Elsevier

NeuroToxicology

Volume 64, January 2018, Pages 204-218
NeuroToxicology

Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes

https://doi.org/10.1016/j.neuro.2017.05.009Get rights and content

Highlights

  • Mn induces mitochondrial dysfunction in astrocytes.

  • Mn exposure induces inflammatory response in astrocytes and exacerbates αSynagg-induced inflammatory response in astrocytes.

  • Mito-apocynin attenuates Mn-induced inflammatory response in astrocytes by reducing mitochondrial damage.

  • Intranasal Mn exposure induces neuroinflammation and behavioral deficits in mouse model.

  • Thus, Mn-induced mitochondrial defects contribute to astroglial neuroinflammation.

Abstract

Chronic manganese (Mn) exposure induces neurotoxicity, which is characterized by Parkinsonian symptoms resulting from impairment in the extrapyramidal motor system of the basal ganglia. Mitochondrial dysfunction and oxidative stress are considered key pathophysiological features of Mn neurotoxicity. Recent evidence suggests astrocytes as a major target of Mn neurotoxicity since Mn accumulates predominantly in astrocytes. However, the primary mechanisms underlying Mn-induced astroglial dysfunction and its role in metal neurotoxicity are not completely understood. In this study, we examined the interrelationship between mitochondrial dysfunction and astrocytic inflammation in Mn neurotoxicity. We first evaluated whether Mn exposure alters mitochondrial bioenergetics in cultured astrocytes. Metabolic activity assessed by MTS assay revealed an IC50 of 92.68 μM Mn at 24 h in primary mouse astrocytes (PMAs) and 50.46 μM in the human astrocytic U373 cell line. Mn treatment reduced mitochondrial mass, indicative of impaired mitochondrial function and biogenesis, which was substantiated by the significant reduction in mRNA of mitofusin-2, a protein that serves as a ubiquitination target for mitophagy. Furthermore, Mn increased mitochondrial circularity indicating augmented mitochondrial fission. Seahorse analysis of bioenergetics status in Mn-treated astrocytes revealed that Mn significantly impaired the basal mitochondrial oxygen consumption rate as well as the ATP-linked respiration rate. The effect of Mn on mitochondrial energy deficits was further supported by a reduction in ATP production. Mn-exposed primary astrocytes also exhibited a severely quiescent energy phenotype, which was substantiated by the inability of oligomycin to increase the extracellular acidification rate. Since astrocytes regulate immune functions in the CNS, we also evaluated whether Mn modulates astrocytic inflammation. Mn exposure in astrocytes not only stimulated the release of proinflammatory cytokines, but also exacerbated the inflammatory response induced by aggregated α-synuclein. The novel mitochondria-targeted antioxidant, mito-apocynin, significantly attenuated Mn-induced inflammatory gene expression, further supporting the role of mitochondria dysfunction and oxidative stress in mediating astrogliosis. Lastly, intranasal delivery of Mn in vivo elevated GFAP and depressed TH levels in the olfactory bulbs, clearly supporting the involvement of astrocytes in Mn-induced dopaminergic neurotoxicity. Collectively, our study demonstrates that Mn drives proinflammatory events in astrocytes by impairing mitochondrial bioenergetics.

Introduction

Central nervous system (CNS) aberrations resulting from prolonged exposure to transition metals, such as iron (Febbraro et al., 2012, Galvin et al., 2000, Gregory et al., 2009), lead (Bihaqi and Zawia, 2013), and manganese (Gorojod et al., 2015, Hesketh et al., 2008), lend support to the potential involvement of environmental metal exposure in the etiology of chronic neurodegeneration (Rokad et al., 2016). Manganese (Mn) toxicity resulting from overexposure to airborne sources was first noted in miners (Peres et al., 2016, Rodier, 1955), and subsequently in industrial workers like welders (Horning et al., 2015, Kwakye et al., 2015). Although Mn has a multitude of physiological functions (e.g., enzyme cofactor for superoxide dismutase and arginase, immunity, bone development, and reproduction) (Karki et al., 2015), the accruing toxic levels has been shown to predominately localize to the globus pallidus, as well as other brain areas of the extrapyramidal motor system, thereby leading to a condition termed manganism, which is phenotypically similar to PD (Kwakye et al., 2015, Milatovic et al., 2007). In addition, diets including grains, legumes, baby formulations and nuts represent the main source of Mn, of which <5% is intestinally absorbed (Karki et al., 2013, Kwakye et al., 2015). Approximately, 30–40% of inhaled Mn is absorbed into the bloodstream, where it is transported primarily bound to albumin and β-globulin in oxidation state 2 (Mn2+) (Karki et al., 2013); however, the absorbed proportion could be smaller (∼10.6%) because of oxidative state-dependent differences in solubility (Ellingsen et al., 2003). Availability of the Mn2+ oxidation state in the central nervous system (CNS) is facilitated by transporters such as divalent metal ion transporter 1 (DMT1) and SLC13A10, while Mn3+ is facilitated by transferrin (Chen et al., 2015, Karki et al., 2015, Leyva-Illades et al., 2014, Sidoryk-Wegrzynowicz and Aschner, 2013).

As a ubiquitous constituent and physiological glue of the CNS, astrocytes are indispensable for cellular homeostatic maintenance (Volterra and Meldolesi, 2005). Astrocytes regulate extracellular glutamate levels among neurons in tripartite synapses (Karki et al., 2013, Popoli et al., 2011). Excessive Mn can disrupt this regulation by inducing tumor necrosis factor-α (TNFα), a secreted proinflammatory cytokine. Previous studies demonstrated that Mn impairs calcium (Ca2+) homeostasis in astrocytes and activates key proinflammatory events such as NFκB, NOS2 and cytokine production (Karki et al., 2014, Karki et al., 2015). This inflammatory response increases expression of yin yang-1, a transcription repressor that inhibits production of excitatory amino acid transporter-2, which is necessary for glutamate reuptake (Karki et al., 2014, Karki et al., 2015). Recent studies also demonstrate that Mn interferes with neurodegenerative disease-specific proteins including prions, alpha-synuclein, and huntingtin (Bichell et al., 2017, Choi et al., 2007, Choi et al., 2010, Harischandra et al., 2015), as well as protein misfolding, which may further contribute to neuroinflammation. Thus, Mn exposure can activate the inflammatory response in astrocytes, which can further contribute to Mn neurotoxicity.

Along with pericytes, the end-feet of astrocytes envelope blood vessels to regulate blood–brain barrier trafficking (Pekny and Pekna, 2014). Interestingly, astrocytes have been demonstrated to have an increased affinity for Mn, with concentrations up to 50-fold greater than in neurons (Aschner et al., 1992, Gonzalez et al., 2008). Once internalized, Mn is sequestered by mitochondria via the Ca2+ uniporter (Gunter et al., 2006). Kinetical analyses revealed the influx of Mn through this mitochondrial uniporter is slower than Ca2+ per se, but Mn efflux is far slower (Gavin et al., 1990, Martinez-Finley et al., 2013). Even though the atomic charge properties and size of Mn are quite analogous to Ca2+, the two Ca2+ efflux mechanisms are poor exporters of Mn, thus explaining the mitochondrial sequestration of Mn (Gunter et al., 1975, Gunter and Sheu, 2009, Martinez-Finley et al., 2013). On the other hand, although sodium (Na+)-independent Ca2+ efflux does transport Mn, Na+-dependent Ca2+ efflux, the predominant mechanism in CNS mitochondria, has not been reported to export Mn (Gavin et al., 1990, Gunter and Sheu, 2009, Martinez-Finley et al., 2013). This extremely sluggish efflux of Mn potentially explains the prolonged CNS clearance and the buildup of toxicity (Martinez-Finley et al., 2013). Mn can also competitively inhibit both efflux pathways, thereby raising the mitochondrial Ca2+ concentration, which can impair aerobic respiration and elicit reactive oxygen species (ROS) generation (Gavin et al., 1990, Martinez-Finley et al., 2013, Streifel et al., 2013, Tjalkens et al., 2006).

Despite the known effects of Mn on astrocytes, the extent of Mn-induced structural and functional impairment of mitochondrial dynamics and its consequent neuroinflammatory processes as it relates to Mn neurotoxicity have not been systematically examined. In the present study, we characterized the impact of Mn on mitochondrial bioenergetic flux in astrocytes (using the Seahorse bioenergetic analyzer) and its functional consequence on neuroinflammation. Our results demonstrate that overexposure to Mn (i) lowers basal respiration in astrocytes, (ii) induces astrocytic inflammation that is not exclusive to TNFα, and (iii) potentiates aggregated α-synuclein (αSynagg)-induced inflammation in astrocytes. Thus, our study establishes a functional interaction between mitochondrial dysfunction and astrocytic inflammation in Mn-induced neurotoxicity. In addition, we report that the mitochondria-targeted anti-oxidant mito-apocynin attenuates Mn-induced astrocyte inflammation.

Section snippets

Chemicals and reagents

Dulbecco's modified eagle medium (DMEM), fetal bovine serum (FBS), l-glutamine (Q), and penicillin/streptomycin (P/S) were obtained from Invitrogen (Carlsbad, CA). MitoTracker Green FM and MitoTracker Red CMXRos, were purchased from Molecular Probes (Eugene, OR). Manganese chloride (MnCl2) was obtained from Sigma–Aldrich (St. Louis, MO). The CellTiter® 961 AQueous Non-Radioactive Cell Proliferation Assay kit and CellTiter® Glo Luminescent Cell Viability Assay kit were obtained from Promega

Mn induced a dose-dependent decrease in mitochondrial metabolic activity in astrocytes

Mn has been shown to accumulate in mitochondria, block the electron transport chain (ETC), and alter mitochondrial permeability in astrocytes (Hazell, 2002, Rao and Norenberg, 2004). In this experiment, we examined the effect of Mn on metabolic activity in PMA and U373 cells. Both sets of astrocytes were exposed to increasing concentrations of Mn (0.001–1 mM) for 24 h. We observed a dose-dependent decrease in metabolic activity as determined by MTS assay. The IC50 for inhibiting metabolic

Discussion

Neuroinflammation plays a key role in neurodegenerative disorders, including Parkinsonian syndrome (Glass et al., 2010, Golde, 2002, Herrero et al., 2015, Tansey and Goldberg, 2010, Whitton, 2007). Even though the etiology of these diseases is not well established, the consensus is that environment plays an important role in these neurodegenerative disorders. Astrocytes, the most abundant glial cells of the brain, are a recognized source of sustained inflammation upon activation (Phillips et

Conflict of interest

A.G.K. and V.A. are shareholders of PK Biosciences Corporation (Ames, IA), which is interested in identifying novel biomarkers and potential therapeutic targets for PD. A.G.K and V.A. do not have any direct financial interest in the work presented in present work.

Transparency document

.

Acknowledgements

This work was supported by National Institutes of Health (NIH) Grants: ES026892, ES10586, and NS088206. The authors would like to thank Dr. Balaraman Kalyanaraman for providing access to mito-apocynin. Lloyds chair to A.G.K. and Salsbury Chair to A.K. are also acknowledged.

References (90)

  • C.K. Glass et al.

    Mechanisms underlying inflammation in neurodegeneration

    Cell

    (2010)
  • L.E. Gonzalez et al.

    Manganese activates the mitochondrial apoptotic pathway in rat astrocytes by modulating the expression of proteins of the Bcl-2 family

    Neurochem. Int.

    (2008)
  • R. Gordon et al.

    A simple magnetic separation method for high-yield isolation of pure primary microglia

    J. Neurosci. Methods

    (2011)
  • R. Gordon et al.

    Protein kinase Cδ upregulation in microglia drives neuroinflammatory responses and dopaminergic neurodegeneration in experimental models of Parkinson's disease

    Neurobiol. Dis.

    (2016)
  • R.M. Gorojod et al.

    The autophagic-lysosomal pathway determines the fate of glial cells under manganese-induced oxidative stress conditions

    Free Radic. Biol. Med.

    (2015)
  • C.L. Graff et al.

    Nasal drug administration: potential for targeted central nervous system delivery

    J. Pharm. Sci.

    (2005)
  • R.E. Gunter et al.

    Quantitative magnetic resonance studies of manganese uptake by mitochondria

    Biophys. J.

    (1975)
  • T.E. Gunter et al.

    Speciation of manganese in cells and mitochondria: a search for the proximal cause of manganese neurotoxicity

    Neurotoxicology

    (2006)
  • T.E. Gunter et al.

    Characteristics and possible functions of mitochondrial Ca(2+) transport mechanisms

    Biochim. Biophys. Acta

    (2009)
  • D.S. Harischandra et al.

    Environmental neurotoxicant manganese regulates exosome-mediated extracellular miRNAs in cell culture model of Parkinson's disease: relevance to α-synuclein misfolding in metal neurotoxicity

    Neurotoxicology

    (2018)
  • A.S. Hazell

    Astrocytes and manganese neurotoxicity

    Neurochem. Int.

    (2002)
  • S. Hesketh et al.

    Elevated manganese levels in blood and CNS in human prion disease

    Mol. Cell. Neurosci.

    (2008)
  • A.G. Kanthasamy et al.

    Effect of divalent metals on the neuronal proteasomal system, prion protein ubiquitination and aggregation

    Toxicol. Lett.

    (2012)
  • P. Karki et al.

    Role of transcription factor yin yang 1 in manganese-induced reduction of astrocytic glutamate transporters: putative mechanism for manganese-induced neurotoxicity

    Neurochem. Int.

    (2015)
  • L. Liu et al.

    IPAF inflammasome is involved in interleukin-1beta production from astrocytes, induced by palmitate; implications for Alzheimer's disease

    Neurobiol. Aging

    (2014)
  • T. Liu et al.

    Overexpression of mitofusin 2 inhibits reactive astrogliosis proliferation in vitro

    Neurosci. Lett.

    (2014)
  • E.J. Martinez-Finley et al.

    Manganese neurotoxicity and the role of reactive oxygen species

    Free Radic. Biol. Med.

    (2013)
  • A.H. Moberly et al.

    Intranasal exposure to manganese disrupts neurotransmitter release from glutamatergic synapses in the central nervous system in vivo

    Neurotoxicology

    (2012)
  • L. Narhi et al.

    Both familial Parkinson's disease mutations accelerate alpha-synuclein aggregation

    J. Biol. Chem.

    (1999)
  • H.A. Ngwa et al.

    Vanadium exposure induces olfactory dysfunction in an animal model of metal neurotoxicity

    Neurotoxicology

    (2014)
  • E.J. Park et al.

    Induction of oxidative stress and inflammatory cytokines by manganese chloride in cultured T98G cells, human brain glioblastoma cell line

    Toxicol. In Vitro

    (2010)
  • K.V. Rao et al.

    Manganese induces the mitochondrial permeability transition in cultured astrocytes

    J. Biol. Chem.

    (2004)
  • K.M. Streifel et al.

    Manganese inhibits ATP-induced calcium entry through the transient receptor potential channel TRPC3 in astrocytes

    Neurotoxicology

    (2013)
  • X. Su et al.

    Synuclein activates microglia in a model of Parkinson's disease

    Neurobiol. Aging

    (2008)
  • F.L. Tang et al.

    VPS35 deficiency or mutation causes dopaminergic neuronal loss by impairing mitochondrial fusion and function

    Cell Rep.

    (2015)
  • M.G. Tansey et al.

    Neuroinflammation in Parkinson's disease: its role in neuronal death and implications for therapeutic intervention

    Neurobiol. Dis.

    (2010)
  • R.B. Tjalkens et al.

    Manganese suppresses ATP-dependent intercellular calcium waves in astrocyte networks through alteration of mitochondrial and endoplasmic reticulum calcium dynamics

    Brain Res.

    (2006)
  • Z. Yin et al.

    Mitochondrial-dependent manganese neurotoxicity in rat primary astrocyte cultures

    Brain Res.

    (2008)
  • S. Alfonso-Loeches et al.

    Role of mitochondria ROS generation in ethanol-induced NLRP3 inflammasome activation and cell death in astroglial cells

    Front. Cell. Neurosci.

    (2014)
  • S. Amor et al.

    Inflammation in neurodegenerative diseases

    Immunology

    (2010)
  • M. Aschner et al.

    Manganese uptake and efflux in cultured rat astrocytes

    J. Neurochem.

    (1992)
  • T.J. Bichell et al.

    Reduced bioavailable manganese causes striatal urea cycle pathology in Huntington's disease mouse model

    Biochim. Biophys. Acta

    (2017)
  • T.M. Brenza et al.

    Neuronal protection against oxidative insult by polyanhydride nanoparticle-based mitochondria-targeted antioxidant therapy

    Nanomedicine

    (2016)
  • P. Chen et al.

    SLC30A10: a novel manganese transporter

    Worm

    (2015)
  • C.J. Choi et al.

    Manganese upregulates cellular prion protein and contributes to altered stabilization and proteolysis: relevance to role of metals in pathogenesis of prion disease

    Toxicol. Sci.

    (2010)
  • Cited by (95)

    • Mitochondrial dysfunction: A fatal blow in depression

      2023, Biomedicine and Pharmacotherapy
    View all citing articles on Scopus
    1

    These authors made equal contributions.

    View full text