Skip to main content
Log in

Sex Differences in Ischemia/Reperfusion Injury: The Role of Mitochondrial Permeability Transition

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Brain and heart ischemia are among the leading causes of death and disability in both men and women, but there are significant sex differences in the incidence and severity of these diseases. Ca2+ dysregulation in response to ischemia/reperfusion injury (I/RI) is a well-recognized pathogenic mechanism leading to the death of affected cells. Excess intracellular Ca2+ causes mitochondrial matrix Ca2+ overload that can result in mitochondrial permeability transition (MPT), which can have severe consequences for mitochondrial function and trigger cell death. Recent findings indicate that estrogens and their related receptors are involved in the regulation of MPT, suggesting that sex differences in I/RI could be linked to estrogen-dependent modulation of mitochondrial Ca2+. Here, we review the evidence supporting sex differences in I/RI and the role of estrogen and estrogen receptors in producing these differences, the involvement of mitochondrial Ca2+ overload in disease pathogenesis, and the estrogen-dependent modulation of MPT that may contribute to sex differences.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Ostadal B, Ostadal P (2014) Sex-based differences in cardiac ischaemic injury and protection: therapeutic implications. Br J Pharmacol 171:541–554

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Gibson CL, Attwood L (2016) The impact of gender on stroke pathology and treatment. Neurosci Biobehav Rev 67:119–124

    PubMed  Google Scholar 

  3. Spychala MS, Honarpisheh P, McCullough LD (2017) Sex differences in neuroinflammation and neuroprotection in ischemic stroke. J Neurosci Res 95:462–471

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Herson PS, Palmateer J, Hurn PD (2013) Biological sex and mechanisms of ischemic brain injury. Transl Stroke Res 4:413–419

    PubMed  Google Scholar 

  5. Haast RA, Gustafson DR, Kiliaan AJ (2012) Sex differences in stroke. J Cereb Blood Flow Metab 32:2100–2107

    PubMed  PubMed Central  Google Scholar 

  6. Ahnstedt H, McCullough LD, Cipolla MJ (2016) The importance of considering sex differences in translational stroke research. Transl Stroke Res 7:261–273

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Gibson CL (2013) Cerebral ischemic stroke: is gender important? J Cereb Blood Flow Metab 33:1355–1361

    PubMed  PubMed Central  Google Scholar 

  8. Chauhan A, Moser H, McCullough LD (2017) Sex differences in ischaemic stroke: potential cellular mechanisms. Clin Sci (Lond) 131:533–552

    CAS  Google Scholar 

  9. Girijala RL, Sohrabji F, Bush RL (2017) Sex differences in stroke: review of current knowledge and evidence. Vasc Med 22:135–145

    PubMed  Google Scholar 

  10. Appelros P, Stegmayr B, Terent A (2009) Sex differences in stroke epidemiology: a systematic review. Stroke 40:1082–1090

    Google Scholar 

  11. Di Carlo A, Lamassa M, Baldereschi M, Pracucci G, Basile AM, Wolfe CD, Giroud M, Rudd A, Ghetti A, Inzitari D, European BSoSCG (2003) Sex differences in the clinical presentation, resource use, and 3-month outcome of acute stroke in Europe: data from a multicenter multinational hospital-based registry. Stroke 34:1114–1119

    PubMed  Google Scholar 

  12. Dodson JA, Arnold SV, Reid KJ, Gill TM, Rich MW, Masoudi FA, Spertus JA, Krumholz HM, Alexander KP (2012) Physical function and independence 1 year after myocardial infarction: observations from the translational research investigating underlying disparities in recovery from acute myocardial infarction: patients’ health status registry. Am Heart J 163:790–796

    PubMed  PubMed Central  Google Scholar 

  13. Humphries KH, Izadnegahdar M, Sedlak T, Saw J, Johnston N, Schenck-Gustafsson K, Shah RU, Regitz-Zagrosek V, Grewal J, Vaccarino V, Wei J, Bairey Merz CN (2017) Sex differences in cardiovascular disease—impact on care and outcomes. Front Neuroendocrinol 46:46–70

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhu C, Xu F, Wang X, Shibata M, Uchiyama Y, Blomgren K, Hagberg H (2006) Different apoptotic mechanisms are activated in male and female brains after neonatal hypoxia-ischaemia. J Neurochem 96:1016–1027

    CAS  PubMed  Google Scholar 

  15. Hurn PD, Vannucci SJ, Hagberg H (2005) Adult or perinatal brain injury: does sex matter? Stroke 36:193–195

    PubMed  Google Scholar 

  16. DiNapoli VA, Huber JD, Houser K, Li X, Rosen CL (2008) Early disruptions of the blood-brain barrier may contribute to exacerbated neuronal damage and prolonged functional recovery following stroke in aged rats. Neurobiol Aging 29:753–764

    CAS  PubMed  Google Scholar 

  17. Johnson MS, Moore RL, Brown DA (2006) Sex differences in myocardial infarct size are abolished by sarcolemmal KATP channel blockade in rat. Am J Physiol Heart Circ Physiol 290:H2644–H2647

    CAS  PubMed  Google Scholar 

  18. Booth EA, Lucchesi BR (2008) Estrogen-mediated protection in myocardial ischemia-reperfusion injury. Cardiovasc Toxicol 8:101–113

    CAS  PubMed  Google Scholar 

  19. Rocca WA, Grossardt BR, Miller VM, Shuster LT, Brown RD Jr (2012) Premature menopause or early menopause and risk of ischemic stroke. Menopause 19:272–277

    PubMed  PubMed Central  Google Scholar 

  20. Lisabeth LD, Beiser AS, Brown DL, Murabito JM, Kelly-Hayes M, Wolf PA (2009) Age at natural menopause and risk of ischemic stroke: the Framingham heart study. Stroke 40:1044–1049

    PubMed  PubMed Central  Google Scholar 

  21. Harman SM, Naftolin F, Brinton EA, Judelson DR (2005) Is the estrogen controversy over? deconstructing the women’s health initiative study: a critical evaluation of the evidence. Ann N Y Acad Sci 1052:43–56

    CAS  PubMed  Google Scholar 

  22. Simpkins JW, Rajakumar G, Zhang YQ, Simpkins CE, Greenwald D, Yu CJ, Bodor N, Day AL (1997) Estrogens may reduce mortality and ischemic damage caused by middle cerebral artery occlusion in the female rat. J Neurosurg 87:724–730

    CAS  PubMed  Google Scholar 

  23. Carswell HV, Dominiczak AF, Macrae IM (2000) Estrogen status affects sensitivity to focal cerebral ischemia in stroke-prone spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 278:H290–H294

    CAS  PubMed  Google Scholar 

  24. Suzuki S, Brown CM, Wise PM (2009) Neuroprotective effects of estrogens following ischemic stroke. Front Neuroendocrinol 30:201–211

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Ross JL, Howlett SE (2012) Age and ovariectomy abolish beneficial effects of female sex on rat ventricular myocytes exposed to simulated ischemia and reperfusion. PLoS ONE 7:e38425

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Hale SL, Birnbaum Y, Kloner RA (1997) Estradiol, administered acutely, protects ischemic myocardium in both female and male rabbits. J Cardiovasc Pharmacol Ther 2:47–52

    CAS  PubMed  Google Scholar 

  27. Miller NR, Jover T, Cohen HW, Zukin RS, Etgen AM (2005) Estrogen can act via estrogen receptor alpha and beta to protect hippocampal neurons against global ischemia-induced cell death. Endocrinology 146:3070–3079

    CAS  PubMed  Google Scholar 

  28. Booth EA, Marchesi M, Kilbourne EJ, Lucchesi BR (2003) 17Beta-estradiol as a receptor-mediated cardioprotective agent. J Pharmacol Exp Ther 307:395–401

    CAS  PubMed  Google Scholar 

  29. Bopassa JC, Eghbali M, Toro L, Stefani E (2010) A novel estrogen receptor GPER inhibits mitochondria permeability transition pore opening and protects the heart against ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 298:H16–H23

    CAS  PubMed  Google Scholar 

  30. Kabir ME, Singh H, Lu R, Olde B, Leeb-Lundberg LM, Bopassa JC (2015) G protein-coupled estrogen receptor 1 mediates acute estrogen-induced cardioprotection via MEK/ERK/GSK-3beta pathway after ischemia/reperfusion. PLoS ONE 10:e0135988

    PubMed  PubMed Central  Google Scholar 

  31. Choi DW (1985) Glutamate neurotoxicity in cortical cell culture is calcium dependent. Neurosci Lett 58:293–297

    CAS  PubMed  Google Scholar 

  32. Brewer GJ, Reichensperger JD, Brinton RD (2006) Prevention of age-related dysregulation of calcium dynamics by estrogen in neurons. Neurobiol Aging 27:306–317

    CAS  PubMed  Google Scholar 

  33. Burstein SR, Kim HJ, Fels JA, Qian L, Zhang S, Zhou P, Starkov AA, Iadecola C, Manfredi G (2018) Estrogen receptor beta modulates permeability transition in brain mitochondria. Biochim Biophys Acta Bioenerg 1859:423–433

    CAS  PubMed  Google Scholar 

  34. Mendelowitsch A, Ritz MF, Ros J, Langemann H, Gratzl O (2001) 17beta-estradiol reduces cortical lesion size in the glutamate excitotoxicity model by enhancing extracellular lactate: a new neuroprotective pathway. Brain Res 901:230–236

    CAS  PubMed  Google Scholar 

  35. Zhao L, Brinton RD (2007) Estrogen receptor alpha and beta differentially regulate intracellular Ca(2+) dynamics leading to ERK phosphorylation and estrogen neuroprotection in hippocampal neurons. Brain Res 1172:48–59

    CAS  PubMed  Google Scholar 

  36. Yang SH, Sarkar SN, Liu R, Perez EJ, Wang X, Wen Y, Yan LJ, Simpkins JW (2009) Estrogen receptor beta as a mitochondrial vulnerability factor. J Biol Chem 284:9540–9548

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Jovanovic S, Jovanovic A, Shen WK, Terzic A (2000) Low concentrations of 17beta-estradiol protect single cardiac cells against metabolic stress-induced Ca2+ loading. J Am Coll Cardiol 36:948–952

    CAS  PubMed  Google Scholar 

  38. Tymianski M, Charlton MP, Carlen PL, Tator CH (1993) Source specificity of early calcium neurotoxicity in cultured embryonic spinal neurons. J Neurosci 13:2085–2104

    CAS  PubMed  Google Scholar 

  39. Peng TI, Greenamyre JT (1998) Privileged access to mitochondria of calcium influx through N-methyl-D-aspartate receptors. Mol Pharmacol 53:974–980

    CAS  PubMed  Google Scholar 

  40. Cross JL, Meloni BP, Bakker AJ, Lee S, Knuckey NW (2010) Modes of neuronal calcium entry and homeostasis following cerebral ischemia. Stroke Res Treat 2010:316862

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Garcia-Dorado D, Ruiz-Meana M, Inserte J, Rodriguez-Sinovas A, Piper HM (2012) Calcium-mediated cell death during myocardial reperfusion. Cardiovasc Res 94:168–180

    CAS  PubMed  Google Scholar 

  42. Nayler WG (1981) The role of calcium in the ischemic myocardium. Am J Pathol 102:262–270

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Chiong M, Wang ZV, Pedrozo Z, Cao DJ, Troncoso R, Ibacache M, Criollo A, Nemchenko A, Hill JA, Lavandero S (2011) Cardiomyocyte death: mechanisms and translational implications. Cell Death Dis 2:e244

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Pallafacchina G, Zanin S, Rizzuto R (2018) Recent advances in the molecular mechanism of mitochondrial calcium uptake. F1000Research. https://doi.org/10.12688/f1000research.15723.1

    Article  PubMed  PubMed Central  Google Scholar 

  45. Bernardi P, von Stockum S (2012) The permeability transition pore as a Ca(2+) release channel: new answers to an old question. Cell Calcium 52:22–27

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Duchen MR (2012) Mitochondria, calcium-dependent neuronal death and neurodegenerative disease. Pflugers Arch 464:111–121

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Gouriou Y, Demaurex N, Bijlenga P, De Marchi U (2011) Mitochondrial calcium handling during ischemia-induced cell death in neurons. Biochimie 93:2060–2067

    CAS  PubMed  Google Scholar 

  48. Stout AK, Raphael HM, Kanterewicz BI, Klann E, Reynolds IJ (1998) Glutamate-induced neuron death requires mitochondrial calcium uptake. Nat Neurosci 1:366–373

    CAS  PubMed  Google Scholar 

  49. Castilho RF, Hansson O, Ward MW, Budd SL, Nicholls DG (1998) Mitochondrial control of acute glutamate excitotoxicity in cultured cerebellar granule cells. J Neurosci 18:10277–10286

    CAS  PubMed  Google Scholar 

  50. Vergun O, Keelan J, Khodorov BI, Duchen MR (1999) Glutamate-induced mitochondrial depolarisation and perturbation of calcium homeostasis in cultured rat hippocampal neurones. J Physiol 519(2):451–466

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Hunter DR, Haworth RA (1979) The Ca2+-induced membrane transition in mitochondria: III. Transitional Ca2+ release. Arch Biochem Biophys 195:468–477

    CAS  PubMed  Google Scholar 

  52. Szabo I, Zoratti M (1992) The mitochondrial megachannel is the permeability transition pore. J Bioenerg Biomembr 24:111–117

    CAS  PubMed  Google Scholar 

  53. Crompton M (1999) The mitochondrial permeability transition pore and its role in cell death. Biochem J 341(2):233–249

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Halestrap AP, Richardson AP (2015) The mitochondrial permeability transition: a current perspective on its identity and role in ischaemia/reperfusion injury. J Mol Cell Cardiol 78:129–141

    CAS  PubMed  Google Scholar 

  55. Petronilli V, Penzo D, Scorrano L, Bernardi P, Di Lisa F (2001) The mitochondrial permeability transition, release of cytochrome c and cell death. Correlation with the duration of pore openings in situ. J Biol Chem 276:12030–12034

    CAS  PubMed  Google Scholar 

  56. Giorgio V, Guo L, Bassot C, Petronilli V, Bernardi P (2018) Calcium and regulation of the mitochondrial permeability transition. Cell Calcium 70:56–63

    CAS  PubMed  Google Scholar 

  57. Petronilli V, Cola C, Bernardi P (1993) Modulation of the mitochondrial cyclosporin A-sensitive permeability transition pore. II. The minimal requirements for pore induction underscore a key role for transmembrane electrical potential, matrix pH, and matrix Ca2+. J Biol Chem 268:1011–1016

    CAS  PubMed  Google Scholar 

  58. Doczi J, Turiak L, Vajda S, Mandi M, Torocsik B, Gerencser AA, Kiss G, Konrad C, Adam-Vizi V, Chinopoulos C (2011) Complex contribution of cyclophilin D to Ca2+-induced permeability transition in brain mitochondria, with relation to the bioenergetic state. J Biol Chem 286:6345–6353

    CAS  PubMed  Google Scholar 

  59. Zoratti M, Szabo I (1995) The mitochondrial permeability transition. Biochim Biophys Acta 1241:139–176

    PubMed  Google Scholar 

  60. Crompton M, Virji S, Ward JM (1998) Cyclophilin-D binds strongly to complexes of the voltage-dependent anion channel and the adenine nucleotide translocase to form the permeability transition pore. Eur J Biochem 258:729–735

    CAS  PubMed  Google Scholar 

  61. Kokoszka JE, Waymire KG, Levy SE, Sligh JE, Cai J, Jones DP, MacGregor GR, Wallace DC (2004) The ADP/ATP translocator is not essential for the mitochondrial permeability transition pore. Nature 427:461–465

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Doczi J, Torocsik B, Echaniz-Laguna A, Mousson de Camaret B, Starkov A, Starkova N, Gal A, Molnar MJ, Kawamata H, Manfredi G, Adam-Vizi V, Chinopoulos C (2016) Alterations in voltage-sensing of the mitochondrial permeability transition pore in ANT1-deficient cells. Sci Rep 6:26700

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Alavian KN, Beutner G, Lazrove E, Sacchetti S, Park HA, Licznerski P, Li H, Nabili P, Hockensmith K, Graham M, Porter GA Jr, Jonas EA (2014) An uncoupling channel within the c-subunit ring of the F1FO ATP synthase is the mitochondrial permeability transition pore. Proc Natl Acad Sci USA 111:10580–10585

    CAS  PubMed  Google Scholar 

  64. Bonora M, Morganti C, Morciano G, Pedriali G, Lebiedzinska-Arciszewska M, Aquila G, Giorgi C, Rizzo P, Campo G, Ferrari R, Kroemer G, Wieckowski MR, Galluzzi L, Pinton P (2017) Mitochondrial permeability transition involves dissociation of F1FO ATP synthase dimers and C-ring conformation. EMBO Rep 18:1077–1089

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhou W, Marinelli F, Nief C, Faraldo-Gomez JD (2017) Atomistic simulations indicate the c-subunit ring of the F1Fo ATP synthase is not the mitochondrial permeability transition pore. Elife 6:e23781

    PubMed  PubMed Central  Google Scholar 

  66. Giorgio V, von Stockum S, Antoniel M, Fabbro A, Fogolari F, Forte M, Glick GD, Petronilli V, Zoratti M, Szabo I, Lippe G, Bernardi P (2013) Dimers of mitochondrial ATP synthase form the permeability transition pore. Proc Natl Acad Sci USA 110:5887–5892

    CAS  PubMed  Google Scholar 

  67. Bernardi P, Di Lisa F, Fogolari F, Lippe G (2015) From ATP to PTP and back: a dual function for the mitochondrial ATP synthase. Circ Res 116:1850–1862

    CAS  PubMed  PubMed Central  Google Scholar 

  68. He J, Ford HC, Carroll J, Ding S, Fearnley IM, Walker JE (2017) Persistence of the mitochondrial permeability transition in the absence of subunit c of human ATP synthase. Proc Natl Acad Sci USA 114:3409–3414

    CAS  PubMed  Google Scholar 

  69. He J, Carroll J, Ding S, Fearnley IM, Walker JE (2017) Permeability transition in human mitochondria persists in the absence of peripheral stalk subunits of ATP synthase. Proc Natl Acad Sci USA 114:9086–9091

    CAS  PubMed  Google Scholar 

  70. Giorgio V, Soriano ME, Basso E, Bisetto E, Lippe G, Forte MA, Bernardi P (2010) Cyclophilin D in mitochondrial pathophysiology. Biochim Biophys Acta 1797:1113–1118

    CAS  PubMed  Google Scholar 

  71. Schinzel AC, Takeuchi O, Huang Z, Fisher JK, Zhou Z, Rubens J, Hetz C, Danial NN, Moskowitz MA, Korsmeyer SJ (2005) Cyclophilin D is a component of mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia. Proc Natl Acad Sci USA 102:12005–12010

    CAS  PubMed  Google Scholar 

  72. Basso E, Fante L, Fowlkes J, Petronilli V, Forte MA, Bernardi P (2005) Properties of the permeability transition pore in mitochondria devoid of cyclophilin D. J Biol Chem 280:18558–18561

    CAS  PubMed  Google Scholar 

  73. Izzo V, Bravo-San Pedro JM, Sica V, Kroemer G, Galluzzi L (2016) Mitochondrial permeability transition: new findings and persisting uncertainties. Trends Cell Biol 26:655–667

    CAS  PubMed  Google Scholar 

  74. Crompton M, Ellinger H, Costi A (1988) Inhibition by cyclosporin A of a Ca2+-dependent pore in heart mitochondria activated by inorganic phosphate and oxidative stress. Biochem J 255:357–360

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Hansson MJ, Morota S, Chen L, Matsuyama N, Suzuki Y, Nakajima S, Tanoue T, Omi A, Shibasaki F, Shimazu M, Ikeda Y, Uchino H, Elmer E (2011) Cyclophilin D-sensitive mitochondrial permeability transition in adult human brain and liver mitochondria. J Neurotrauma 28:143–153

    PubMed  PubMed Central  Google Scholar 

  76. Diaz-Ruiz A, Vergara P, Perez-Severiano F, Segovia J, Guizar-Sahagun G, Ibarra A, Rios C (2005) Cyclosporin-A inhibits constitutive nitric oxide synthase activity and neuronal and endothelial nitric oxide synthase expressions after spinal cord injury in rats. Neurochem Res 30:245–251

    CAS  PubMed  Google Scholar 

  77. Gauba E, Guo L, Du H (2017) Cyclophilin D promotes brain mitochondrial F1FO ATP synthase dysfunction in aging mice. J Alzheimers Dis 55:1351–1362

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Giorgio V, Bisetto E, Soriano ME, Dabbeni-Sala F, Basso E, Petronilli V, Forte MA, Bernardi P, Lippe G (2009) Cyclophilin D modulates mitochondrial F0F1-ATP synthase by interacting with the lateral stalk of the complex. J Biol Chem 284:33982–33988

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Chinopoulos C, Konrad C, Kiss G, Metelkin E, Torocsik B, Zhang SF, Starkov AA (2011) Modulation of F0F1-ATP synthase activity by cyclophilin D regulates matrix adenine nucleotide levels. FEBS J 278:1112–1125

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Beck SJ, Guo L, Phensy A, Tian J, Wang L, Tandon N, Gauba E, Lu L, Pascual JM, Kroener S, Du H (2016) Deregulation of mitochondrial F1FO-ATP synthase via OSCP in Alzheimer’s disease. Nat Commun 7:11483

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Bernardi P, Petronilli V (1996) The permeability transition pore as a mitochondrial calcium release channel: a critical appraisal. J Bioenerg Biomembr 28:131–138

    CAS  PubMed  Google Scholar 

  82. Ichas F, Jouaville LS, Mazat JP (1997) Mitochondria are excitable organelles capable of generating and conveying electrical and calcium signals. Cell 89:1145–1153

    CAS  PubMed  Google Scholar 

  83. Ichas F, Mazat JP (1998) From calcium signaling to cell death: two conformations for the mitochondrial permeability transition pore. Switching from low- to high-conductance state. Biochim Biophys Acta 1366:33–50

    CAS  PubMed  Google Scholar 

  84. Huser J, Blatter LA (1999) Fluctuations in mitochondrial membrane potential caused by repetitive gating of the permeability transition pore. Biochem J 343(2):311–317

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Wang W, Fang H, Groom L, Cheng A, Zhang W, Liu J, Wang X, Li K, Han P, Zheng M, Yin J, Wang W, Mattson MP, Kao JP, Lakatta EG, Sheu SS, Ouyang K, Chen J, Dirksen RT, Cheng H (2008) Superoxide flashes in single mitochondria. Cell 134:279–290

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Agarwal A, Wu PH, Hughes EG, Fukaya M, Tischfield MA, Langseth AJ, Wirtz D, Bergles DE (2017) Transient opening of the mitochondrial permeability transition pore induces microdomain calcium transients in astrocyte processes. Neuron 93:587–605 (e587)

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Korge P, Yang L, Yang JH, Wang Y, Qu Z, Weiss JN (2011) Protective role of transient pore openings in calcium handling by cardiac mitochondria. J Biol Chem 286:34851–34857

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Altschuld RA, Hohl CM, Castillo LC, Garleb AA, Starling RC, Brierley GP (1992) Cyclosporin inhibits mitochondrial calcium efflux in isolated adult rat ventricular cardiomyocytes. Am J Physiol 262:H1699–H1704

    CAS  PubMed  Google Scholar 

  89. De Marchi E, Bonora M, Giorgi C, Pinton P (2014) The mitochondrial permeability transition pore is a dispensable element for mitochondrial calcium efflux. Cell Calcium 56:1–13

    PubMed  PubMed Central  Google Scholar 

  90. Lemasters JJ, Nieminen AL, Qian T, Trost LC, Elmore SP, Nishimura Y, Crowe RA, Cascio WE, Bradham CA, Brenner DA, Herman B (1998) The mitochondrial permeability transition in cell death: a common mechanism in necrosis, apoptosis and autophagy. Biochim Biophys Acta 1366:177–196

    CAS  PubMed  Google Scholar 

  91. Brown MR, Sullivan PG, Geddes JW (2006) Synaptic mitochondria are more susceptible to Ca2+ overload than nonsynaptic mitochondria. J Biol Chem 281:11658–11668

    CAS  PubMed  Google Scholar 

  92. Naga KK, Sullivan PG, Geddes JW (2007) High cyclophilin D content of synaptic mitochondria results in increased vulnerability to permeability transition. J Neurosci 27:7469–7475

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Porter GA Jr, Beutner G (2018) Cyclophilin D, somehow a master regulator of mitochondrial function. Biomolecules 8:176

    PubMed Central  Google Scholar 

  94. Lemasters JJ, Theruvath TP, Zhong Z, Nieminen AL (2009) Mitochondrial calcium and the permeability transition in cell death. Biochim Biophys Acta 1787:1395–1401

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Rasola A, Bernardi P (2007) The mitochondrial permeability transition pore and its involvement in cell death and in disease pathogenesis. Apoptosis 12:815–833

    CAS  PubMed  Google Scholar 

  96. Starkov AA, Chinopoulos C, Fiskum G (2004) Mitochondrial calcium and oxidative stress as mediators of ischemic brain injury. Cell Calcium 36:257–264

    CAS  PubMed  Google Scholar 

  97. Shiga Y, Onodera H, Matsuo Y, Kogure K (1992) Cyclosporin A protects against ischemia-reperfusion injury in the brain. Brain Res 595:145–148

    CAS  PubMed  Google Scholar 

  98. Uchino H, Elmer E, Uchino K, Lindvall O, Siesjo BK (1995) Cyclosporin A dramatically ameliorates CA1 hippocampal damage following transient forebrain ischaemia in the rat. Acta Physiol Scand 155:469–471

    CAS  PubMed  Google Scholar 

  99. Argaud L, Gateau-Roesch O, Muntean D, Chalabreysse L, Loufouat J, Robert D, Ovize M (2005) Specific inhibition of the mitochondrial permeability transition prevents lethal reperfusion injury. J Mol Cell Cardiol 38:367–374

    CAS  PubMed  Google Scholar 

  100. Nakagawa T, Shimizu S, Watanabe T, Yamaguchi O, Otsu K, Yamagata H, Inohara H, Kubo T, Tsujimoto Y (2005) Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature 434:652–658

    CAS  PubMed  Google Scholar 

  101. Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, Hambleton MA, Brunskill EW, Sayen MR, Gottlieb RA, Dorn GW, Robbins J, Molkentin JD (2005) Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 434:658–662

    CAS  PubMed  Google Scholar 

  102. Tsujimoto Y, Shimizu S (2007) Role of the mitochondrial membrane permeability transition in cell death. Apoptosis 12:835–840

    CAS  PubMed  Google Scholar 

  103. Simpkins JW, Dykens JA (2008) Mitochondrial mechanisms of estrogen neuroprotection. Brain Res Rev 57:421–430

    CAS  PubMed  Google Scholar 

  104. Lobaton CD, Vay L, Hernandez-Sanmiguel E, Santodomingo J, Moreno A, Montero M, Alvarez J (2005) Modulation of mitochondrial Ca(2+) uptake by estrogen receptor agonists and antagonists. Br J Pharmacol 145:862–871

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Horvat A, Petrovic S, Nedeljkovic N, Martinovic JV, Nikezic G (2000) Estradiol affect Na-dependent Ca2+ efflux from synaptosomal mitochondria. Gen Physiol Biophys 19:59–71

    CAS  PubMed  Google Scholar 

  106. Nilsen J, Diaz Brinton R (2003) Mechanism of estrogen-mediated neuroprotection: regulation of mitochondrial calcium and Bcl-2 expression. Proc Natl Acad Sci USA 100:2842–2847

    CAS  PubMed  Google Scholar 

  107. Kim HJ, Magrane J, Starkov AA, Manfredi G (2012) The mitochondrial calcium regulator cyclophilin D is an essential component of oestrogen-mediated neuroprotection in amyotrophic lateral sclerosis. Brain 135:2865–2874

    PubMed  PubMed Central  Google Scholar 

  108. Ribeiro RF Jr, Ronconi KS, Morra EA, Do Val Lima PR, Porto ML, Vassallo DV, Figueiredo SG, Stefanon I (2016) Sex differences in the regulation of spatially distinct cardiac mitochondrial subpopulations. Mol Cell Biochem 419:41–51

    CAS  PubMed  Google Scholar 

  109. Milerova M, Drahota Z, Chytilova A, Tauchmannova K, Houstek J, Ostadal B (2016) Sex difference in the sensitivity of cardiac mitochondrial permeability transition pore to calcium load. Mol Cell Biochem 412:147–154

    CAS  PubMed  Google Scholar 

  110. Arieli Y, Gursahani H, Eaton MM, Hernandez LA, Schaefer S (2004) Gender modulation of Ca(2+) uptake in cardiac mitochondria. J Mol Cell Cardiol 37:507–513

    CAS  PubMed  Google Scholar 

  111. Yang SH, Liu R, Perez EJ, Wen Y, Stevens SM Jr, Valencia T, Brun-Zinkernagel AM, Prokai L, Will Y, Dykens J, Koulen P, Simpkins JW (2004) Mitochondrial localization of estrogen receptor beta. Proc Natl Acad Sci USA 101:4130–4135

    CAS  PubMed  Google Scholar 

  112. Zheng J, Ramirez VD (1999) Purification and identification of an estrogen binding protein from rat brain: oligomycin sensitivity-conferring protein (OSCP), a subunit of mitochondrial F0F1-ATP synthase/ATPase. J Steroid Biochem Mol Biol 68:65–75

    CAS  PubMed  Google Scholar 

  113. Moreno AJ, Moreira PI, Custodio JB, Santos MS (2013) Mechanism of inhibition of mitochondrial ATP synthase by 17beta-estradiol. J Bioenerg Biomembr 45:261–270

    CAS  PubMed  Google Scholar 

  114. Du H, Guo L, Fang F, Chen D, Sosunov AA, McKhann GM, Yan Y, Wang C, Zhang H, Molkentin JD, Gunn-Moore FJ, Vonsattel JP, Arancio O, Chen JX, Yan SD (2008) Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer’s disease. Nat Med 14:1097–1105

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Warne J, Pryce G, Hill JM, Shi X, Lenneras F, Puentes F, Kip M, Hilditch L, Walker P, Simone MI, Chan AW, Towers GJ, Coker AR, Duchen MR, Szabadkai G, Baker D, Selwood DL (2016) Selective inhibition of the mitochondrial permeability transition pore protects against neurodegeneration in experimental multiple sclerosis. J Biol Chem 291:4356–4373

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH/NINDS Grant No. 1R01NS095692.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Giovanni Manfredi.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Special Issue: In Honor of Professor Vera Adam-Vizi.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fels, J.A., Manfredi, G. Sex Differences in Ischemia/Reperfusion Injury: The Role of Mitochondrial Permeability Transition. Neurochem Res 44, 2336–2345 (2019). https://doi.org/10.1007/s11064-019-02769-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-019-02769-6

Keywords

Navigation