Increase in Cardiac Ischemia-Reperfusion Injuries in Opa1+/- Mouse Model

Sophie Le Page, Marjorie Niro, Jérémy Fauconnier, Laura Cellier, Sophie Tamareille, Abdallah Gharib, Arnaud Chevrollier, Laurent Loufrani, Céline Grenier, Rima Kamel, Emmanuelle Sarzi, Alain Lacampagne, Michel Ovize, Daniel Henrion, Pascal Reynier, Guy Lenaers, Delphine Mirebeau-Prunier, Fabrice Prunier, Sophie Le Page, Marjorie Niro, Jérémy Fauconnier, Laura Cellier, Sophie Tamareille, Abdallah Gharib, Arnaud Chevrollier, Laurent Loufrani, Céline Grenier, Rima Kamel, Emmanuelle Sarzi, Alain Lacampagne, Michel Ovize, Daniel Henrion, Pascal Reynier, Guy Lenaers, Delphine Mirebeau-Prunier, Fabrice Prunier

Abstract

Background: Recent data suggests the involvement of mitochondrial dynamics in cardiac ischemia/reperfusion (I/R) injuries. Whilst excessive mitochondrial fission has been described as detrimental, the role of fusion proteins in this context remains uncertain.

Objectives: To investigate whether Opa1 (protein involved in mitochondrial inner-membrane fusion) deficiency affects I/R injuries.

Methods and results: We examined mice exhibiting Opa1delTTAG mutations (Opa1+/-), showing 70% Opa1 protein expression in the myocardium as compared to their wild-type (WT) littermates. Cardiac left-ventricular systolic function assessed by means of echocardiography was observed to be similar in 3-month-old WT and Opa1+/- mice. After subjection to I/R, infarct size was significantly greater in Opa1+/- than in WTs both in vivo (43.2±4.1% vs. 28.4±3.5%, respectively; p<0.01) and ex vivo (71.1±3.2% vs. 59.6±8.5%, respectively; p<0.05). No difference was observed in the expression of other main fission/fusion protein, oxidative phosphorylation, apoptotic markers, or mitochondrial permeability transition pore (mPTP) function. Analysis of calcium transients in isolated ventricular cardiomyocytes demonstrated a lower sarcoplasmic reticulum Ca2+ uptake, whereas cytosolic Ca2+ removal from the Na+/Ca2+ exchanger (NCX) was increased, whilst SERCA2a, phospholamban, and NCX protein expression levels were unaffected in Opa1+/- compared to WT mice. Simultaneous whole-cell patch-clamp recordings of mitochondrial Ca2+ movements and ventricular action potential (AP) showed impairment of dynamic mitochondrial Ca2+ uptake and a marked increase in the AP late repolarization phase in conjunction with greater occurrence of arrhythmia in Opa1+/- mice.

Conclusion: Opa1 deficiency was associated with increased sensitivity to I/R, imbalance in dynamic mitochondrial Ca2+ uptake, and subsequent increase in NCX activity.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Cardiac morphology and function.
Fig 1. Cardiac morphology and function.
BW (A) and HW/BW ratio (B) in 3- and 6-month-old Opa1+/- (n = 11 and 12 respectively) and WT (n = 6 and 15 respectively) mice. C, D: Echocardiography parameters in 3- and 6-month-old mice,(n = 9/group at 3 months and 10/group at 6 months) whereby C represents left-ventricular end-diastolic diameter (LVEDD), and D depicts fractional shortening (FS). E, F: Examples of electron microscopy images at 12,000x magnification in 3-month-old WT (E) and Opa+/- (F) mice. Values are mean ± SEM. * p<0.05 and ** p<0.01. Non-parametric Mann-Whitney test (HW/BW) and t-test (BW, LVEDD and FS) according to distribution.
Fig 2. Infarct size.
Fig 2. Infarct size.
A, B: Examples of left-ventricular sections with TTC staining after 30 minutes of ischemia and 2 hours of reperfusion ex vivo in WT (A) and Opa1+/- (B) mice, and histograms showing AN as a percentage of total LV in WT (n = 11) and OPA1+/- (n = 12) mice (C). D, E: Examples of left-ventricular sections with TTC-staining after 45 minutes of ischemia and 2 hours of reperfusion in vivo in WT (D) and OPA1+/- (E) mice. On the left side, images before TTC staining showing Evans blue coloration of the perfused myocardium and AAR as the non-blue area. On the right side, images after TTC staining showing AN in white. F: Histograms showing AN as a percentage of AAR and AAR as a percentage of total LV area in Opa1+/- and WT mice (n = 8/group). Values are mean ± SEM. * p<0.05 and ** p<0.01. Non-parametric Mann-Whitney test (AN/AAR) and t-test (AAR/LV).
Fig 3. Baseline and post-I/R fission and…
Fig 3. Baseline and post-I/R fission and fusion protein expression.
Opa1, Mfn2, Drp1, and Fis1 expression assessed by means of western blotting at baseline (n = 6/group) and after I/R (n = 3-6/group). GAPDH was used as a loading control. Values are mean ± SEM. * p

Fig 4. Baseline and post-I/R oxidative phosphorylation.

Fig 4. Baseline and post-I/R oxidative phosphorylation.

Complex I, II, and IV oxygen consumption were…

Fig 4. Baseline and post-I/R oxidative phosphorylation.
Complex I, II, and IV oxygen consumption were measured in WT and Opa1+/- mouse hearts at baseline and after I/R (n = 6/group). The I/R protocol consisted of 45 minutes of ischemia and 2 hours of reperfusion. The ratio [state 3 rate]: [state 4 rate] is represented by means of the respiratory control index (RCI) in both groups at baseline and after I/R. Values are mean ± SEM. *p<0.05, **p<0.01, ***p<0.001.

Fig 5. Apoptosis assessment and sensitivity to…

Fig 5. Apoptosis assessment and sensitivity to mPTP.

A : Examples of TUNEL images of…
Fig 5. Apoptosis assessment and sensitivity to mPTP.
A: Examples of TUNEL images of left-ventricular sections in confocal microscopy (70x magnification) after I/R. Cell nuclei are stained in red (propidium iodide, wavelength = 620 nm). TUNEL-positive nuclei appear in green (fluorescein, wavelength = 460 nm). B: Bax and Bcl-2 protein expressions (n = 4-6/group). C: Percentage of TUNEL-positive nuclei compared to total nuclei (n = 8 in Opa1+/- and 7 in WT). D: Caspase 3 activity amongst Opa1+/- (n = 7) and WT (n = 7) groups. E: mPTP opening sensitivity at baseline and after I/R (n = 6/group). Required calcium overload for mPTP opening in WT and Opa1+/- groups at baseline, after I/R, with or without use of cyclosporine analog (CsA). The I/R protocol included 45 minutes of ischemia and 2 hours of reperfusion. Values are mean ± SEM. *p<0.05, **p<0.01, ***p<0.001. $p< 0.05 compared to the same group at baseline + CsA. Non-parametric Mann-Whitney test (TUNEL and Bax/Bcl2 ratio) and t-test (Bax expression and caspase 3 activity).

Fig 6. Calcium transients and SR calcium…

Fig 6. Calcium transients and SR calcium load in Opa1 +/- isolated left-ventricular cardiomyocytes.

A :…
Fig 6. Calcium transients and SR calcium load in Opa1+/- isolated left-ventricular cardiomyocytes.
A: Typical calcium transients recorded under field stimulation at 1Hz in WT (black) and Opa1+/- (red) isolated left-ventricular cardiomyocytes using fluo-4 calcium dye. B: Mean values of peak calcium transients (WT, n = 32 cells and 4 animals, vs. Opa1+/-, n = 43 cells and 5 animals; *p<0.05). C: Mean values of decay time constant of steady-state Ca2+ transients (WT, n = 32 cells and 4 animals, vs. Opa1+/-, n = 43 cells and 5 animals; *p<0.05). D: Typical example of a caffeine-induced SR Ca2+ release in WT (black) and Opa1+/- (red) left-ventricular cardiomyocytes using fluo-4 calcium dye. E: Mean values of maximum amplitude of caffeine-induced SR Ca2+ release, indicative of SR Ca2+ load (WT, n = 7 cells and 4 animals, vs. Opa1+/-, n = 11 cells and 5 animals; p>0.05). F: Mean values of decay time constant of caffeine-induced SR Ca2+ release, indicative of cytosolic Ca2+ extrusion (WT, n = 7 cells and 4 animals, vs. Opa1+/-, n = 11 cells and 5 animals; *p<0.05). Statistical test was t-test.

Fig 7. Opa1 +/- left-ventricular cardiomyocyte AP.

A…

Fig 7. Opa1 +/- left-ventricular cardiomyocyte AP.

A : Typical AP recorded using whole-cell patch-clamp technique…
Fig 7. Opa1+/- left-ventricular cardiomyocyte AP.
A: Typical AP recorded using whole-cell patch-clamp technique with current clamp at 1Hz in WT (black) and Opa1+/- (red) isolated left-ventricular cardiomyocytes. B: Mean values of AP duration at different percentages of AP repolarization (WT: n = 12 cells and 3 animals vs. Opa1+/-: n = 12 cells and 4 animals; *p<0.05). C, D: Frame C displays 30 seconds of continuous AP recording at a pacing rate of 1Hz early after depolarization, whilst frame D shows a magnified view of the early post-depolarization period. Note that 6/12 Opa1+/- cells present such arrhythmic events, whereas WT myocytes underwent no early subsequent depolarization. Statistical test was t-test.

Fig 8. Dynamic mitochondrial Ca 2+ movements…

Fig 8. Dynamic mitochondrial Ca 2+ movements during APs recorded using Rhod-2 in conjunction with…

Fig 8. Dynamic mitochondrial Ca2+ movements during APs recorded using Rhod-2 in conjunction with a whole-cell patch-clamp technique.
A: Typical rhod-2 signal during a steady-state AP recorded at 1Hz in WT (black) and Opa1+/- (red) isolated left-ventricular cardiomyocytes. B: Mean values of peak rhod-2 signal. C: Rate of rise. D: Decay time constant in WT (n = 12 cells and 3 animals) and Opa1+/- cells (n = 12 cells and 4 animals; *p<0.05). Statistical test was t-test.
All figures (8)
Fig 4. Baseline and post-I/R oxidative phosphorylation.
Fig 4. Baseline and post-I/R oxidative phosphorylation.
Complex I, II, and IV oxygen consumption were measured in WT and Opa1+/- mouse hearts at baseline and after I/R (n = 6/group). The I/R protocol consisted of 45 minutes of ischemia and 2 hours of reperfusion. The ratio [state 3 rate]: [state 4 rate] is represented by means of the respiratory control index (RCI) in both groups at baseline and after I/R. Values are mean ± SEM. *p<0.05, **p<0.01, ***p<0.001.
Fig 5. Apoptosis assessment and sensitivity to…
Fig 5. Apoptosis assessment and sensitivity to mPTP.
A: Examples of TUNEL images of left-ventricular sections in confocal microscopy (70x magnification) after I/R. Cell nuclei are stained in red (propidium iodide, wavelength = 620 nm). TUNEL-positive nuclei appear in green (fluorescein, wavelength = 460 nm). B: Bax and Bcl-2 protein expressions (n = 4-6/group). C: Percentage of TUNEL-positive nuclei compared to total nuclei (n = 8 in Opa1+/- and 7 in WT). D: Caspase 3 activity amongst Opa1+/- (n = 7) and WT (n = 7) groups. E: mPTP opening sensitivity at baseline and after I/R (n = 6/group). Required calcium overload for mPTP opening in WT and Opa1+/- groups at baseline, after I/R, with or without use of cyclosporine analog (CsA). The I/R protocol included 45 minutes of ischemia and 2 hours of reperfusion. Values are mean ± SEM. *p<0.05, **p<0.01, ***p<0.001. $p< 0.05 compared to the same group at baseline + CsA. Non-parametric Mann-Whitney test (TUNEL and Bax/Bcl2 ratio) and t-test (Bax expression and caspase 3 activity).
Fig 6. Calcium transients and SR calcium…
Fig 6. Calcium transients and SR calcium load in Opa1+/- isolated left-ventricular cardiomyocytes.
A: Typical calcium transients recorded under field stimulation at 1Hz in WT (black) and Opa1+/- (red) isolated left-ventricular cardiomyocytes using fluo-4 calcium dye. B: Mean values of peak calcium transients (WT, n = 32 cells and 4 animals, vs. Opa1+/-, n = 43 cells and 5 animals; *p<0.05). C: Mean values of decay time constant of steady-state Ca2+ transients (WT, n = 32 cells and 4 animals, vs. Opa1+/-, n = 43 cells and 5 animals; *p<0.05). D: Typical example of a caffeine-induced SR Ca2+ release in WT (black) and Opa1+/- (red) left-ventricular cardiomyocytes using fluo-4 calcium dye. E: Mean values of maximum amplitude of caffeine-induced SR Ca2+ release, indicative of SR Ca2+ load (WT, n = 7 cells and 4 animals, vs. Opa1+/-, n = 11 cells and 5 animals; p>0.05). F: Mean values of decay time constant of caffeine-induced SR Ca2+ release, indicative of cytosolic Ca2+ extrusion (WT, n = 7 cells and 4 animals, vs. Opa1+/-, n = 11 cells and 5 animals; *p<0.05). Statistical test was t-test.
Fig 7. Opa1 +/- left-ventricular cardiomyocyte AP.
Fig 7. Opa1+/- left-ventricular cardiomyocyte AP.
A: Typical AP recorded using whole-cell patch-clamp technique with current clamp at 1Hz in WT (black) and Opa1+/- (red) isolated left-ventricular cardiomyocytes. B: Mean values of AP duration at different percentages of AP repolarization (WT: n = 12 cells and 3 animals vs. Opa1+/-: n = 12 cells and 4 animals; *p<0.05). C, D: Frame C displays 30 seconds of continuous AP recording at a pacing rate of 1Hz early after depolarization, whilst frame D shows a magnified view of the early post-depolarization period. Note that 6/12 Opa1+/- cells present such arrhythmic events, whereas WT myocytes underwent no early subsequent depolarization. Statistical test was t-test.
Fig 8. Dynamic mitochondrial Ca 2+ movements…
Fig 8. Dynamic mitochondrial Ca2+ movements during APs recorded using Rhod-2 in conjunction with a whole-cell patch-clamp technique.
A: Typical rhod-2 signal during a steady-state AP recorded at 1Hz in WT (black) and Opa1+/- (red) isolated left-ventricular cardiomyocytes. B: Mean values of peak rhod-2 signal. C: Rate of rise. D: Decay time constant in WT (n = 12 cells and 3 animals) and Opa1+/- cells (n = 12 cells and 4 animals; *p<0.05). Statistical test was t-test.

References

    1. Hall AR, Burke N, Dongworth RK, Hausenloy DJ. Mitochondrial fusion and fission proteins: novel therapeutic targets for combating cardiovascular disease. Br J Pharmacol 2014;171:1890–1906. 10.1111/bph.12516
    1. Youle RJ, van der Bliek AM. Mitochondrial fission, fusion, and stress. Science 2012;337:1062–1065. 10.1126/science.1219855
    1. Alexander C, Votruba M, Pesch UE, Thiselton DL, Mayer S, Moore A et al. OPA1, encoding a dynamin-related GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28. Nat Genet 2000;26:211–215. 10.1038/79944
    1. Delettre C, Lenaers G, Griffoin JM, Gigarel N, Lorenzo C, Belenguer P et al. Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy. Nat Genet 2000;26:207–210. 10.1038/79936
    1. Chao de la Barca JM, Prunier-Mirebeau D, Amati-Bonneau P, Ferre M, Sarzi E, Bris C, et al. OPA1-related disorders: Diversity of clinical expression, modes of inheritance and pathophysiology. Neurobiol Dis 2016;90:20–26. 10.1016/j.nbd.2015.08.015
    1. Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation 2010;121:2012–2022. 10.1161/CIRCULATIONAHA.109.906610
    1. Disatnik MH, Ferreira JC, Campos JC, Gomes KS, Dourado PM, Qi X et al. Acute inhibition of excessive mitochondrial fission after myocardial infarction prevents long-term cardiac dysfunction. J Am Heart Assoc 2013;2:e000461 10.1161/JAHA.113.000461
    1. Sharp WW, Fang YH, Han M, Zhang HJ, Hong Z, Banathy A et al. Dynamin-related protein 1 (Drp1)-mediated diastolic dysfunction in myocardial ischemia-reperfusion injury: therapeutic benefits of Drp1 inhibition to reduce mitochondrial fission. Faseb j 2014;28:316–326. 10.1096/fj.12-226225
    1. Zepeda R, Kuzmicic J, Parra V, Troncoso R, Pennanen C, Riquelme JA et al. Drp1 loss-of-function reduces cardiomyocyte oxygen dependence protecting the heart from ischemia-reperfusion injury. J Cardiovasc Pharmacol 2014;63:477–487. 10.1097/FJC.0000000000000071
    1. Papanicolaou KN, Khairallah RJ, Ngoh GA, Chikando A, Luptak I, O'Shea KM et al. Mitofusin-2 maintains mitochondrial structure and contributes to stress-induced permeability transition in cardiac myocytes. Mol Cell Biol 2011;31:1309–1328. 10.1128/MCB.00911-10
    1. Chen L, Gong Q, Stice JP, Knowlton AA. Mitochondrial OPA1, apoptosis, and heart failure. Cardiovasc Res 2009;84:91–99. 10.1093/cvr/cvp181
    1. Chen L, Liu T, Tran A, Lu X, Tomilov AA, Davies V et al. OPA1 mutation and late-onset cardiomyopathy: mitochondrial dysfunction and mtDNA instability. J Am Heart Assoc 2012;1:e003012 10.1161/JAHA.112.003012
    1. Piquereau J, Caffin F, Novotova M, Prola A, Garnier A, Mateo P et al. Down-regulation of OPA1 alters mouse mitochondrial morphology, PTP function, and cardiac adaptation to pressure overload. Cardiovasc Res 2012;94:408–417. 10.1093/cvr/cvs117
    1. Ferre M, Caignard A, Milea D, Leruez S, Cassereau J, Chevrollier A et al. Improved locus-specific database for OPA1 mutations allows inclusion of advanced clinical data. Hum Mutat 2015;36:20–25. 10.1002/humu.22703
    1. Sarzi E, Angebault C, Seveno M, Gueguen N, Chaix B, Bielicki G et al. The human OPA1delTTAG mutation induces premature age-related systemic neurodegeneration in mouse. Brain 2012;135:3599–3613. 10.1093/brain/aws303
    1. Prunier F, Gaertner R, Louedec L, Michel JB, Mercadier JJ, Escoubet B. Doppler echocardiographic estimation of left ventricular end-diastolic pressure after MI in rats. Am J Physiol Heart Circ Physiol 2002;283:H346–352. 10.1152/ajpheart.01050.2001
    1. Fauconnier J, Lanner JT, Zhang SJ, Tavi P, Bruton JD, Katz A et al. Insulin and inositol 1,4,5-trisphosphate trigger abnormal cytosolic Ca2+ transients and reveal mitochondrial Ca2+ handling defects in cardiomyocytes of ob/ob mice. Diabetes 2005;54:2375–2381. 10.2337/diabetes.54.8.2375
    1. Paillard M, Tubbs E, Thiebaut PA, Gomez L, Fauconnier J, Da Silva CC et al. Depressing mitochondria-reticulum interactions protects cardiomyocytes from lethal hypoxia-reoxygenation injury. Circulation 2013;128:1555–1565. 10.1161/CIRCULATIONAHA.113.001225
    1. Dague E, Genet G, Lachaize V, Guilbeau-Frugier C, Fauconnier J, Mias C et al. Atomic force and electron microscopic-based study of sarcolemmal surface of living cardiomyocytes unveils unexpected mitochondrial shift in heart failure. J Mol Cell Cardiol 2014;74:162–172. 10.1016/j.yjmcc.2014.05.006
    1. Fauconnier J, Bedut S, Le Guennec JY, Babuty D, Richard S. Ca2+ current-mediated regulation of action potential by pacing rate in rat ventricular myocytes. Cardiovasc Res 2003;57:670–680. 10.1016/S0008-6363(02)00731-9
    1. Wilding JR, Joubert F, de Araujo C, Fortin D, Novotova M, Veksler V et al. Altered energy transfer from mitochondria to sarcoplasmic reticulum after cytoarchitectural perturbations in mice hearts. J Physiol 2006;575:191–200. 10.1113/jphysiol.2006.114116
    1. Teixeira G, Chiari P, Fauconnier J, Abrial M, Couture-Lepetit E, Harisseh R et al. Involvement of Cyclophilin D and Calcium in Isoflurane-induced Preconditioning. Anesthesiology 2015;123:1374–1384. 10.1097/ALN.0000000000000876
    1. Gharib A, De Paulis D, Li B, Augeul L, Couture-Lepetit E, Gomez L et al. Opposite and tissue-specific effects of coenzyme Q2 on mPTP opening and ROS production between heart and liver mitochondria: role of complex I. J Mol Cell Cardiol 2012;52:1091–1095. 10.1016/j.yjmcc.2012.02.005
    1. Kalakech H, Tamareille S, Pons S, Godin-Ribuot D, Carmeliet P, Furber A et al. Role of hypoxia inducible factor-1alpha in remote limb ischemic preconditioning. J Mol Cell Cardiol 2013;65:98–104. 10.1016/j.yjmcc.2013.10.001
    1. Tamareille S, Mateus V, Ghaboura N, Jeanneteau J, Croue A, Henrion D et al. RISK and SAFE signaling pathway interactions in remote limb ischemic perconditioning in combination with local ischemic postconditioning. Basic Res Cardiol 2011;106:1329–1339. 10.1007/s00395-011-0210-z
    1. Heusch G. Molecular basis of cardioprotection: signal transduction in ischemic pre-, post-, and remote conditioning. Circ Res 2015;116:674–699. 10.1161/CIRCRESAHA.116.305348
    1. Gomez L, Li B, Mewton N, Sanchez I, Piot C, Elbaz M et al. Inhibition of mitochondrial permeability transition pore opening: translation to patients. Cardiovasc Res 2009;83:226–233. 10.1093/cvr/cvp063
    1. Heusch G, Boengler K, Schulz R. Inhibition of mitochondrial permeability transition pore opening: the Holy Grail of cardioprotection. Basic Res Cardiol 2010;105:151–154. 10.1007/s00395-009-0080-9
    1. Brady NR, Hamacher-Brady A, Gottlieb RA. Proapoptotic BCL-2 family members and mitochondrial dysfunction during ischemia/reperfusion injury, a study employing cardiac HL-1 cells and GFP biosensors. Biochim Biophys Acta 2006;1757:667–678. 10.1016/j.bbabio.2006.04.011
    1. Spiegel R, Saada A, Flannery PJ, Burte F, Soiferman D, Khayat M et al. Fatal infantile mitochondrial encephalomyopathy, hypertrophic cardiomyopathy and optic atrophy associated with a homozygous OPA1 mutation. J Med Genet 2016;53:127–131. 10.1136/jmedgenet-2015-103361
    1. Fauconnier J, Roberge S, Saint N, Lacampagne A. Type 2 ryanodine receptor: a novel therapeutic target in myocardial ischemia/reperfusion. Pharmacol Ther 2013;138:323–332. 10.1016/j.pharmthera.2013.01.015
    1. Prunier F, Kawase Y, Gianni D, Scapin C, Danik SB, Ellinor PT et al. Prevention of ventricular arrhythmias with sarcoplasmic reticulum Ca2+ ATPase pump overexpression in a porcine model of ischemia reperfusion. Circulation 2008;118:614–624. 10.1161/CIRCULATIONAHA.108.770883
    1. Bogeholz N, Pauls P, Bauer BK, Schulte JS, Dechering DG, Frommeyer G et al. Suppression of Early and Late Afterdepolarizations by Heterozygous Knockout of the Na+/Ca2+ Exchanger in a Murine Model. Circ Arrhythm Electrophysiol 2015;8:1210–1218. 10.1161/CIRCEP.115.002927
    1. Yao A, Su Z, Nonaka A, Zubair I, Lu L, Philipson KD et al. Effects of overexpression of the Na+-Ca2+ exchanger on [Ca2+]i transients in murine ventricular myocytes. Circ Res 1998;82:657–665. 10.1161/01.RES.82.6.657
    1. Pott C, Eckardt L, Goldhaber JI. Triple threat: the Na+/Ca2+ exchanger in the pathophysiology of cardiac arrhythmia, ischemia and heart failure. Curr Drug Targets 2011;12:737–747. 10.2174/138945011795378559
    1. Qu Z, Xie LH, Olcese R, Karagueuzian HS, Chen PS, Garfinkel A et al. Early afterdepolarizations in cardiac myocytes: beyond reduced repolarization reserve. Cardiovasc Res 2013;99:6–15. 10.1093/cvr/cvt104
    1. Bers DM. Calcium fluxes involved in control of cardiac myocyte contraction. Circ Res 2000;87:275–281. 10.1161/01.RES.87.4.275
    1. Varanita T, Soriano ME, Romanello V, Zaglia T, Quintana-Cabrera R, Semenzato M et al. The OPA1-dependent mitochondrial cristae remodeling pathway controls atrophic, apoptotic, and ischemic tissue damage. Cell Metab 2015;21:834–844. 10.1016/j.cmet.2015.05.007
    1. Ikeda Y, Shirakabe A, Maejima Y, Zhai P, Sciarretta S, Toli J et al. Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res 2015;116:264–278. 10.1161/CIRCRESAHA.116.303356

Source: PubMed

3
Předplatit