Elsevier

Biochemical Pharmacology

Volume 73, Issue 5, 1 March 2007, Pages 694-708
Biochemical Pharmacology

Toxic effects of cobalt in primary cultures of mouse astrocytes: Similarities with hypoxia and role of HIF-1α

https://doi.org/10.1016/j.bcp.2006.11.008Get rights and content

Abstract

Cobalt is suspected to cause memory deficit in humans and was reported to induce neurotoxicity in animal models. We have studied the effects of cobalt in primary cultures of mouse astrocytes. CoCl2 (0.2–0.8 mM) caused dose-dependent ATP depletion, apoptosis (cell shrinkage, phosphatidylserine externalization and chromatin rearrangements) and secondary necrosis. The mitochondria appeared to be a main target of cobalt toxicity, as shown by the loss of mitochondrial membrane potential (ΔΨm) and release from the mitochondria of apoptogenic factors, e.g. apoptosis inducing factor (AIF). Pre-treatment with bongkrekic acid reduced ATP depletion, implicating the involvement of the mitochondrial permeability transition (MPT) pore. Cobalt increased the generation of oxygen radicals, but antioxidants did not prevent toxicity. There was also an impaired response to ATP stimulation, evaluated as a lower raise in intracellular calcium. Similarly to hypoxia and dymethyloxallyl glycine (DMOG), cobalt triggered stabilization of the α-subunit of hypoxia-inducible factor HIF-1 (HIF-1α). This early event was followed by an increased expression of HIF-1 regulated genes, e.g. stress protein HO-1, pro-apoptotic factor Nip3 and iNOS. Although all of the three stimuli activated the HIF-1α pathway and decreased ATP levels, the downstream effects were different. DMOG only inhibited cell proliferation, whereas the other two conditions caused cell death by apoptosis and necrosis. This points to cobalt and hypoxia not only inducing HIF-1α regulated genes but also affecting similarly other cellular functions, including metabolism.

Introduction

Cobalt is an essential element for humans since it is a necessary constituent for the formation of vitamin B12 (hydroxycobalamin). The human body contains about 1–2 mg of cobalt; most of it is found in the liver, kidney, heart and spleen, whereas low concentrations are detected in serum, brain and pancreas [1]. The minimum recommended daily intake of hydroxycobalamin of an adult is 3 μg, corresponding to 0.012 μg of cobalt. Dietary vitamin B12 deficiency is a cause of anemia and increases the risk of developmental abnormalities and growth failure in infants [2]. However, excessive levels of cobalt can be detrimental to the organism. The route of exposure is frequently dermal or via inhalation [3] and occurs mostly in industrial refining, in the production of alloys and in the tungsten carbide hard metal industry. In urban areas mean concentration of cobalt in air is small (about 1–2 ng Co/m3) but in heavily industrialised cities concentrations up to 10 ng/m3 have been reported with concentrations near industrial sources ranging up to 600 ng Co/m3[4]. In occupational setting levels can be as high as 1100 μg Co/m3[5]. Increased levels of cobalt were found in urine and blood of occupationally exposed workers. For example, Ichikawa et al. [6] have reported that cobalt concentration was 0.57–0.79 μg/dl in blood and 59–78 μg/dl in urine following exposure to 100 μg/m3.

Cobalt toxicity includes cardiomyopathy [7], adverse pulmonary effects [8] and carcinogenicity [9]. This heavy metal is also suspected to cause neurotoxic effects, as indicated by the report of memory deficit among workers exposed to hard metal both as dust powder and in mist form [10]. Uptake of cobalt in the nasal mucosa and transport to the olfactory bulbs may be an important route of brain exposure [11]. Experiments performed in the rat have shown that cobalt can cause decreased exploratory behavior [12] and depletion of neurotransmitters [13]. Besides inhibiting synaptic transmission by presynaptic blockade of calcium channels, cobalt can also block postsynaptic responses induced by neurotransmitters in vitro [14].

The data available in the literature indicate that cobalt is cytotoxic to many cell types, including neural cells [15], [16], [17] and can induce cell death by apoptosis and necrosis [18]. Cobalt can cause DNA fragmentation [19], [20], [21], activation of caspases [22], increased production of reactive oxygen species (ROS) [16], [19], [23], augmented phosphorylation of mitogen activated protein (MAP) kinases [17], [22] and elevated levels of p53 [23].

Cobalt has been used to induce ischemic pre-conditioning in vivo [24]. Some of the characteristic effects of cobalt are thought to be mediated by interaction with the cellular oxygen-sensing machinery. Like low oxygen tension, cobalt at normoxic conditions is able to stabilize the α-subunit of hypoxia-inducible factor HIF-1 by blocking its ubiquitination and proteasomal degradation [25]. Increased levels of HIF-1α result in higher transcription of a set of genes that encode several proteins, e.g. glycolytic enzymes, erythropoietin and heat shock proteins, important for the adaptation of cells to hypoxic stress [26], [27]. Many of the gene products regulated by HIF-1α are involved in a physiological response to promote cell survival and recovery after hypoxia, e.g. by maintenance of cellular energy supplies and stimulation of angiogenesis. However, recent work has also pointed to the increased transcription of pro-apoptotic factors, i.e. NIP3/BNIP3 and NIX [28], which can lead to cell death. Because of these effects cobalt ions may be used to elucidate the roles of HIF-1α in different types of cells.

Astrocytes play a crucial role in regulating functions and survival of neurons and controlling synapses [29], [30]. Although glia are generally considered more resistant to stress conditions as compared to neuronal cells, recent studies have indicated that astrocytes are also susceptible to a variety of neurotoxic stimuli, including hypoxic injury and heavy metals [31], [32], [33], [34], [35], [36], [37], [38]. It was reported that cobalt can activate extracellular signal-regulated protein kinase1/2 (ERK1/2) and cause cell death by apoptosis in C6 glioma cells [17], but otherwise little is known about cobalt effects in astrocytes. The purpose of the present study was twofold. First, we aimed to characterize cobalt cytotoxicity in primary astrocytes, by investigating the possible involvement of HIF-1 regulated genes, oxidative stress and calcium signaling, as well as by looking at mechanisms of cell death. On the other hand, we wanted to verify the hypothesis that cobalt is a valuable mimic of hypoxia in astrocytes. For this purpose, we have compared the effects of cobalt exposure to oxygen deprivation.

Section snippets

Chemicals

All cell culture reagents, including media, antibiotics, fetal bovine serum (FBS) and phosphate buffered saline (PBS) were purchased from Gibco-Life Technologies (Täby, Sweden) and cell culture plastics from Corning Incorporated (Schipol-Rijk, NE). The RNeasy kit was obtained from Qiagen (VWR International, Stockholm, Sweden). The GeneAmp RNA PCR kit and the TaqMan Universal PCR master mix were purchased from Applied Biosystems (Applera Sweden, Stockholm). The fluorescein isothiocyanate

Features of cobalt toxicity

Primary mouse astrocytes exposed to 0.2–0.8 mM CoCl2 displayed both biochemical and morphological signs of cellular toxicity. These levels of cobalt are several orders of magnitude higher than plasma levels encountered in human exposure but similar to the doses used in previous experiments to affect metabolism. All of the doses tested induced a significant decrease of the total ATP level after 24 h (Fig. 1A). Depletion of ATP can be a consequence of metabolic alterations resulting in deficient

Discussion

Exposure to cobalt induced multiple alterations in primary astrocytes. Early events included stabilization of HIF-1α, resulting in increased expression of HIF-1 regulated genes, e.g. stress protein HO-1, pro-apoptotic factor NIP3 and iNOS. There was also altered calcium signaling and increased generation of oxygen radicals. Mitochondria were a key target of cobalt toxicity, as shown by loss of ΔΨm, opening of the MPT pore and release of AIF. This led to apoptosis, i.e. cell shrinkage, PS

Acknowledgments

The authors are grateful to Dr. Ping Sheng Hu for valuable help with the intracellular calcium measurements. This work was supported by Karolinska Institutet, the Swedish Science Research Council (2553), European Commission (LSHM-CT-2005-518189), the Swedish Brain Foundation, the T. Nilson Foundation, the Swedish Society of Medicine, the Å. Wiberg Foundation and the General Maternity Hospital foundation. The funding agencies do not take any responsibility for the contents of the article.

References (80)

  • S. Gupta

    Molecular steps of death receptor and mitochondrial pathways of apoptosis

    Life Sci

    (2001)
  • V.L. Gabai et al.

    Role of Hsp70 in regulation of stress-kinase JNK: implications in apoptosis and aging

    FEBS Lett

    (1998)
  • J. Pourahmad et al.

    Carcinogenic metal induced sites of reactive oxygen species formation in hepatocytes

    Toxicol In Vitro

    (2003)
  • O.I. Aruoma et al.

    The antioxidant action of N-acetylcysteine: its reaction with hydrogen peroxide, hydroxyl radical, superoxide, and hypochlorous acid

    Free Radic Biol Med

    (1989)
  • T.W. Wu et al.

    Propyl gallate as a hepatoprotector in vitro and in vivo

    Biochem Pharmacol

    (1994)
  • A.D. Mooradian

    Antioxidant properties of steroids

    J Steroid Biochem Mol Biol

    (1993)
  • N. Hatori et al.

    Acute cobalt exposure and oxygen radical scavengers in the rat myocardium

    Biochim Biophys Acta

    (1993)
  • M.B. Kadiiska et al.

    A comparison of cobalt(II) and iron(II) hydroxyl and superoxide free radical formation

    Arch Biochem Biophys

    (1989)
  • M.L. Tomaro et al.

    Heme oxygenase induction by CoCl2, Co-protoporphyrin IX, phenylhydrazine, and diamide: evidence for oxidative stress involvement

    Arch Biochem Biophys

    (1991)
  • P.K. Aley et al.

    Hypoxia stimulates Ca2+ release from intracellular stores in astrocytes via cyclic ADP ribose-mediated activation of ryanodine receptors

    Cell Calcium

    (2006)
  • C.M. Wiener et al.

    In vivo expression of mRNAs encoding hypoxia-inducible factor 1

    Biochem Biophys Res Commun

    (1996)
  • K. Yamaura et al.

    Contribution of epoxyeicosatrienoic acids to the hypoxia-induced activation of Ca(2+)-activated K(+) channel current in cultured rat hippocampal astrocytes

    Neuroscience

    (2006)
  • B.A. MacVicar

    Morphological differentiation of cultured astrocytes is blocked by cadmium or cobalt

    Brain Res

    (1987)
  • C. Coddou et al.

    Heavy metals modulate the activity of the purinergic P2X4 receptor

    Toxicol Appl Pharmacol

    (2005)
  • C. Elinder et al.

    Cobalt

  • S.P. Stabler et al.

    Vitamin B12 deficiency as a worldwide problem

    Annu Rev Nutr

    (2004)
  • A.A. Jensen et al.

    Cobalt exposure and cancer risk

    Crit Rev Toxicol

    (1990)
  • D.G. Barceloux

    Cobalt

    J Toxicol Clin Toxicol

    (1999)
  • N. Palmen

    Cobalt and cobalt compounds. Criteria document for Swedish occupational standards

  • Y. Ichikawa et al.

    Biological monitoring of cobalt exposure, based on cobalt concentrations in blood and urine

    Int Arch Occup Environ Health

    (1985)
  • H. Kesteloot et al.

    An enquiry into the role of cobalt in the heart disease of chronic beer drinkers

    Circulation

    (1968)
  • A.P. Wehner et al.

    Chronic inhalation of cobalt oxide and cigarette smoke by hamsters

    Am Ind Hyg Assoc J

    (1977)
  • J.C. Heath

    The production of malignant tumours by cobalt in the rat

    Br J Cancer

    (1956)
  • C.M. Jordan et al.

    Memory deficits and industrial toxicant exposure: a comparative study of hard metal, solvent and asbestos workers

    Int J Neurosci

    (1997)
  • M. Czarnota et al.

    Activity and passive-avoidance learning in cobalt-injected rats

    Int J Neurosci

    (1998)
  • M. Hasan et al.

    Cobalt-induced depletion of dopamine, norepinephrine & 5-hydroxytryptamine concentration in different regions of the rat brain

    Indian J Exp Biol

    (1980)
  • G. Wang et al.

    Mitochondrial DNA damage and a hypoxic response are induced by CoCl(2) in rat neuronal PC12 cells

    Nucleic Acids Res

    (2000)
  • G. Olivieri et al.

    Melatonin protects SHSY5Y neuroblastoma cells from cobalt-induced oxidative stress, neurotoxicity and increased beta-amyloid secretion

    J Pineal Res

    (2001)
  • S.J. Yang et al.

    Cobalt chloride-induced apoptosis and extracellular signal-regulated protein kinase 1/2 activation in rat C6 glioma cells

    J Biochem Mol Biol

    (2004)
  • W. Zou et al.

    Cobalt chloride induces PC12 cells apoptosis through reactive oxygen species and accompanied by AP-1 activation

    J Neurosci Res

    (2001)
  • Cited by (142)

    • Colorimetric and fluorescent Schiff base sensors for trace detection of pollutants and biologically significant cations: A review (2010–2021)

      2022, Microchemical Journal
      Citation Excerpt :

      Cobalamin is a water-soluble and cobalt-containing co-enzyme that is important for red blood cell formation, protein synthesis, in the treatment of high cholesterol and fatigue, and the health of nerve tissues [237–239]. Unregulated exposure to cobalt may lead to several health issues, including pneumonia, dermatitis, hypothyroidism, lung cancer, hearing loss, asthma, etc. [240–243]. So, molecular sensors are needed to control the amount of cobalt in biological samples.

    View all citing articles on Scopus

    This work was supported by Karolinska Institutet, the Swedish Science Research Council (2553), European Commission (LSHM-CT-2005-518189), the Swedish Brain Foundation, the T. Nilson Foundation, the Swedish Society of Medicine, the Å. Wiberg Foundation and the General Maternity Hospital foundation. The funding agencies do not take any responsibility for the contents of the article.

    View full text