Elsevier

NeuroToxicology

Volume 28, Issue 3, May 2007, Pages 478-489
NeuroToxicology

Gene expression profiling of human primary astrocytes exposed to manganese chloride indicates selective effects on several functions of the cells

https://doi.org/10.1016/j.neuro.2006.10.005Get rights and content

Abstract

Exposure of adult humans to manganese (Mn) has long been known to cause neurotoxicity. Recent evidence also suggests that exposure of children to Mn is associated with developmental neurotoxicity. Astrocytes are critical for the proper functioning of the nervous system, and they play active roles in neurogenesis, synaptogenesis and synaptic neurotransmission. In this report, to help elucidate the molecular events underlying Mn neurotoxicity, we systematically identified the molecular targets of Mn in primary human astrocytes at a genome-wide level, by using microarray gene expression profiling and computational data analysis algorithms. We found that Mn altered the expression of diverse genes ranging from those encoding cytokines and transporters to signal transducers and transcriptional regulators. Particularly, 28 genes encoding proinflammatory chemokines, cytokines and related functions were up-regulated, whereas 15 genes encoding functions involved in DNA replication and repair and cell cycle checkpoint control were down-regulated. Consistent with the increased expression of proinflammatory factors, analysis of common regulators revealed that 16 targets known to be positively affected by the interferon-γ signaling pathway were up-regulated by Mn2+. In addition, 68 genes were found to be similarly up- or down-regulated by both Mn2+ and hypoxia. These results from genomic analysis are further supported by data from real-time RT-PCR, Western blotting, flow cytometric and toxicological analyses. Together, these analyses show that Mn2+ selectively affects cell cycle progression, the expression of hypoxia-responsive genes, and the expression of proinflammatory factors in primary human astrocytes. These results provide important insights into the molecular mechanisms underlying Mn neurotoxicity.

Introduction

Manganese (Mn) is an essential metal required for many enzymatic reactions such as those catalyzed by arginase, glutamine synthetase and manganese-dependent superoxide dismutase. However, elevated Mn exposures can lead to its accumulation in the brain and causes serious neurotoxicity (Crossgrove and Zheng, 2004, Dobson et al., 2004, Erikson et al., 2004a, Hazell, 2002, Newland, 1999, Pal et al., 1999). In humans, Mn deficiency is rare whereas Mn neurotoxicity or manganism can occur due to occupational and environmental exposures (Crossgrove and Zheng, 2004, Pal et al., 1999). It is estimated that over 3700 tons of Mn are released into the atm osphere every year, particularly from the gasoline additive, methylcyclopentadienyl manganese tricarbonyl (MMT). Signs of manganism resemble those of idiopathic Parkinson's disease, including dystonia, bradykinesia, rigidity, and tremor (Barbeau, 1984, Newland, 1999, Pal et al., 1999). In exposed humans and monkeys, Mn accumulates at the highest levels in the striatum, globus pallidus, and substantia nigra (Erikson et al., 2004a). In monkeys dosed for 3 months with MnO2, Mn concentrations reached 264 μM in the striatum and 334 μM in the globus pallidus (Suzuki et al., 1975). Mn exposure may also cause developmental neurotoxicity in children. A recent cross-sectional study of 201 10-year-old children living in Araihazar, Bangladesh showed that water Mn was significantly and adversely associated with Performance and Full Scale raw intelligence score (Wasserman et al., 2006).

Previous studies have suggested that Mn2+ can induce oxidative stress and affect iron metabolism and cellular energy metabolism (Dobson et al., 2004, Erikson et al., 2004a, Hazell, 2002, Li et al., 2005, Lu et al., 2005). Nonetheless, the understanding of the specific mechanisms underlying Mn neurotoxicity in humans remains far from clear. In mammals, Mn is absorbed and transported to the brain by the mechanisms responsible for the uptake of divalent metal ions, including the DMT-1-mediated and transferrin-mediated pathways (Erikson and Aschner, 2006, Erikson et al., 2004b). In the brain, astrocytes are a “sink” for Mn, with concentrations 10–50-fold higher than in neurons (Erikson and Aschner, 2006, Erikson et al., 2004b). Mn transport to astrocytes is significantly affected by Fe status. Also, evidence suggests that Mn transport involves multiple pathways that may be competitive or synergistic with iron transport (Crossgrove and Yokel, 2005, Erikson and Aschner, 2006, Erikson et al., 2004b).

In this report, we focus on the potential effects of Mn on human astrocytes. Astrocytes play active roles in many neuronal functions: maintaining ion and pH homeostasis, promoting the synthesis and removal of neurotransmitters, providing glucose supply and antioxidant defense, and regulating synaptic activity, synaptogenesis and neurogenesis (Auld and Robitaille, 2003, Magisretti and Ransom, 2002, Newman, 2003). Astrocytes are sensors of the brain environment, to which they immediately react on the genomic (gene expression) and non-genomic levels (Nedergaard and Dirnagl, 2005). Inflammatory activation of astrocytes or astrocyte dysfunctions are believed to be associated with several chronic neurological diseases, including prion, Alzheimer's and Parkinson's Diseases, and HIV-1 associated dementia (HAD) (Burwinkel et al., 2004, Choi et al., 2005, Meda et al., 2001, Wang et al., 2004). If Mn neurotoxicity is linked with inflammation, it is plausible that the initiating events may occur in astrocytes. One purpose of this study is to test the hypothesis that Mn may induce the expression of inflammatory mediators in astrocytes.

A new powerful way to begin identifying molecular targets of pathogenic insults to cells is to use microarray technology to obtain a general gene expression profile of affected versus control cells.

Candidate affected genes and their functions can then be subjected to in-depth analysis to determine the mechanisms of their responses to a given stimulus. Here we applied this approach to identify candidate genes and gene families in human primary astrocytes that are affected by exposure of the cells to Mn2+. Furthermore, we applied biochemical and toxicological methods to confirm the prominent effects of Mn2+ in primary human astrocytes. Our studies suggest that Mn2+ selectively affects cell cycle progression and the expression of hypoxia-responsive genes and proinflammatory factors. These results provide new insights into the mechanisms underlying Mn neurotoxicity.

Section snippets

Primary human astrocytes and treatment

Astrocytes were isolated from second trimester (14–19 weeks of gestational age) human fetal brains obtained from elective abortions in full compliance with NIH guidelines, as previously described by Volsky and colleagues (Canki et al., 2001, Wang et al., 2004). Homogenous preparations of astrocytes were obtained using high-density culture conditions in the absence of growth factors in F12 Dulbecco's modified Eagles Medium (DMEM-F12) (GIBCO-Invitrogen, Carlsbad, CA) containing 10% fetal bovine

Mn2+ alters the transcript levels of a subset of genes with distinctive functional characteristics

Previous studies of Mn-exposed monkeys and rats showed that Mn concentration in the striatum and globus pallidus can reach concentrations higher than 200 μM (Erikson et al., 2004a, Suzuki et al., 1975). In addition, Mn concentration in astrocytes can be much higher than in neurons. Thus, to maximize the likelihood of identifying Mn targets that may be relevant to human exposures, we chose to examine the effect of 200 μM Mn on primary human astrocytes. We performed microarray gene expression

Discussion

In this report, we applied microarray gene expression profiling and computational algorithms to identify molecular targets of the common environmental toxicant Mn2+ in primary human astrocytes. Further, we used real-time RT-PCR, Western blotting, flow cytometric, and toxicological analyses to confirm the key results from microarray expression analysis. Our analyses provide several new insights into the molecular neurotoxic actions of Mn. Such insights may provide a molecular basis for further

Acknowledgement

This work is supported in part by public health grants HL65568 (LZ) and NS31492 (GB and DJV).

References (53)

  • A.S. Hazell

    Astrocytes and manganese neurotoxicity

    Neurochem Int

    (2002)
  • G.J. Li et al.

    Alteration at translational but not transcriptional level of transferring receptor expression following manganese exposure at the blood-CSF barrier in vitro

    Toxicol Appl Pharmacol

    (2005)
  • K.J. Livak et al.

    Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method

    Methods

    (2001)
  • L. Lu et al.

    Alteration of serum concentrations of manganese, iron, ferritin, and transferrin receptor following exposure to welding fumes among career welders

    Neurotoxicology

    (2005)
  • D.J. Manalo et al.

    Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1

    Blood

    (2005)
  • R.L. Margolis

    Bub1, a gatekeeper for Cdc20-dependent mitotic exit

    Dev Cell

    (2004)
  • L. Meda et al.

    Glial activation in Alzheimer's disease: the role of Abeta and its associated proteins

    Neurobiol Aging

    (2001)
  • E.A. Newman

    New roles for astrocytes: regulation of synaptic transmission

    Trends Neurosci

    (2003)
  • R.T. Abraham et al.

    Cell biology. Guiding ATM to broken DNA

    Science

    (2005)
  • A. Barbeau

    Manganese and extrapyramidal disorders (a critical review and tribute to Dr. George C. Cotzias)

    Neurotoxicology

    (1984)
  • S.A. Bustin

    Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems

    J Mol Endocrinol

    (2002)
  • M. Canki et al.

    Highly productive infection with pseudotyped human immunodeficiency virus type 1 (HIV-1) indicates no intracellular restrictions to HIV-1 replication in primary human astrocytes

    J Virol

    (2001)
  • V. Castellani

    The function of neuropilin/L1 complex

    Adv Exp Med Biol

    (2002)
  • J. Crossgrove et al.

    Manganese toxicity upon overexposure

    NMR Biomed

    (2004)
  • Z. Darzynkiewicz et al.

    Determining cell cyle stages by flow cytometry

  • A.W. Dobson et al.

    Manganese neurotoxicity

    Ann NY Acad Sci

    (2004)
  • Cited by (35)

    • Neurotoxicity and gene expression alterations in zebrafish larvae in response to manganese exposure

      2022, Science of the Total Environment
      Citation Excerpt :

      Neuroinflammation is one of the principal underlying mechanisms of neuronal injury, and glial cells play an important role in it. It was found that proinflammatory cytokines genes were up-regulated and genes involved in cell cycle regulation and DNA replication and repair were down-regulated in primitive human astrocytes exposed to Mn (Sengupta et al., 2007). Oxidative stress plays an important role in Mn-induced neuronal apoptosis.

    • Neurotoxicity mechanisms of manganese in the central nervous system

      2021, Advances in Neurotoxicology
      Citation Excerpt :

      Astrocytes may also be an important source of proinflammatory factors as Mn induced astrogliosis in various mouse brain regions including striatum and substantia nigra (Krishna et al., 2014; Tomas-Camardiel et al., 2002). Mn increased inflammatory cytokines such as TNF-α, ILs, and interferons, and substances such as cyclooxygenase-2 (COX-2) and prostaglandins in astrocytes (Chen and Liao, 2002; Filipov et al., 2005; Liao et al., 2007; Sengupta et al., 2007). Given that Mn increases proinflammatory cytokines and chemokines in various experimental settings, leading to neuronal cell death, understanding the signaling molecules and pathways (e.g., MAPK, NF-κB) by which Mn promotes the production of proinflammatory factors and inflammation is critical to mitigate the effects of Mn-induced inflammation in the brain.

    • The effects of manganese overexposure on brain health

      2020, Neurochemistry International
      Citation Excerpt :

      Exposure of Mn to astrocytes induced the release of pro-inflammatory cytokines and aggravated the inflammatory response induced by aggregated α-synuclein (Harischandra et al., 2014; Sarkar et al., 2018). Genes encoding pro-inflammatory chemokines and cytokines were altered in the astrocytes treated with Mn, implicating involvement of an inflammatory response in Mn neurotoxicity (Sengupta et al., 2007). Pro-inflammatory cytokines such as tumor necrotic factor (TNF-α) and interleukin 1-beta (IL-1β), primarily produced by glial cells in response to activation of environmental toxicants, increased in Mn-neuroinflammatory-induced injury (Guilarte, 2010).

    • Potential for stem cell treatment in manganism

      2018, Neurochemistry International
      Citation Excerpt :

      Indeed, Mn treatment results in neuronal cell death in which apoptosis is a significant contributor (Desole et al., 1996; Hirata et al., 1998). Oxidative stress in astrocytes causes changes in the expression of genes involved in cellular regulation, inflammation and signal transduction (Sengupta et al., 2007). In MN, neurons die but astrocytes survive, although many of the latter cells appear to be significantly damaged by the insult, reflected in the development of Alzheimer type II astrocytosis (Fig. 3).

    View all citing articles on Scopus
    1

    Contributed to this work equally.

    View full text