mtDNA T8993G mutation-induced F1F0-ATP synthase defect augments mitochondrial dysfunction associated with hypoxia/reoxygenation: the protective role of melatonin

PLoS One. 2013 Nov 29;8(11):e81546. doi: 10.1371/journal.pone.0081546. eCollection 2013.

Abstract

Background: F1F0-ATP synthase (F1F0-ATPase) plays important roles in regulating mitochondrial function during hypoxia, but the effect of F1F0-ATPase defect on hypoxia/reoxygenation (H/RO) is unknown. The aim of this study was to investigate how mtDNA T8993G mutation (NARP)-induced inhibition of F1F0-ATPase modulates the H/RO-induced mitochondrial dysfunction. In addition, the potential for melatonin, a potent antioxidant with multiple mitochondrial protective properties, to protect NARP cells exposed to H/RO was assessed.

Methods and findings: NARP cybrids harboring 98% of mtDNA T8993G genes were established as an in vitro model for cells with F1F0-ATPase defect; their parental osteosarcoma 143B cells were studied for comparison. Treating the cells with H/RO using a hypoxic chamber resembles ischemia/reperfusion in vivo. NARP significantly enhanced apoptotic death upon H/RO detected by MTT assay and the trypan blue exclusion test of cell viability. Based on fluorescence probe-coupled laser scanning imaging microscopy, NARP significantly enhanced mitochondrial reactive oxygen species (mROS) formation and mitochondrial Ca(2+) (mCa(2+)) accumulation in response to H/RO, which augmented the depletion of cardiolipin, resulting in the retardation of mitochondrial movement. With stronger H/RO stress (either with longer reoxygenation duration, longer hypoxia duration, or administrating secondary oxidative stress following H/RO), NARP augmented H/RO-induced mROS formation to significantly depolarize mitochondrial membrane potential (ΔΨm), and enhance mCa(2+) accumulation and nitric oxide formation. Also, NARP augmented H/RO-induced mROS oxidized and depleted cardiolipin, thereby promoting permanent mitochondrial permeability transition, retarded mitochondrial movement, and enhanced apoptosis. Melatonin markedly reduced NARP-augmented H/RO-induced mROS formation and therefore significantly reduced mROS-mediated depolarization of ΔΨm and accumulation of mCa(2+), stabilized cardiolipin, and then improved mitochondrial movement and cell survival.

Conclusion: NARP-induced inhibition of F1F0-ATPase enhances mROS formation upon H/RO, which augments the depletion of cardiolipin and retardation of mitochondrial movement. Melatonin may have the potential to rescue patients with ischemia/reperfusion insults, even those associated with NARP symptoms.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Apoptosis / drug effects
  • Apoptosis / genetics
  • Calcium / metabolism
  • Cardiolipins / metabolism
  • Cell Hypoxia / drug effects
  • Cell Line, Tumor
  • DNA, Mitochondrial / genetics*
  • Humans
  • Melatonin / pharmacology*
  • Mitochondria / drug effects*
  • Mitochondria / metabolism*
  • Mitochondrial Proton-Translocating ATPases / genetics*
  • Mutation*
  • Neuroprotective Agents / pharmacology
  • Oxygen / metabolism*
  • Reactive Oxygen Species / metabolism

Substances

  • Cardiolipins
  • DNA, Mitochondrial
  • Neuroprotective Agents
  • Reactive Oxygen Species
  • F1F0-ATP synthase
  • Mitochondrial Proton-Translocating ATPases
  • Melatonin
  • Oxygen
  • Calcium

Grants and funding

This work was supported by the grants CMRPG 270331-3 (to Huang), CMRPG 270341-2 (to Peng), and CMRPD 180491-3 and CMRPD 170411-3 (to Jou) from the Chang Gung Memorial Hospital and Chang Gung Medical Research Foundation, Taiwan. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.