine phosphate CrP[18O] 18O-labeling rates indicating diminished ATP utilization-synthesis and AK and CK phosphotransfer fluxes. Shift in dynamics of glycolytic phosphotransfer was reflected in the diminished G6P[18O] turnover with relatively constant glycogenolytic flux or G1P[18O] 18O-labeling. Labeling of G3P[18O], an indicator of G3P-shuttle activity and substrate supply to mitochondria, was depressed in aged myocardium. Aged atrial myocardium displayed reduced incorporation of 18O into second (18O2), third (18O3), and fourth (18O4) positions of Pi[18O] and a lower Pi[18O]/γ-ATP[18 O]-labeling ratio, indicating delayed energetic communication and ATP cycling between mitochondria and cellular ATPases. Adrenergic stress alleviated diminished CK flux, AK catalyzed β-ATP turnover and energetic communication in aging atria. Thus, 18O-assisted phosphometabolomics uncovered simultaneous phosphotransfer through AK, CK, and glycolytic pathways and G3P substrate shuttle deficits hindering energetic communication and ATP cycling, which may underlie energetic vulnerability of aging atrial myocardium.
Collapse
Affiliation(s)
- Emirhan Nemutlu
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Anu Gupta
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Song Zhang
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Maria Viqar
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ekhson Holmuhamedov
- Center for Integrative Research on Cardiovascular Aging (CIRCA), Aurora University of Wisconsin Medical Group, Aurora Health Care, Milwaukee, Wisconsin, United States of America
| | - Andre Terzic
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Arshad Jahangir
- Center for Integrative Research on Cardiovascular Aging (CIRCA), Aurora University of Wisconsin Medical Group, Aurora Health Care, Milwaukee, Wisconsin, United States of America
- * E-mail: (PD); (AJ)
| | - Petras Dzeja
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (PD); (AJ)
| |
Collapse
|
50
|
Salimi A, Eybagi S, Seydi E, Naserzadeh P, Kazerouni NP, Pourahmad J. Toxicity of macrolide antibiotics on isolated heart mitochondria: a justification for their cardiotoxic adverse effect. Xenobiotica 2015; 46:82-93. [PMID: 26068526 DOI: 10.3109/00498254.2015.1046975] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
1. Macrolides belong to the polyketide class of natural products. These products are a group of drugs (typically antibiotics) which their activity stems from the presence of a macrolide ring. Antibiotic macrolides are used to treat infections caused by Gram-positive bacteria and Haemophilus influenzae infections such as respiratory tract and soft-tissue infections. Macrolides, mainly erythromycin and clarithromycin, rarely show QT prolongation, as their infamous adverse reaction which can lead to torsades de pointes. Electrophysiological studies showed that macrolides prolonging the QT interval inhibit the rapid component of the delayed rectifier K(+) current (IKr) through the block of potassium channels encoded by the human ether-a-go-go-related gene (HERG). Other studies suggest that increased ROS generation alters the kinetics of hERG K(+) conductance. 2. In our study, rat cardiomyocytes were isolated with collagen perfusion technique. Finally, mitochondria isolated from cardiomyocytes were exposed to erythromycin, azithromycin and clarithromycin for their probable toxicity effects. 3. Our results demonstrated that macrolides induced reactive oxygen species formation, mitochondrial membrane permeabilization and mitochondrial swelling and finally cytochrome c release in cardiomyocyte mitochondria. 4. These findings suggested that the toxicity of heart mitochondria is a starting point for cardiotoxic effects of macrolides including QT prolongation, torsades de pointes and arrhythmia.
Collapse
Affiliation(s)
- Ahmad Salimi
- a Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran and
| | - Sadaf Eybagi
- a Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran and
| | - Enayatollah Seydi
- a Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran and
| | - Parvaneh Naserzadeh
- a Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran and
| | | | - Jalal Pourahmad
- a Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran and
| |
Collapse
|