Elsevier

Life Sciences

Volume 121, 15 January 2015, Pages 158-165
Life Sciences

Identification of mitochondrial deficits and melatonin targets in liver of septic mice by high-resolution respirometry

https://doi.org/10.1016/j.lfs.2014.11.031Get rights and content

Abstract

Aims

Previous data showed that melatonin maintains liver mitochondrial homeostasis during sepsis, but neither the mechanisms underlying mitochondrial dysfunction nor the target of melatonin are known.

Main methods

Here, we analyzed mitochondrial respiration in isolated mouse liver mitochondria with different substrate combinations (glutamate/malate, glutamate/malate/sucinate or succinate/rotenone) to identify mitochondrial defects and melatonin targets during sepsis. Other bioenergetic parameters including a + a3, b, and c + c1 content, mitochondrial mass, and mitochondrial supercomplexes formation were analyzed. Mitochondrial function was assessed during experimental sepsis induced by cecal ligation and puncture (CLP) in livers of 3 mo. C57BL/6 mice at early and late phases of sepsis, i.e., at 8 and 24 h after sepsis induction.

Key findings

Septic mice showed mitochondrial injury with a decrease in state 3, respiratory control rate, mitochondrial mass, and cytochrome b and c + c1 content, which was prevented by melatonin treatment. Mitochondrial dysfunction in sepsis was mainly linked to complex I damage, because complex II was far less impaired. These mitochondria preserved the respiratory supramolecular organization, maintaining their electron transport system capacity.

Significance

This work strengthens the use of substrate combinations to identify specific respiratory defects and selective melatonin actions in septic mitochondria. Targeting mitochondrial complex I should be a main therapeutical approach in the treatment of sepsis, whereas the use of melatonin should be considered in the therapy of clinical sepsis.

Introduction

Sepsis, a systemic inflammatory response of the organism against infection, represents the major cause of death in the intensive care units of developed countries [50], [51]. Liver dysfunction during sepsis is closely associated with high mortality in septic patients [54]. Moreover, a substantial body of evidence supports a relationship between mitochondrial dysfunction and sepsis [14], [15]. Thus, exhaustive study of the mitochondrial function may reveal new insights related to the pathophysiology of sepsis.

There are several pathways involved in mitochondrial dysfunction and ATP deficit during sepsis. At the early-onset of sepsis, the inflammatory response triggers mitochondrial morphological and structural alterations [8], [22], [66]. Then, poly (A) polymerase 1 (PARP-1) activation leads to a massive consumption of NAD+, decreasing its mitochondrial availability and reducing ATP production [38]. Later, proinflammatory cytokines generated during sepsis participate in the inhibition of pyruvate dehydrogenase (PDH), which reduces the substrates of OXPHOS, favoring an additional drop in ATP production [62], [73]. In parallel, the induction of inducible nitric oxide synthase (iNOS) during sepsis results in a significant elevation of nitric oxide (NO•) levels. In the mitochondria, NO• reversibly inhibits the complex IV of the ETC and, in a concentration-dependent manner, progressively inhibits other respiratory complexes [16], [19], [20], [72]. Moreover, NO• reacts with O2 generating the highly toxic peroxynitrite (ONOO) anions which irreversibly inhibit mitochondrial respiration [16]. Therefore, sepsis courses with an oxidative–nitrosative stress able to damage proteins, including respiratory complexes, DNA and membrane lipids, yielding severe mitochondrial dysfunction and bioenergetic failure [6], [23], [45]. Moreover, mitochondrial respiratory complexes can be assembled in supramolecular structures called supercomplexes (SC). This association provides greater efficiency facilitating the electron transfer through enzymatic respiratory complexes. SC formation is associated with an increase in electron flow efficiency and, therefore, in the ATP synthesis, minimizing the ROS generation [48]. The participation of each respiratory complex and/or SC in mitochondrial dysfunction during sepsis remains, however, unclear.

Melatonin (N-acetyl-5-methoxytryptamine) is a highly conserved and amphipathic indoleamine, present in all cellular compartments including mitochondria [4], [68]. There is consistent experimental background showing that mitochondria are the main intracellular target of melatonin [2], [3], [5]. Melatonin prevents septic shock and multiple organ failure reducing, iNOS expression and ROS generation, restoring ETC activity and ATP production [18], [24], [25], [44], [45], [47]. Therefore, an exhaustive study of the mitochondrial function with different substrate combinations and with the aid of high-resolution respirometry may identify new insights related to the pathophysiology of sepsis and the targets of melatonin.

Section snippets

Animals

Adult C57/BL6 male mice (3 months old) were provided by Harland (Barcelona, Spain). The animals were maintained in the University of Granada's facility under a 12 h light/dark cycle with lights on at 08:00 h, and a constant room temperature (22 °C ± 1 °C). Mice were housed in groups of five individuals per clear plastic cage with food and water ad libitum. All experiments were conducted in accordance with the University of Granada's Ethical Committee; the Spanish Protection Guide for Animal

Measurement of leak state

Leak state represents the oxygen consumption mainly used to compensate for the proton leak across the inner mitochondrial membrane in a non-phosphorylating respiration. To avoid an overestimation of leak state due to any contamination of ATPase activity (where ATP recycling can happen), we determined the leak state without adenylates (for CI and CI + II) or with oligomycin (for CII) [12]. Septic mice exhibited an upshift of leak during the early phase of sepsis (S8) with substrates for CI and for

Discussion

Mitochondrial impairment and bioenergetic failure have been related to elevated mortality in sepsis [13], [23], [73], although the mechanisms underlying mitochondrial dysfunction are unclear. The excess of NO• and ROS produced during the inflammatory response may inhibit the mitochondrial ETC complexes, reducing the electron flow through the ETC [6], [9], [23], [46], [62]. The subsequent electron leak promotes the formation of ROS and RNS, including the highly toxic ONOO, which irreversibly

Conclusion

Here, using HRR, the analysis of mitochondrial respiration allowed us to identify the different changes in CII and CI function with sepsis, which could not be identify when respiration is analyzed through CI + CII together. Thus, we confirmed that liver mitochondrial dysfunction in sepsis mainly depends on CI impairment, and identified the targets involved in the beneficial effects of melatonin treatment. The results support that mitochondrial CI should be a main therapeutical approach in the

Conflict of interest statement

The authors declare that there are no conflicts of interest.

Acknowledgments

This study was partially supported by grants from the Instituto de Salud Carlos III (RD12/0043/0005, PI08-1664), and from the Consejería de Innovación, Ciencia y Empresa, Junta de Andalucía (P07-CTS-03135 and CTS-101), Spain; CD and JAG are supported by the Instituto de Salud Carlos III, Spain; HV is a PhD student supported by a FPU fellowship (Ministerio de Educación, Spain); MED-C and ML-S are PhD students from the Consejería de Innovación, Ciencia y Empresa, Junta de Andalucía, Spain, and

References (75)

  • M. Frenzel et al.

    Ageing alters the supramolecular architecture of OxPhos complexes in rat brain cortex

    Exp. Gerontol.

    (2010)
  • J.J. García et al.

    N-acetylserotonin suppresses hepatic microsomal membrane rigidity associated with lipid peroxidation

    Eur. J. Pharmacol.

    (2001)
  • E. Gnaiger

    Capacity of oxidative phosphorylation in human skeletal muscle: new perspectives of mitochondrial physiology

    Int. J. Biochem. Cell Biol.

    (2009)
  • L.A. Gómez et al.

    Supercomplexes of the mitochondrial electron transport chain decline in the aging rat heart

    Arch. Biochem. Biophys.

    (2009)
  • M. Huttemann et al.

    The multiple functions of cytochrome c and their regulation in life and death decisions of the mammalian cell: From respiration to apoptosis

    Mitochondrion

    (2011)
  • S.P. Kantrow et al.

    Oxidative metabolism in rat hepatocytes and mitochondria during sepsis

    Arch. Biochem. Biophys.

    (1997)
  • S.S. Korshunov et al.

    The antioxidant functions of cytochrome c

    FEBS Lett.

    (1999)
  • L.C. López et al.

    Identification of an inducible nitric oxide synthase in diaphragm mitochondria from septic mice: its relation with mitochondrial dysfunction and prevention by melatonin

    Int. J. Biochem. Cell Biol.

    (2006)
  • D.A. Lowes et al.

    Antioxidants that protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial function, and reduce biochemical markers of organ dysfunction in a rat model of acute sepsis

    Br. J. Anaesth.

    (2013)
  • A. Mourier et al.

    Active proton leak in mitochondria: a new way to regulate substrate oxidation

    Biochim. Biophys. Acta

    (2010)
  • A. Navarro et al.

    Human brain cortex: mitochondrial oxidative damage and adaptive response in Parkinson disease and in dementia with Lewy bodies

    Free Radic. Biol. Med.

    (2009)
  • K. Pfeiffer et al.

    Cardiolipin stabilizes respiratory chain supercomplexes

    J. Biol. Chem.

    (2003)
  • R. Radi et al.

    Peroxynitrite reactions and formation in mitochondria

    Free Radic. Biol. Med.

    (2002)
  • T. Regueira et al.

    Mitochondrial dysfunction during sepsis, impact and possible regulating role of hypoxia-inducible factor-1alpha

    Med. Int.

    (2009)
  • M. Rosca et al.

    Cardiac mitochondria in heart failure: normal cardiolipin profile and increased threonine phosphorylation of complex IV

    Biochim. Biophys. Acta

    (2011)
  • R. Welter et al.

    The effects of nitric oxide on electron transport complexes

    Arch. Biochem. Biophys.

    (1996)
  • K.A. Wichterman et al.

    Sepsis and septic shock--a review of laboratory models and a proposal

    J. Surg. Res.

    (1980)
  • M. Zhang et al.

    Gluing the respiratory chain together. Cardiolipin is required for supercomplex formation in the inner mitochondrial membrane

    J. Biol. Chem.

    (2002)
  • D. Acuña-Castroviejo et al.

    Melatonin-mitochondria interplay in health and disease

    Curr. Top. Med. Chem.

    (2011)
  • D. Acuña-Castroviejo et al.

    Melatonin role in the mitochondrial function

    Front. Biosci.

    (2007)
  • D. Acuña-Castroviejo et al.

    Extrapineal melatonin: sources, regulation, and potential functions

    Cell. Mol. Life Sci.

    (2014)
  • D. Acuña-Castroviejo et al.

    Melatonin, mitochondria, and cellular bioenergetics

    J. Pineal Res.

    (2001)
  • R.M. Berg et al.

    Neuro-oxidative-nitrosative stress in sepsis

    J. Cereb. Blood Flow Metab.

    (2011)
  • M.D. Brand et al.

    Assessing mitochondrial dysfunction in cells

    Biochem. J.

    (2011)
  • D. Brealey et al.

    Mitochondrial dysfunction in a long-term rodent model of sepsis and organ failure

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2004)
  • D. Brealey et al.

    Mitochondrial dysfunction in sepsis

    Curr. Infect. Dis. Rep.

    (2003)
  • L.A. Callahan et al.

    Sepsis induces diaphragm electron transport chain dysfunction and protein depletion

    Am. J. Respir. Crit. Care Med.

    (2005)
  • Cited by (22)

    • Perturbed Mitochondrial Dynamics Is a Novel Feature of Colitis That Can Be Targeted to Lessen Disease

      2020, Cellular and Molecular Gastroenterology and Hepatology
      Citation Excerpt :

      The mitochondrial network was quantified for 20 epithelial cells and 50 macrophages/preparation as fused, intermediate, or fragmented morphology expressed as a percent of mitochondrial morphology.58 Real-time, continuous assessment of oxidative phosphorylation (OXPHOS) in permeabilized IEC 4.1 cells was performed by using the Oxygraph-2k C-series (Oroboros Instruments, Innsbruck, Austria) according to the substrate inhibitor uncoupler titration protocol-002 with modifications to digitonin and carbonyl cyanide p-(trifluoromethoxy) phenylhydrazone titration protocol (http://wiki.oroboros.at/index.php/SUIT-002).59 Cells (1.5 × 106) were seeded in 100 × 20 mm culture dishes for 24 hours, followed by 24-hour exposure to 2% DSS ± P110 (1.5 μmol/L).

    • Myalgic encephalomyelitis/chronic fatigue syndrome: From pathophysiological insights to novel therapeutic opportunities

      2019, Pharmacological Research
      Citation Excerpt :

      Melatonin supplementation also has profound anti-apoptotic consequences in other areas, reducing the release of cytochrome c into the cytosol and inhibiting caspase-3 activity [332]. Importantly, there is research to suggest that the protective effects of melatonin supplementation are not limited to in vitro experiments as these effects have also been observed in animal models of sepsis [333,334], ischaemia/reperfusion [335,336], Parkinson’s disease (MPP+: 1-methyl-4-phenylpyridinium) [322], and Alzheimer’s disease β-amyloid peptide [Aβ 25–35: amyloid beta (25–35] [337,338], HNE [339] and LPS [328]. Melatonin exerts a number of different effects on the immune system that are broadly pro-inflammatory when the immune system is operating physiologically and anti-inflammatory when the immune system is in an activated state [340].

    • Targeting of reactive isolevuglandins in mitochondrial dysfunction and inflammation

      2019, Redox Biology
      Citation Excerpt :

      Impaired oxidative phosphorylation significantly contribute to organ damage in sepsis [37]. Mitochondrial dysfunction in sepsis was recently linked to complex I damage, and targeted protection of complex I was proposed as a treatment for sepsis [38]. Sepsis was previously shown to increase IsoLG production [11] and IsoLG can induce the mitochondrial permeability transition [9].

    • Permeabilized myocardial fibers as model to detect mitochondrial dysfunction during sepsis and melatonin effects without disruption of mitochondrial network

      2016, Mitochondrion
      Citation Excerpt :

      Melatonin (30 mg/ml dissolved in ethanol: saline, 0.25% v/v) was administered to mice at a dose of 30 mg/kg body weight: one dose 30 min before surgery (intraperitoneal); a second dose just after the surgery (subcutaneous), and the third and fourth doses 4 and 8 h later, respectively (subcutaneous). S8M group received only the first three doses of melatonin (Doerrier et al., 2015). The dose of melatonin employed in the present work was obtained from preliminary dose-dependent studies aimed at inhibiting the expression and activity of iNOS in CLP septic mice.

    View all citing articles on Scopus
    View full text