Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Environ Sci Technol ; 58(36): 15926-15937, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39190186

ABSTRACT

This study demonstrated the strengths of in vivo molecular staining coupled with automated imaging analysis in Daphnia magna. A multiwell plate protocol was developed to assess mitochondrial membrane potential using the JC-1 dye. The suitability of five common anesthetics was initially tested, and 5% ethanol performed best in terms of anesthetic effects and healthy recovery. The staining conditions were optimized to 30 min staining with 2 µM JC-1 for best J-aggregate formation. The protocol was validated with the model compound carbonyl cyanide 3-chlorophenylhydrazone (CCCP) and used to measure the effect of four environmental contaminants, 2,4-dinitrophenol, triclosan, n-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD), and ibuprofen, on mitochondrial health. Test organisms were imaged using an automated confocal microscope, and fluorescence intensities were automatically quantified. The effect concentrations for CCCP were lower by a factor of 30 compared with the traditional OECD 202 acute toxicity test. Mitochondrial effects were also detected at lower concentrations for all tested environmental contaminants compared to the OCED 202 test. For 2,4-dinitrophenol, mitochondria effects were detectable after 2 h exposure to environmentally relevant concentrations and predicted organism death was observed after 24 h. The high sensitivity and time efficiency of this novel automated imaging method make it a valuable tool for advancing ecotoxicological testing.


Subject(s)
Daphnia , Membrane Potential, Mitochondrial , Animals , Daphnia/drug effects , Membrane Potential, Mitochondrial/drug effects , Ecotoxicology , Fluorescence , Water Pollutants, Chemical/toxicity , Daphnia magna
2.
Arch Toxicol ; 98(8): 2695-2709, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38769170

ABSTRACT

To improve the mechanistic screening of reproductive toxicants in  chemical-risk assessment and drug development, we have developed a three-dimensional (3D) heterogenous testicular co-culture model from neonatal mice. Di-n-butyl phthalate (DBP), an environmental contaminant that can affect reproductive health negatively, was used as a model compound to illustrate the utility of the in vitro model. The cells were treated with DBP (1 nM to 100 µM) for 7 days. Automated high-content imaging confirmed the presence of cell-specific markers of Leydig cells (CYP11A1 +), Sertoli cells (SOX9 +), and germ cells (DAZL +). Steroidogenic activity of Leydig cells was demonstrated by analyzing testosterone levels in the culture medium. DBP induced a concentration-dependent reduction in testosterone levels and decreased the number of Leydig cells compared to vehicle control. The levels of steroidogenic regulator StAR and the steroidogenic enzyme CYP11A1 were decreased already at the lowest DBP concentration (1 nM), demonstrating upstream effects in the testosterone biosynthesis pathway. Furthermore, exposure to 10 nM DBP decreased the levels of the germ cell-specific RNA binding protein DAZL, central for the spermatogenesis. The 3D model also captured the development of the Sertoli cell junction proteins, N-cadherin and Zonula occludens protein 1 (ZO-1), critical for the blood-testis barrier. However, DBP exposure did not significantly alter the cadherin and ZO-1 levels. Altogether, this 3D in vitro system models testicular cellular signaling and function, making it a powerful tool for mechanistic screening of developmental testicular toxicity. This can open a new avenue for high throughput screening of chemically-induced reproductive toxicity during sensitive developmental phases.


Subject(s)
Coculture Techniques , Dibutyl Phthalate , Leydig Cells , Sertoli Cells , Testis , Testosterone , Animals , Male , Testis/drug effects , Testis/metabolism , Leydig Cells/drug effects , Leydig Cells/metabolism , Dibutyl Phthalate/toxicity , Testosterone/metabolism , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Mice , Reproduction/drug effects , Dose-Response Relationship, Drug , Environmental Pollutants/toxicity , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Animals, Newborn
3.
Ecotoxicol Environ Saf ; 262: 115321, 2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37549549

ABSTRACT

Di-n-butyl phthalate (DBP) is a ubiquitous environmental contaminant linked with various adverse health effects, including immune system dysfunction. Gut microbial dysbiosis can contribute to a wide range of pathogenesis, particularly immune disease. Here, we investigated the impact of DBP on the gut microbiome and examined correlations with immune system changes after five weeks oral exposure (10 or 100 mg/kg/day) in adult male mice. The fecal microbiome composition was characterized using 16S rRNA sequencing. DBP-treated mice displayed a significantly distinct microbial community composition, indicated by Bray-Curtis distance. Numerous amplicon sequence variants (ASVs) at the genus level were altered. Compared to the vehicle control group, the 10 mg/kg/day DBP group had 63 more abundant and 65 less abundant ASVs, while 60 ASVs were increased and 76 ASVs were decreased in the 100 mg/kg/day DBP group. Both DBP treatment groups showed higher abundances of ASVs assigned to Desulfovibrio (Proteobacteria phylum) and Enterorhabdus genera, while ASVs belonging to Parabacteroides, Lachnospiraceae UCG-006 and Lachnoclostridium were less common compared to the control group. Interestingly, an ASV belonging to Rumniniclostridium 6, which was less abundant in DBP-treated mice, demonstrated a negative correlation with the increased number of non-classical monocytes observed in the blood of DBP-treated animals. In addition, an ASV from Lachnospiraceae UCG-001, which was more abundant in the DBP-treated animals, showed a positive correlation with the non-classical monocyte increase. This study shows that DBP exposure greatly modifies the gut bacterial microbiome and indicates a potential contribution of microbial dysbiosis to DBP-induced immune system impairment, illustrating the importance of investigating how interactions between exposome components can affect health.

4.
Int J Mol Sci ; 23(15)2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35955852

ABSTRACT

Studies indicate that phthalates are endocrine disruptors affecting reproductive health. One of the most commonly used phthalates, di-n-butyl phthalate (DBP), has been linked with adverse reproductive health outcomes in men, but the mechanisms behind these effects are still poorly understood. Here, adult male mice were orally exposed to DBP (10 or 100 mg/kg/day) for five weeks, and the testis and adrenal glands were collected one week after the last dose, to examine more persistent effects. Quantification of testosterone, androstenedione, progesterone and corticosterone concentrations by liquid chromatography-mass spectrometry showed that testicular testosterone was significantly decreased in both DBP treatment groups, whereas the other steroids were not significantly altered. Western blot analysis of testis revealed that DBP exposure increased the levels of the steroidogenic enzymes CYP11A1, HSD3ß2, and CYP17A1, the oxidative stress marker nitrotyrosine, and the luteinizing hormone receptor (LHR). The analysis further demonstrated increased levels of the germ cell marker DAZL, the Sertoli cell markers vimentin and SOX9, and the Leydig cell marker SULT1E1. Overall, the present work provides more mechanistic understanding of how adult DBP exposure can induce effects on the male reproductive system by affecting several key cells and proteins important for testosterone biosynthesis and spermatogenesis, and for the first time shows that these effects persist at least one week after the last dose. It also demonstrates impairment of testosterone biosynthesis at a lower dose than previously reported.


Subject(s)
Dibutyl Phthalate , Testis , Animals , Dibutyl Phthalate/metabolism , Humans , Leydig Cells/metabolism , Male , Mice , Spermatogenesis , Testis/metabolism , Testosterone/metabolism
6.
Arch Toxicol ; 94(11): 3893-3906, 2020 11.
Article in English | MEDLINE | ID: mdl-32700164

ABSTRACT

Gene-environment interactions are involved in the development of breast cancer, the tumor type that accounts for the majority of the cancer-related deaths among women. Here, we demonstrate that exposure to PFOS (10 µM) and PFOA (100 µM)-two contaminants ubiquitously found in human blood-for 72 h induced breast epithelial cell (MCF-10A cell line) proliferation and alteration of regulatory cell-cycle proteins (cyclin D1, CDK6, p21, p53, p27, ERK 1/2 and p38) that persisted after a multitude of cell divisions. The contaminants also promoted cell migration and invasion by reducing the levels of E-cadherin, occludin and ß-integrin in the unexposed daughter cells. The compounds further induced an increase in global DNA methylation and differentially altered histone modifications, epigenetic mechanisms implicated in tumorigenesis. This mechanistic evidence for PFOS- and PFOA-induced malignant transformation of human breast cells supports a role of these abundant contaminants in the development and progression of breast cancer. Increased knowledge of contaminant-induced effects and their contribution to breast tumorigenesis is important for a better understanding of gene-environment interactions in the etiology of breast cancer.


Subject(s)
Alkanesulfonic Acids/toxicity , Breast Neoplasms/chemically induced , Caprylates/toxicity , Carcinogenesis/chemically induced , Cell Cycle Proteins/metabolism , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Fluorocarbons/toxicity , Breast Neoplasms/genetics , Carcinogenesis/genetics , Cell Cycle/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Female , Humans
7.
Arch Toxicol ; 94(8): 2799-2808, 2020 08.
Article in English | MEDLINE | ID: mdl-32435914

ABSTRACT

Olfactory dysfunction is implicated in neurodegenerative disorders and typically manifests years before other symptoms. The cyanobacterial neurotoxin ß-N-methylamino-L-alanine (BMAA) is suggested as a risk factor for neurodegenerative disease. Detection of BMAA in air filters has increased the concern that aerosolization may lead to human BMAA exposure through the air. The aim of this study was to determine if BMAA targets the olfactory system. Autoradiographic imaging showed a distinct localization of radioactivity in the right olfactory mucosa and bulb following a unilateral intranasal instillation of 3H-BMAA (0.018 µg) in mice, demonstrating a direct transfer of BMAA via the olfactory pathways to the brain circumventing the blood-brain barrier, which was confirmed by liquid scintillation. Treatment of mouse primary olfactory bulb cells with 100 µM BMAA for 24 h caused a disruption of the neurite network, formation of dendritic varicosities and reduced cell viability. The NMDA receptor antagonist MK-801 and the metabotropic glutamate receptor antagonist MCPG protected against the BMAA-induced alterations, demonstrating the importance of glutamatergic mechanisms. The ionotropic non-NMDA receptor antagonist CNQX prevented the BMAA-induced decrease of cell viability in mixed cultures containing both neuronal and glial cells, but not in cultures with neurons only, suggesting a role of neuron-glial interactions and glial AMPA receptors in the BMAA-induced toxicity. The results show that the olfactory region may be a target for BMAA following inhalation exposure. Further studies on the relations between environmental olfactory toxicants and neurodegenerative disorders are warranted.


Subject(s)
Amino Acids, Diamino/toxicity , Bacterial Toxins/toxicity , Cyanobacteria/metabolism , Neuroglia/drug effects , Neurons/drug effects , Olfactory Bulb/drug effects , Administration, Intranasal , Amino Acids, Diamino/administration & dosage , Amino Acids, Diamino/metabolism , Animals , Bacterial Toxins/administration & dosage , Bacterial Toxins/metabolism , Cell Survival/drug effects , Cells, Cultured , Cyanobacteria Toxins , Glutamic Acid/metabolism , Male , Mice, Inbred C57BL , Neuroglia/metabolism , Neuroglia/pathology , Neuronal Outgrowth/drug effects , Neurons/metabolism , Neurons/pathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Olfactory Mucosa/metabolism
8.
J Pineal Res ; 65(1): e12488, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29528516

ABSTRACT

The environmental neurotoxin ß-N-methylamino-L-alanine (BMAA) is a glutamate receptor agonist that can induce oxidative stress and has been implicated as a possible risk factor for neurodegenerative disease. Detection of BMAA in mussels, crustaceans, and fish illustrates that the sources of human exposure to this toxin are more abundant than previously anticipated. The aim of this study was to determine uptake of BMAA in the pineal gland and subsequent effects on melatonin production in primary pinealocyte cultures and a rat model. Autoradiographic imaging of 10-day-old male rats revealed a high and selective uptake in the pineal gland at 30 minutes to 24 hours after 14 C-L-BMAA administration (0.68 mg/kg). Primary pinealocyte cultures exposed to 0.05-3 mmol/L BMAA showed a 57%-93% decrease in melatonin synthesis in vitro. Both the metabotropic glutamate receptor 3 (mGluR3) antagonist Ly341495 and the protein kinase C (PKC) activator phorbol-12-myristate-13-acetate prevented the decrease in melatonin secretion, suggesting that BMAA inhibits melatonin synthesis by mGluR3 activation and PKC inhibition. Serum analysis revealed a 45% decrease in melatonin concentration in neonatal rats assessed 2 weeks after BMAA administration (460 mg/kg) and confirmed an inhibition of melatonin synthesis in vivo. Given that melatonin is a most important neuroprotective molecule in the brain, the etiology of BMAA-induced neurodegeneration may include mechanisms beyond direct excitotoxicity and oxidative stress.


Subject(s)
Amino Acids, Diamino/pharmacology , Melatonin/metabolism , Amino Acids/pharmacology , Animals , Cyanobacteria Toxins , Female , Male , Oxidative Stress/drug effects , Phorbol Esters/pharmacology , Pineal Gland/cytology , Pineal Gland/metabolism , Protein Kinase C/metabolism , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Xanthenes/pharmacology
9.
Arch Toxicol ; 92(2): 705-716, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29063134

ABSTRACT

Perfluorooctanesulfonic acid (PFOS) is a synthetic fluorosurfactant widely used in the industry and a prominent environmental toxicant. PFOS is persistent, bioaccumulative, and toxic to mammalian species. Growing evidence suggests that PFOS has the potential to interfere with estrogen homeostasis, posing a risk of endocrine-disrupting effects. Recently, concerns about a potential link between PFOS and breast cancer have been raised, but the mechanisms underlying its actions as a potential carcinogen are unknown. By utilizing cell proliferation assays, flow cytometry, immunocytochemistry, and cell migration/invasion assays, we examined the potentially tumorigenic activity of PFOS (100 nM-1 mM) in MCF-10A breast cell line. The results showed that the growth of MCF-10A cells exposed to 1 and 10 µM PFOS was higher compared to that of the control. Mechanistic studies using 10 µM PFOS demonstrated that the compound promotes MCF-10A proliferation through accelerating G0/G1-to-S phase transition of the cell cycle after 24, 48, and 72 h of treatment. In addition, PFOS exposure increased CDK4 and decreased p27, p21, and p53 levels in the cells. Importantly, treatment with 10 µM PFOS for 72 h also stimulated MCF-10A cell migration and invasion, illustrating its capability to induce neoplastic transformation of human breast epithelial cells. Our experimental results suggest that exposure to low levels of PFOS might be a potential risk factor in human breast cancer initiation and development.


Subject(s)
Alkanesulfonic Acids/toxicity , Breast/cytology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Fluorocarbons/toxicity , Breast Neoplasms/pathology , Cell Line , Cell Survival , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epithelial Cells/cytology , Estrogen Receptor alpha/metabolism , Female , Humans , Tumor Suppressor Protein p53/metabolism
10.
Arch Toxicol ; 92(5): 1729-1739, 2018 05.
Article in English | MEDLINE | ID: mdl-29502166

ABSTRACT

Despite significant advances in early detection and treatment, breast cancer remains a major cause of morbidity and mortality. Perfluorooctanoic acid (PFOA) is a suspected endocrine disruptor and a common environmental pollutant associated with various diseases including cancer. However, the effects of PFOA and its mechanisms of action on hormone-responsive cells remain unclear. Here, we explored the potential tumorigenic activity of PFOA (100 nM-1 mM) in human breast epithelial cells (MCF-10A). MCF-10A cells exposed to 50 and 100 µM PFOA demonstrated a higher growth rate compared to controls. The compound promoted MCF-10A proliferation by accelerating G0/G1 to S phase transition of the cell cycle. PFOA increased cyclin D1 and CDK4/6 levels, concomitant with a decrease in p27. In contrast to previous studies of perfluorooctane sulfate (PFOS), the estrogen receptor antagonist ICI 182,780 had no effect on PFOA-induced cell proliferation, whereas the PPARα antagonist GW 6471 was able to prevent the MCF-10A proliferation, indicating that the underlying mechanisms involve PPARα-dependent pathways. Interestingly, we also showed that PFOA is able to stimulate cell migration and invasion, demonstrating its potential to induce neoplastic transformation of human breast epithelial cells. These results suggest that more attention should be paid to the roles of PFOA in the development and progression of breast cancer.


Subject(s)
Caprylates/toxicity , Epithelial Cells/drug effects , Fluorocarbons/toxicity , Mammary Glands, Human/cytology , Breast Neoplasms/chemically induced , Breast Neoplasms/pathology , Caprylates/administration & dosage , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Endocrine Disruptors/administration & dosage , Endocrine Disruptors/toxicity , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fluorocarbons/administration & dosage , Humans , Mammary Glands, Human/drug effects , Oxazoles/pharmacology , PPAR alpha/antagonists & inhibitors , Receptors, Estrogen/metabolism , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
12.
Metab Brain Dis ; 33(3): 693-704, 2018 06.
Article in English | MEDLINE | ID: mdl-29288365

ABSTRACT

Although methylphenidate (MPH) is ubiquitously prescribed to children and adolescents, the consequences of chronic utilization of this psychostimulant are poorly understood. In this study, we investigated the effects of MPH on cytoskeletal homeostasis and lipid content in rat hippocampus. Wistar rats received intraperitoneal injections of MPH (2.0 mg/kg) or saline solution (controls), once a day, from the 15th to the 44th day of age. Results showed that MPH provoked hypophosphorylation of glial fibrillary acidic protein (GFAP) and reduced its immunocontent. Middle and high molecular weight neurofilament subunits (NF-M, NF-H) were hypophosphorylated by MPH on KSP repeat tail domains, while NFL, NFM and NFH immunocontents were not altered. MPH increased protein phosphatase 1 (PP1) and 2A (PP2A) immunocontents. MPH also decreased the total content of ganglioside and phospholipid, as well as the main brain gangliosides (GM1, GD1a, and GD1b) and the major brain phospholipids (sphingomyelin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, and phosphatidylserine). Total cholesterol content was also reduced in the hippocampi of juvenile rats treated with MPH. These results provide evidence that disruptions of cytoskeletal and lipid homeostasis in hippocampus of juvenile rats are triggers by chronic MPH treatment and present a new basis for understanding the effects and consequences associated with chronic use of this psychostimulant during the development of the central nervous system.


Subject(s)
Cytoskeleton/drug effects , Hippocampus/drug effects , Homeostasis/drug effects , Lipid Metabolism/drug effects , Methylphenidate/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Lipids , Male , Rats, Wistar
13.
Biochim Biophys Acta ; 1863(12): 3001-3014, 2016 12.
Article in English | MEDLINE | ID: mdl-27663072

ABSTRACT

QUIN is a glutamate agonist playing a role in the misregulation of the cytoskeleton, which is associated with neurodegeneration in rats. In this study, we focused on microglial activation, FGF2/Erk signaling, gap junctions (GJs), inflammatory parameters and redox imbalance acting on cytoskeletal dynamics of the in QUIN-treated neural cells of rat striatum. FGF-2/Erk signaling was not altered in QUIN-treated primary astrocytes or neurons, however cytoskeleton was disrupted. In co-cultured astrocytes and neurons, QUIN-activated FGF2/Erk signaling prevented the cytoskeleton from remodeling. In mixed cultures (astrocyte, neuron, microglia), QUIN-induced FGF-2 increased level failed to activate Erk and promoted cytoskeletal destabilization. The effects of QUIN in mixed cultures involved redox imbalance upstream of Erk activation. Decreased connexin 43 (Cx43) immunocontent and functional GJs, was also coincident with disruption of the cytoskeleton in primary astrocytes and mixed cultures. We postulate that in interacting astrocytes and neurons the cytoskeleton is preserved against the insult of QUIN by activation of FGF-2/Erk signaling and proper cell-cell interaction through GJs. In mixed cultures, the FGF-2/Erk signaling is blocked by the redox imbalance associated with microglial activation and disturbed cell communication, disrupting the cytoskeleton. Thus, QUIN signal activates differential mechanisms that could stabilize or destabilize the cytoskeleton of striatal astrocytes and neurons in culture, and glial cells play a pivotal role in these responses preserving or disrupting a combination of signaling pathways and cell-cell interactions. Taken together, our findings shed light into the complex role of the active interaction of astrocytes, neurons and microglia in the neurotoxicity of QUIN.


Subject(s)
Astrocytes/drug effects , Cytoskeleton/drug effects , Excitatory Amino Acid Agonists/toxicity , Microglia/drug effects , Quinolinic Acid/toxicity , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Communication/drug effects , Coculture Techniques , Connexin 43/genetics , Connexin 43/metabolism , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Gene Expression Regulation , MAP Kinase Signaling System , Microglia/cytology , Microglia/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction , Pregnancy , Primary Cell Culture , Rats , Rats, Wistar
14.
Biochim Biophys Acta ; 1860(11 Pt A): 2510-2520, 2016 11.
Article in English | MEDLINE | ID: mdl-27475002

ABSTRACT

BACKGROUND: Diphenylditelluride (PhTe)2 is a potent neurotoxin disrupting the homeostasis of the cytoskeleton. METHODS: Cultured astrocytes and neurons were incubated with (PhTe)2, receptor antagonists and enzyme inhibitors followed by measurement of the incorporation of [32P]orthophosphate into intermediate filaments (IFs). RESULTS: (PhTe)2 caused hyperphosphorylation of glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits (NFL, NFM and NFH) from primary astrocytes and neurons, respectively. These mechanisms were mediated by N-methyl-d-aspartate (NMDA) receptors, L-type voltage-dependent calcium channels (L-VDCCs) as well as metabotropic glutamate receptors upstream of phospholipase C (PLC). Upregulated Ca(2+) influx activated protein kinase A (PKA) and protein kinase C (PKC) in astrocytes causing hyperphosphorylation of GFAP and vimentin. Hyperphosphorylated (IF) together with RhoA-activated stress fiber formation, disrupted the cytoskeleton leading to altered cell morphology. In neurons, the high intracellular Ca(2+) levels activated the MAPKs, Erk and p38MAPK, beyond PKA and PKC, provoking hyperphosphorylation of NFM, NFH and NFL. CONCLUSIONS: Our findings support that intracellular Ca(2+) is one of the crucial signals that modulate the action of (PhTe)2 in isolated cortical astrocytes and neurons modulating the response of the cytoskeleton against the insult. GENERAL SIGNIFICANCE: Cytoskeletal misregulation is associated with neurodegeneration. This compound could be a valuable tool to induce molecular changes similar to those found in different pathologies of the brain.


Subject(s)
Actin Cytoskeleton/metabolism , Astrocytes/drug effects , Benzene Derivatives/pharmacology , Calcium Signaling , Neurons/drug effects , Organometallic Compounds/pharmacology , Animals , Astrocytes/metabolism , Benzene Derivatives/toxicity , Cells, Cultured , Neurons/metabolism , Organometallic Compounds/toxicity , Rats , Rats, Wistar
15.
J Neurosci Res ; 93(2): 268-84, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25306914

ABSTRACT

Quinolinic acid (QUIN) is an endogenous metabolite of the kynurenine pathway involved in several neurological disorders. Among the several mechanisms involved in QUIN-mediated toxicity, disruption of the cytoskeleton has been demonstrated in striatally injected rats and in striatal slices. The present work searched for the actions of QUIN in primary striatal neurons. Neurons exposed to 10 µM QUIN presented hyperphosphorylated neurofilament (NF) subunits (NFL, NFM, and NFH). Hyperphosphorylation was abrogated in the presence of protein kinase A and protein kinase C inhibitors H89 (20 µM) and staurosporine (10 nM), respectively, as well as by specific antagonists to N-methyl-D-aspartate (50 µM DL-AP5) and metabotropic glutamate receptor 1 (100 µM MPEP). Also, intra- and extracellular Ca(2+) chelators (10 µM BAPTA-AM and 1 mM EGTA, respectively) and Ca(2+) influx through L-type voltage-dependent Ca(2+) channel (10 µM verapamil) are implicated in QUIN-mediated effects. Cells immunostained for the neuronal markers ßIII-tubulin and microtubule-associated protein 2 showed altered neurite/neuron ratios and neurite outgrowth. NF hyperphosphorylation and morphological alterations were totally prevented by conditioned medium from QUIN-treated astrocytes. Cocultured astrocytes and neurons interacted with one another reciprocally, protecting them against QUIN injury. Cocultured cells preserved their cytoskeletal organization and cell morphology together with unaltered activity of the phosphorylating system associated with the cytoskeleton. This article describes cytoskeletal disruption as one of the most relevant actions of QUIN toxicity in striatal neurons in culture with soluble factors secreted by astrocytes, with neuron-astrocyte interaction playing a role in neuroprotection.


Subject(s)
Astrocytes/physiology , Cell Communication/physiology , Corpus Striatum/cytology , Cytoskeleton/metabolism , Homeostasis/drug effects , Neurons/drug effects , Quinolinic Acid/pharmacology , Animals , Animals, Newborn , Astrocytes/chemistry , Cell Communication/drug effects , Cell Survival/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Phosphorylation/drug effects , Pregnancy , Rats , Rats, Wistar , Valine/analogs & derivatives , Valine/pharmacology
16.
Exp Cell Res ; 322(2): 313-23, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24583400

ABSTRACT

Quinolinic acid (QUIN) is a glutamate agonist which markedly enhances the vulnerability of neural cells to excitotoxicity. QUIN is produced from the amino acid tryptophan through the kynurenine pathway (KP). Dysregulation of this pathway is associated with neurodegenerative conditions. In this study we treated striatal astrocytes in culture with QUIN and assayed the endogenous phosphorylating system associated with glial fibrillary acidic protein (GFAP) and vimentin as well as cytoskeletal remodeling. After 24h incubation with 100 µM QUIN, cells were exposed to (32)P-orthophosphate and/or protein kinase A (PKA), protein kinase dependent of Ca(2+)/calmodulin II (PKCaMII) or protein kinase C (PKC) inhibitors, H89 (20 µM), KN93 (10 µM) and staurosporin (10nM), respectively. Results showed that hyperphosphorylation was abrogated by PKA and PKC inhibitors but not by the PKCaMII inhibitor. The specific antagonists to ionotropic NMDA and non-NMDA (50 µM DL-AP5 and CNQX, respectively) glutamate receptors as well as to metabotropic glutamate receptor (mGLUR; 50 µM MCPG), mGLUR1 (100 µM MPEP) and mGLUR5 (10 µM 4C3HPG) prevented the hyperphosphorylation provoked by QUIN. Also, intra and extracellular Ca(2+) quelators (1mM EGTA; 10 µM BAPTA-AM, respectively) prevented QUIN-mediated effect, while Ca(2+) influx through voltage-dependent Ca(2+) channel type L (L-VDCC) (blocker: 10 µM verapamil) is not implicated in this effect. Morphological analysis showed dramatically altered actin cytoskeleton with concomitant change of morphology to fusiform and/or flattened cells with retracted cytoplasm and disruption of the GFAP meshwork, supporting misregulation of actin cytoskeleton. Both hyperphosphorylation and cytoskeletal remodeling were reversed 24h after QUIN removal. Astrocytes are highly plastic cells and the vulnerability of astrocyte cytoskeleton may have important implications for understanding the neurotoxicity of QUIN in neurodegenerative disorders.


Subject(s)
Actin Cytoskeleton/drug effects , Astrocytes/cytology , Corpus Striatum/cytology , Quinolinic Acid/pharmacology , Actin Cytoskeleton/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamates/metabolism , Immunoenzyme Techniques , Phosphorylation/drug effects , Pregnancy , Rats , Rats, Wistar , Vimentin/metabolism
17.
Sci Total Environ ; 949: 174864, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39032741

ABSTRACT

DNA methylation plays a pivotal role in cancer. The ubiquitous contaminant perfluorooctanesulfonic acid (PFOS) has been epidemiologically associated with breast cancer, and can induce proliferation and malignant transformation of normal human breast epithelial cells (MCF-10A), but the information about its effect on DNA methylation is sparse. The aim of this study was to characterize the whole-genome methylome effects of PFOS in our breast cell model and compare the findings with previously demonstrated DNA methylation alterations in breast tumor tissues. The DNA methylation profile was assessed at single CpG resolution in MCF-10A cells treated with 1 µM PFOS for 72 h by using Enzymatic Methyl sequencing (EM-seq). We found 12,591 differentially methylated CpG-sites and 13,360 differentially methylated 100 bp tiles in the PFOS exposed breast cells. These differentially methylated regions (DMRs) overlapped with 2406 genes of which 494 were long non-coding RNA and 1841 protein coding genes. We identified 339 affected genes that have been shown to display altered DNA methylation in breast cancer tissue and several other genes related to cancer development. This includes hypermethylation of GACAT3, DELEC1, CASC2, LCIIAR, MUC16, SYNE1 and hypomethylation of TTN and KMT2C. DMRs were also found in estrogen receptor genes (ESR1, ESR2, ESRRG, ESRRB, GREB1) and estrogen responsive genes (GPER1, EEIG1, RERG). The gene ontology analysis revealed pathways related to cancer phenotypes such as cell adhesion and growth. These findings improve the understanding of PFOS's potential role in breast cancer and illustrate the value of whole-genome methylome analysis in uncovering mechanisms of chemical effects, identifying biomarker candidates, and strengthening epidemiological associations, potentially impacting risk assessment.


Subject(s)
Alkanesulfonic Acids , Breast Neoplasms , DNA Methylation , Fluorocarbons , Humans , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , DNA Methylation/drug effects , Breast Neoplasms/genetics , Female , Epigenome
18.
Biochim Biophys Acta ; 1823(10): 1708-19, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22743040

ABSTRACT

The involvement of calcium-mediated signaling pathways in the mechanism of action of 1α,25-dihydroxyvitamin D(3) (1,25D) is currently demonstrated. In this study we found that 1,25D induces nongenomic effects mediated by membrane vitamin D receptor (VDRm) by modulating intermediate filament (IF) phosphorylation and calcium uptake through L-type voltage-dependent calcium channels (L-VDCC) in cerebral cortex of 10 day-old rats. Results showed that the mechanism of action of 1,25D involves intra- and extracellular calcium levels, as well as the modulation of chloride and potassium channels. The effects of L-VDCCs on membrane voltage occur over a broad potential range and could involve depolarizing or hyperpolarizing coupling modes, supporting a cross-talk among Ca(2+) uptake and potassium and chloride channels. Also, the Na(+)/K(+)-ATPase inactivation by ouabain mimicked the 1,25D action on (45)Ca(2+) uptake. The Na(+)/K(+)-ATPase inhibition observed herein might lead to intracellular Na(+) accumulation with subsequent L-VDCC opening and consequently increased (45)Ca(2+) (calcium, isotope of mass 45) uptake. Moreover, the 1,25D effect is dependent on the activation of the following protein kinases: cAMP-dependent protein kinase (PKA), Ca(2+)/calmodulin-dependent protein kinase (PKCaMII), phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase p38 (p38(MAPK)). The modulation of calcium entry into neural cells by the 1,25D we are highlighting, might take a role in the regulation of a plethora of intracellular processes. Considering that vitamin D deficiency can lead to brain illness, 1,25D may be a possible candidate to be used, at least as an adjuvant, in the pharmacological therapy of neuropathological conditions.


Subject(s)
Aging/metabolism , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Cerebral Cortex/metabolism , Intermediate Filaments/metabolism , Ion Channel Gating/drug effects , Vitamin D/analogs & derivatives , Aging/drug effects , Animals , Antigens, Nuclear/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebral Cortex/drug effects , Chloride Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Intermediate Filament Proteins/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , MAP Kinase Signaling System/drug effects , Models, Biological , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Potassium Channels/metabolism , Protein Kinase C/metabolism , Rats , Receptors, Calcitriol/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Vitamin D/pharmacology
19.
Mol Genet Metab ; 109(2): 144-53, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23611578

ABSTRACT

3-Hydroxy-3-methylglutaryl-CoA lyase (HL) deficiency is a genetic disorder biochemically characterized by predominant accumulation of 3-hydroxy-3-methylglutaric (HMG) and 3-methylglutaric (MGA) acids in tissues and biological fluids of affected individuals. Clinically, the patients present neurological symptoms and basal ganglia injury, whose pathomechanisms are partially understood. In the present study, we investigated the ex vivo effects of intrastriatal administration of HMG and MGA on important parameters of oxidative stress in striatum of developing rats. Our results demonstrate that HMG and MGA induce lipid and protein oxidative damage. HMG and MGA also increased 2',7'-dichlorofluorescein oxidation, whereas only HMG elicited nitric oxide production, indicating a role for reactive oxygen (HMG and MGA) and nitrogen (HMG) species in these effects. Regarding the enzymatic antioxidant defenses, both organic acids decreased reduced glutathione concentrations and the activities of superoxide dismutase and glutathione reductase and increased glutathione peroxidase activity. HMG also provoked an increase of catalase activity and a diminution of glucose-6-phosphate dehydrogenase activity. We finally observed that antioxidants fully prevented or attenuated HMG-induced alterations of the oxidative stress parameters, further indicating the participation of reactive species in these effects. We also observed that MK-801, a non-competitive antagonist of the N-methyl-D-aspartate (NMDA) receptor, prevented some of these effects, indicating the involvement of the NMDA receptor in HMG effects. The present data provide solid evidence that oxidative stress is induced in vivo by HMG and MGA in rat striatum and it is presumed that this pathomechanism may explain, at least in part, the cerebral alterations observed in HL deficiency.


Subject(s)
Acetyl-CoA C-Acetyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Basal Ganglia/metabolism , Meglutol/analogs & derivatives , Meglutol/metabolism , Oxidative Stress , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Antioxidants/pharmacology , Basal Ganglia/growth & development , Basal Ganglia/pathology , Catalase/metabolism , Dizocilpine Maleate/pharmacology , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Male , Malondialdehyde/metabolism , Protein Carbonylation , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Superoxide Dismutase/metabolism , Vitamin E/pharmacology
20.
Metab Brain Dis ; 28(3): 429-38, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23378107

ABSTRACT

Carbonyl compounds such as methylglyoxal (MGO) seem to play an important role in complications resulting from diabetes mellitus, in aging and neurodegenerative disorders. In this study, we are showing, that MGO is able to suppress cell viability and induce apoptosis in the cerebral cortex and hippocampus of neonatal rats ex-vivo. These effects are partially related with ROS production, evaluated by DCFH-DA assay. Coincubation of MGO and reduced glutathione (GSH) or Trolox (vitamin E) totally prevented ROS production but only partially prevented the MGO-induced decreased cell viability in the two brain structures, as evaluated by the MTT assay. Otherwise, L-NAME, a nitric oxide (NO) inhibitor, partially prevented ROS production in the two structures but partially prevented cytotoxicity in the hippocampus. Pharmacological inhibition of Erk, has totally attenuated MGO-induced ROS production and cytotoxicity, suggesting that MEK/Erk pathway could be upstream of ROS generation and cell survival. Otherwise, p38MAPK and JNK failed to prevent ROS generation but induced decreased cell survival consistent with ROS-independent mechanisms. We can propose that Erk, p38MAPK and JNK are involved in the cytotoxicity induced by MGO through different signaling pathways. While Erk could be an upstream effector of ROS generation, p38MAPK and JNK seem to be associated with ROS-independent cytotoxicity in neonatal rat brain. The cytotoxic damage progressed to apoptotic cell death at MGO concentration higher than those described for adult brain, suggesting that the neonatal brain is resistant to MGO-induced cell death. The consequences of MGO-induced brain damage early in life, remains to be clarified. However, it is feasible that high MGO levels during cortical and hippocampal development could be, at least in part, responsible for the impairment of cognitive functions in adulthood.


Subject(s)
Brain/pathology , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress/drug effects , Pyruvaldehyde/toxicity , Animals , Animals, Newborn , Annexin A5/metabolism , Antioxidants/pharmacology , Blotting, Western , Brain/drug effects , Brain/enzymology , Cell Survival/drug effects , Coloring Agents , Fluorescent Dyes , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , MAP Kinase Kinase 4/metabolism , Nerve Tissue Proteins/metabolism , Pyruvaldehyde/antagonists & inhibitors , Pyruvaldehyde/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Tetrazolium Salts , Thiazoles , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL