Elsevier

Brain, Behavior, and Immunity

Volume 92, February 2021, Pages 90-101
Brain, Behavior, and Immunity

Mitochondrial pyruvate carrier as a key regulator of fever and neuroinflammation

https://doi.org/10.1016/j.bbi.2020.11.031Get rights and content

Highlights

  • MPC inhibition attenuates fever and neuroinflammation.

  • LPS induces an increase in oxygen consumption in the hypothalamus.

  • MPC inhibition prevents the LPS-induced increase in oxygen consumption.

  • Changes in neurometabolism regulate the levels of inflammatory markers.

Abstract

The mitochondrial pyruvate carrier (MPC) is an inner-membrane transporter that facilitates pyruvate uptake from the cytoplasm into mitochondria. We previously reported that MPC1 protein levels increase in the hypothalamus of animals during fever induced by lipopolysaccharide (LPS), but how this increase contributes to the LPS responses remains to be studied. Therefore, we investigated the effect of UK 5099, a classical MPC inhibitor, in a rat model of fever, on hypothalamic mitochondrial function and neuroinflammation in LPS-stimulated preoptic area (POA) primary microcultures. Intracerebroventricular administration of UK 5099 reduced the LPS-induced fever. High-resolution respirometry revealed an increase in oxygen consumption and oxygen flux related to ATP synthesis in the hypothalamic homogenate from LPS-treated animals linked to mitochondrial complex I plus II. Preincubation with UK 5099 prevented the LPS-induced increase in oxygen consumption, ATP synthesis and spare capacity only in complex I-linked respiration and reduced mitochondrial H2O2 production. In addition, treatment of rat POA microcultures with UK 5099 reduced the secretion of the proinflammatory and pyrogenic cytokines TNFα and IL-6 as well as the immunoreactivity of inflammatory transcription factors NF-κB and NF-IL6 four hours after LPS stimulation. These results suggest that the regulation of mitochondrial pyruvate metabolism through MPC inhibition may be effective in reducing neuroinflammation and fever.

Introduction

Over the last years, neuroinflammation has been linked to changes in cellular metabolism (Ghosh et al., 2016, Mills et al., 2017, Nair et al., 2019, Peruzzotti-Jametti and Pluchino, 2018). Under inflammatory conditions, neuronal and non-neuronal cell types have an increased need in energy to fulfill their protective functions. Thus, changes in the bioenergetic pathways are required to supply the increased demand for energy (Lynch, 2020).

Fever is a brain-mediated increase in body core temperature, a nonspecific host defense response to infectious or inflammatory insults. The increase and maintenance of body temperature associated with fever is due to behavioral changes and autonomic responses aimed at increasing heat production and decreasing heat loss (Blomqvist and Engblom, 2018, Roth and Blatteis, 2014). The body metabolic rate is estimated to increase by about 10% to 12.5% for every 1 °C rise in body temperature (Evans et al., 2015, Kluger, 1986). Nevertheless, little is known about the metabolic changes that occur in the hypothalamus and the preoptic hypothalamic area (POA), the major thermoregulatory region in the central nervous system (Roth and Blatteis, 2014). Indeed, metabolomic profiling of the hypothalamus in yeast-induced fever in rats revealed an increase in the metabolism of purines, lipids, amino acids, and energy metabolism in the febrile group (Liu et al., 2015). Additionally, we recently performed label-free quantitative proteomics of the hypothalamus in a rat model of fever induced by lipopolysaccharide (LPS). Pathway analysis showed changes in several proteins related to cellular metabolic processes, including glycolysis, citric acid cycle and oxidative phosphorylation (Firmino et al., 2018).

Pyruvate is a key metabolite linking cytoplasmic and mitochondrial metabolism. It is generated in the cytoplasm mainly through glycolysis and is subsequently transported into the mitochondria by the mitochondrial pyruvate carrier (MPC), a protein complex in the inner mitochondrial membrane that is composed of two essential components, MPC1 and MPC2 (Olson et al., 2016). The activity of this transporter, its kinetics and specificity for substrates and inhibitors was described decades ago (Halestrap, 1975), but its molecular identity was only recently determined (Bricker et al., 2012, Herzig et al., 2012). Constitutive knockout of MPC1 (Bowman et al., 2016, Vanderperre et al.,2016) or MPC2 (Vigueira et al., 2014) in mice leads to embryonic lethality, whereas partial deletion of MPC1 results in low body weight, decreased movement, low body shell temperature, and less adipose tissue accumulation in mice (Zou et al., 2018).

Interestingly, in the previously mentioned proteomic analysis study, we also found that MPC1 is upregulated in the hypothalamus of febrile rats (Firmino et al., 2018). The functional cause of MPC1 upregulation remains to be investigated, but given the central metabolic position of the MPC complex, changes in its activity, either through post-translational modifications or protein abundance, may regulate overall cellular metabolism (Gray et al., 2016). Since changes in cellular metabolism also influence neuroinflammation and redox signaling (Aguilera et al., 2018), the reduction in the inflammatory response and the promotion of neuroprotection as a consequence of blocking pyruvate transport by MPC inhibition have been documented in experimental models of diabetes, aging, and neurodegenerative diseases (Divakaruni et al., 2017, Quansah et al., 2018, Ghosh et al., 2016).

In this study, we evaluated the functional capacity of hypothalamic mitochondria in a model of fever induced by LPS. We found a critical modulation of hypothalamic mitochondrial function during the peak of fever that was related to increased MPC activity. We also investigated the link between MPC inhibition and the reduction of fever, mitochondrial H2O2 production and neuroinflammation.

Section snippets

Animals

Fever experiments were performed in female and male Wistar rats (180–200 g body weight). The animals were obtained from the Animal House of the University of Brasília and housed at 24 ± 1 °C, under a 12:12-hour light–dark cycle (lights on at 7 a.m.), with access to food and water ad libitum. Animal handling and experiments were approved by the Animal Research Ethics Committee of the University of Brasília (Protocol no. 99/2017). This study was carried out according to the Guide for the Care and

Inhibition of mitochondrial pyruvate carrier attenuates fever induced by LPS

Administration of LPS (5 µg/kg, intravenously) induced an increase in body temperature, which started approximately one hour after injection and produced a biphasic fever with the first peak occurring around 2.5 and the second peak around 5 h [FLPS (1, 25) = 32.01, p < 0.0001; Ftime (26, 650) = 16.91, p < 0.0001; Ftime x LPS (26, 650) = 7.807, p < 0.0001, Fig. 1A]. Pretreatment of the animals with a selective MPC inhibitor, UK 5099 (1 µg, i.c.v.), decreased the febrile response mainly during

Discussion

This study revealed an important association between hypothalamic mitochondrial metabolism and the modulation of fever and neuroinflammation. We recently demonstrated the upregulation of MPC1 in the hypothalamus of febrile rats (Firmino et al., 2018), which was associated with a higher metabolic demand. Since the pharmacological inhibition of hypothalamic MPC attenuated fever and neuroinflammation induced by LPS, we postulated that pyruvate metabolism has a key role under these conditions.

Funding

This work was supported by the Fundação de Apoio à Pesquisa do Distrito Federal (FAPDF grants 193.001.730/2017, 00193-00000106/2019-75, 00193–00001324/2019-27); Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq grant 424809-2018-4); Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) – Finance Code 001; and Instituto Nacional de Ciência e Tecnologia e Neuro-ImunoModulação (INCT-NIM grant 485489/2014-1). N.C.G is a recipient of a scholarship from CNPq

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

We gratefully acknowledge Prof. Dr. Connie McManus (Institute of Biological Sciences, University of Brasília, Brazil) for the valid discussions about the statistical analysis, which contributed to the quality of our article and we thank Priscila Batista da Rosa for excellent graphical design.

References (75)

  • S. Leisengang et al.

    Primary Cultures from Rat Dorsal Root Ganglia: Responses of Neurons and Glial Cells to Somatosensory or Inflammatory Stimulation

    Neuroscience

    (2018)
  • M.A. Lynch

    Can the emerging field of immunometabolism provide insights into neuroinflammation?

    Prog. Neurobiol.

    (2020)
  • A.P. Muller et al.

    Insulin prevents mitochondrial generation of H2O2 in rat brain

    Exp. Neurol.

    (2013)
  • K.A. Olson et al.

    Pyruvate and Metabolic Flexibility: Illuminating a Path Toward Selective Cancer Therapies

    Trends Biochem. Sci.

    (2016)
  • L. Peruzzotti-Jametti et al.

    Targeting Mitochondrial Metabolism in Neuroinflammation: Towards a Therapy for Progressive Multiple Sclerosis

    Trends Mol. Med.

    (2018)
  • K. Riester et al.

    In vivo characterization of functional states of cortical microglia during peripheral inflammation

    Brain Behav. Immun.

    (2020)
  • C. Rummel

    Inflammatory transcription factors as activation markers and functional readouts in immune-to-brain communication

    Brain Behav. Immun.

    (2016)
  • E.A. Saramago et al.

    Molecular hydrogen potentiates hypothermia and prevents hypotension and fever in LPS-induced systemic inflammation

    Brain Behav. Immun.

    (2019)
  • G.A. Shaw et al.

    Traumatic stress history interacts with sex and chronic peripheral inflammation to alter mitochondrial function of synaptosomes

    Brain Behav. Immun.

    (2020)
  • D.d.M. Soares et al.

    The relevance of kalikrein-kinin system via activation of B2 receptor in LPS-induced fever in rats

    Neuropharmacology

    (2017)
  • D.M. Soares et al.

    Chemokine ligand (CCL)-3 promotes an integrated febrile response when injected within pre-optic area (POA) of rats and induces calcium signaling in cells of POA microcultures but not TNF-α or IL-6 synthesis

    Brain Behav. Immun.

    (2013)
  • N. Vacanti et al.

    Regulation of Substrate Utilization by the Mitochondrial Pyruvate Carrier

    Mol. Cell

    (2014)
  • P. Vigueira et al.

    Mitochondrial Pyruvate Carrier 2 Hypomorphism in Mice Leads to Defects in Glucose-Stimulated Insulin Secretion

    Cell Reports

    (2014)
  • S. Zou et al.

    Fatty acid oxidation alleviates the energy deficiency caused by the loss of MPC1 in MPC1+/− mice

    Biochem. Biophys. Res. Commun.

    (2018)
  • L.A. Aarden et al.

    Production of hybridoma growth factor by human monocytes

    Eur. J. Immunol.

    (1987)
  • G. Aguilera et al.

    Redox Signaling, Neuroinflammation, and Neurodegeneration

    Antioxid. Redox Signal.

    (2018)
  • D.A. Bader et al.

    Mitochondrial pyruvate import is a metabolic vulnerability in androgen receptor-driven prostate cancer

    Nat Metab

    (2019)
  • A.L. Bastos-Pereira et al.

    Involvement of brain cytokines in zymosan-induced febrile response

    J. Appl. Physiol.

    (2014)
  • A. Blomqvist et al.

    Neural Mechanisms of Inflammation-Induced Fever

    Neuroscientist

    (2018)
  • C.E. Bowman et al.

    Requirement for the Mitochondrial Pyruvate Carrier in Mammalian Development Revealed by a Hypomorphic Allelic Series

    Mol. Cell. Biol.

    (2016)
  • D.K. Bricker et al.

    A Mitochondrial Pyruvate Carrier Required for Pyruvate Uptake in Yeast, Drosophila , and Humans

    Science

    (2012)
  • Cartmell, T., Luheshi, G.N., Rothwell, N.J., 1999. Brain sites of action of endogenous interleukin-1 in the febrile...
  • E. de‐Souza‐Ferreira et al.

    Mitochondria‐coupled glucose phosphorylation develops after birth to modulate H2O2 release and calcium handling in rat brain

    J. Neurochem.

    (2019)
  • C.A. Dinarello

    Infection, fever, and exogenous and endogenous pyrogens: some concepts have changed

    J. Endotoxin Res.

    (2004)
  • A.S. Divakaruni et al.

    Inhibition of the mitochondrial pyruvate carrier protects from excitotoxic neuronal death

    J. Cell Biol.

    (2017)
  • O. Errea et al.

    The disruption of mitochondrial axonal transport is an early event in neuroinflammation

    J. Neuroinflammation

    (2015)
  • A. Eskilsson et al.

    Immune-Induced Fever Is Dependent on Local But Not Generalized Prostaglandin E2 Synthesis in the Brain

    J. Neurosci.

    (2017)
  • Cited by (0)

    View full text