Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 269(1): 51-60, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23500012

ABSTRACT

Perfluorinated compounds are ubiquitous chemicals of major concern for their potential adverse effects on the human population. We have used primary rat embryonic neural stem cells (NSCs) to study the effects of perfluorooctane sulfonate (PFOS) on the process of NSC spontaneous differentiation. Upon removal of basic fibroblast growth factor, NSCs were exposed to nanomolar concentrations of PFOS for 48 h, and then allowed to differentiate for additional 5 days. Exposure to 25 or 50 nM concentration resulted in a lower number of proliferating cells and a higher number of neurite-bearing TuJ1-positive cells, indicating an increase in neuronal differentiation. Exposure to 50 nM also significantly increased the number of CNPase-positive cells, pointing to facilitation of oligodendrocytic differentiation. PPAR genes have been shown to be involved in PFOS toxicity. By q-PCR we detected an upregulation of PPARγ with no changes in PPARα or PPARδ genes. One of the downstream targets of PPARs, the mitochondrial uncoupling protein 2 (UCP2) was also upregulated. The number of TuJ1- and CNPase-positive cells increased after exposure to PPARγ agonist rosiglitazone (RGZ, 3 µM) and decreased after pre-incubation with the PPARγ antagonist GW9662 (5 µM). RGZ also upregulated the expression of PPARγ and UCP2 genes. Meanwhile GW9662 abolished the UCP2 upregulation and decreased Ca²âº activity induced by PFOS. Interestingly, a significantly higher expression of PPARγ and UCP3 genes was also detected in mouse neonatal brain after prenatal exposure to PFOS. These data suggest that PPARγ plays a role in the alteration of spontaneous differentiation of NSCs induced by nanomolar concentrations of PFOS.


Subject(s)
Alkanesulfonic Acids/toxicity , Brain/drug effects , Embryonic Stem Cells/drug effects , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Oligodendroglia/drug effects , PPAR gamma/drug effects , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Gene Expression Regulation/drug effects , Ion Channels/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurons/metabolism , Oligodendroglia/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Time Factors , Tubulin/metabolism , Uncoupling Protein 2 , Uncoupling Protein 3
2.
Neuropeptides ; 46(5): 195-201, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22889491

ABSTRACT

Galanin, a 29-aminoacid peptide (30 in humans), is widely distributed in the nervous and endocrine systems and exerts its actions via three G-protein-coupled receptors, GalR1-3. The galanin system has, among others, been associated with tumorigenesis. Our objective was to assess the expression of galanin and its receptors in pituitary tumors. Transcript levels of galanin and galanin receptors 1-3 (GalR1-3) were measured using quantitative real-time PCR (q-PCR) in pituitary tumors, surgically removed from thirteen patients, and twelve post mortem pituitaries. Galanin, GalR1 and GalR2 mRNA, but not GalR3 mRNA, were found in the twelve human post-mortem pituitaries. Expression of GalR1 was relatively increased in most, whereas GalR2 was decreased in some tumors. High levels of GalR3 were only found in tumors of five patients, who all relapsed shortly after surgical intervention. The results suggest that GalR3, a receptor for the neuroendocrine peptide galanin, is a potential marker for relapsing pituitary tumors. Thus, galanin receptors may play an important role in pituitary tumors, also for surgical outcome and prognosis, and may serve as a diagnostic tool. The association of GalR3 with tumor relapse suggests that antagonists to this receptor represent a potential therapeutic approach to treatment of pituitary tumors.


Subject(s)
Adenoma/metabolism , Galanin/metabolism , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , Receptors, Galanin/metabolism , Adenoma/genetics , Adult , Aged , Female , Galanin/genetics , Humans , Male , Middle Aged , Pituitary Neoplasms/genetics , Receptors, Galanin/genetics
3.
Neurotox Res ; 22(2): 127-37, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22237944

ABSTRACT

Glucocorticoids (GCs) are essential for normal brain development; however, there is consistent evidence that prenatal exposure of the fetal brain to excess GCs permanently modifies the phenotype of neuronal cells. In this paper, the murine-derived multipotent stem cell line C17.2 was used, as an in vitro model, to investigate the impact of GCs on neural stem cell survival. Our results indicate that dexamethasone (Dex) increases the sensitivity of murine neural stem cells (NSCs) to 2,3-methoxy-1,4-naphthoquinone-induced apoptosis, and this effect could be blocked by the glucocorticoid-receptor (GR) antagonist mifepristone, strongly suggesting the involvement of the GR. Furthermore, our results show that Dex decreases cell number and induces a G1-arrest. We hypothesized that the mitochondria are the main target of Dex. Interestingly, after treatment with Dex, 72% of the investigated genes involved in the mitochondrial respiratory chain are down-regulated, as well as 29% of the genes encoding for antioxidant enzymes. In conclusion, using the C17.2 cell line as a model to study developmental neurotoxicity in vitro, we have shown that GCs can increase cellular sensitivity to oxidative stress and alter the phenotype of NCSs.


Subject(s)
Anti-Inflammatory Agents/toxicity , Cell Death/drug effects , Dexamethasone/toxicity , Neural Stem Cells/drug effects , Oxidative Stress/drug effects , Animals , Anti-Inflammatory Agents/antagonists & inhibitors , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Dexamethasone/antagonists & inhibitors , Electron Transport/drug effects , Flow Cytometry , G1 Phase/drug effects , Gene Expression/drug effects , Mice , Mifepristone/pharmacology , Naphthoquinones/toxicity , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/drug effects , Trypan Blue
4.
Toxicol Sci ; 124(1): 192-201, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21908764

ABSTRACT

Developmental exposure to food contaminants, such as polychlorinated biphenyls (PCBs), has been considered as a possible cause of neurodevelopmental disorders. We have investigated the effects of noncytotoxic concentrations of PCBs 153 and 180 on spontaneous differentiation of rat embryonic neural stem cells (NSCs). Upon removal of basic fibroblast growth factor to induce spontaneous differentiation, cells were exposed to 100 nM of the selected PCBs for 48 h and analyzed after 5 days. Both PCBs 153 and 180 induced a significant increase in the number of neurite-bearing Tuj1-positive cells with a concomitant decrease in proliferating cells, as detected by FUCCI transfection and EdU staining. Measurements of spontaneous Ca²âº oscillations showed a decreased number of cells with Ca²âº activity after PCB exposure, further confirming the increase in neuronal cells. Conversely, exposure to methylmercury (MeHg), which we evaluated in parallel, led to an increased number of cells with Ca²âº activity, in agreement with the previously observed inhibition of neuronal differentiation. Analysis with quantitative PCR of the Notch pathway revealed that PCBs have a repressive action on Notch signaling, whereas MeHg activates it. Altogether, the data indicate that nanomolar concentrations of the selected non-dioxin-like PCBs and MeHg interfere in opposite directions with neuronal spontaneous differentiation of NSCs through Notch signaling. Combined exposures to PCBs and MeHg resulted in an induction of apoptosis and an antagonistic interaction on spontaneous neuronal differentiation. NSCs are further proven to be a valuable in vitro model to identify potential developmental neurotoxicants.


Subject(s)
Cell Differentiation/drug effects , Cerebral Cortex/embryology , Neural Stem Cells/drug effects , Neurons/drug effects , Polychlorinated Biphenyls/toxicity , Animals , Apoptosis/drug effects , Cell Culture Techniques , Cell Cycle/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Drug Synergism , Female , Food Contamination , Gestational Age , Immunoblotting , Immunohistochemistry , Methylmercury Compounds/toxicity , Neural Stem Cells/pathology , Neurons/pathology , Rats , Rats, Sprague-Dawley , Receptors, Notch/metabolism , Signal Transduction/drug effects
5.
Methods Mol Biol ; 758: 67-80, 2011.
Article in English | MEDLINE | ID: mdl-21815059

ABSTRACT

The developing nervous system is particularly susceptible to toxicants, and exposure during development may result in long-lasting neurological impairments. The damage can range from subtle to severe, and it may impose substantial burdens on affected individuals, their families, and society. Given the little information available on developmental neurotoxicity (DNT) and the growing number of chemicals that need to be tested, new testing strategies and approaches are necessary to identify developmental neurotoxic agents with speed, reliability, and respect for animal welfare. So far, there are no validated alternative methods for DNT testing. Recently, neural stem/progenitor cells have been proposed as relevant models for alternative DNT testing. In this chapter, we provide detailed protocols for culturing neural stem cells (NSCs), in vitro experimental models, including primary cultures of rat and human embryonic NSCs, rat and mouse adult NSCs, as well as the mouse NSC line C17.2 that we have implemented and successfully used for neurotoxicity studies.


Subject(s)
Cell Culture Techniques , Nervous System/drug effects , Neural Stem Cells/cytology , Toxicity Tests/methods , Animals , Cell Differentiation , Cell Proliferation , Environmental Pollutants/toxicity , Humans , Ki-67 Antigen/metabolism , Mice , Nervous System/growth & development , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Rats
6.
Neurotox Res ; 19(1): 183-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20169435

ABSTRACT

Methylmercury (MeHg) and polychlorinated biphenyls (PCBs) are widespread environmental pollutants commonly found as contaminants in the same food sources. Even though their neurotoxic effects are established, the mechanisms of action are not fully understood. In the present study, we have used the mouse hippocampal neuronal cell line HT22 to investigate the mechanisms of neuronal death induced by MeHg, PCB 153, and PCB 126, alone or in combination. All chemicals induced cell death with morphological changes compatible with either apoptosis or necrosis. Mitochondrial functions were impaired as shown by the significant decrease in mitochondrial Ca²+ uptake capacity and ATP levels. MeHg, but not the PCBs, induced loss of mitochondrial membrane potential and release of cytochrome c into the cytosol. Also, pre-treatment with the antioxidant MnTBAP was protective only against cell death induced by MeHg. While caspase activation was absent, the Ca²+-dependent proteases calpains were activated after exposure to MeHg or the selected PCBs. Furthermore, lysosomal disruption was observed in the exposed cells. Accordingly, pre-treatment with the calpain specific inhibitor PD150606 and/or the cathepsin D inhibitor Pepstatin protected against the cytotoxicity of MeHg and PCBs, and the protection was significantly enhanced when the two inhibitors were combined. Simultaneous exposures to lower doses of MeHg and PCBs suggested mostly antagonistic interactions. Taken together, these data indicate that MeHg and PCBs induce caspase-independent cell death via parallel activation of calpains and lysosomal proteases, and that in this model oxidative stress does not play a major role in PCB toxicity.


Subject(s)
Calpain/metabolism , Lysosomes/enzymology , Methylmercury Compounds/toxicity , Neurons/enzymology , Peptide Hydrolases/metabolism , Polychlorinated Biphenyls/toxicity , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line , Environmental Pollutants/toxicity , Enzyme Activation/drug effects , Enzyme Activation/physiology , Lysosomes/drug effects , Lysosomes/pathology , Mice , Neurons/drug effects , Neurons/pathology
7.
Cell Cycle ; 7(20): 3225-34, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18927501

ABSTRACT

One of the earliest morphological changes occurring in apoptosis is cell shrinkage associated with an increased efflux of K(+) and Cl(-) ions. Block of K(+) or Cl(-) channels prevents cell shrinkage and death. Recently, we found evidences for the activation of a voltage-dependent anion channel in the plasma membrane (pl-VDAC) of a hippocampal cell line undergoing apoptosis. Nothing is known on pl-VDAC in apoptotic cell death of neural cells at different stages of differentiation. We have addressed this issue in primary cultures of differentiated hippocampal neurons and embryonic neural stem cells (NSCs). In control hippocampal neurons, pl-VDAC is closed but acts as an NADH-ferricyanide reductase, while in apoptotic neurons, pl-VDAC is opened and the enzymatic activity is increased. Anti-VDAC antibodies block pl-VDAC and prevent apoptosis, as well as the increase in enzymatic activity. Conversely, in NSCs, pl-VDAC is scarcely seen and there is no NADH-ferricyanide reductase activity. In agreement, anti-VDAC antibodies do not affect the apoptotic process. Instead, we find activation of a Na(+) channel that has low voltage dependency, a conductance of 26 pS, and is blocked by amiloride, which also prevents apoptosis. Thus, it appears that activation of pl-VDAC during apoptosis is a critical event in differentiated neurons, but not in NSCs.


Subject(s)
Apoptosis/physiology , Cell Membrane/metabolism , Hippocampus/cytology , Neurons/physiology , Stem Cells/physiology , Voltage-Dependent Anion Channels/metabolism , Amiloride/pharmacology , Animals , Cell Differentiation/physiology , Cells, Cultured , Cytochromes c/metabolism , Enzyme Inhibitors/pharmacology , Female , Hippocampus/metabolism , NADH, NADPH Oxidoreductases/metabolism , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Pregnancy , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Staurosporine/pharmacology , Stem Cells/cytology , Stem Cells/drug effects
8.
Toxicol Lett ; 167(3): 183-90, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17049763

ABSTRACT

Polychlorinated biphenyls (PCBs) are persistent food contaminants that can have adverse effects on the endocrine and nervous systems, including the pituitary. In the present study, we have investigated cell death in the AtT20 pituitary cell line after exposure to coplanar PCB 126 and non-coplanar PCB 153. In addition, co-exposure to the PCBs and another neurotoxic food contaminant, methylmercury (MeHg), was studied to test possible interactive effects. Our results show that mainly necrosis is induced after exposure to the selected toxicants. Simultaneous exposure to moderately toxic doses of PCBs and MeHg resulted in additive or slightly synergistic effects on the induction of cell death. Furthermore, our data suggest that both PCB congeners trigger cell death in AtT20 cells via activation of calcium regulated calpains and lysosomal cathepsins, possibly through disruption of mitochondrial function and intracellular calcium signaling. However, caspase-activity appears not to be critical for PCB induced cell death in these cells. Presence of reactive oxygen species (ROS) and protective effects of pre-treatment with antioxidants were only found after MeHg exposure, suggesting that oxidative stress plays a major role in MeHg but not PCB toxicity in this experimental model.


Subject(s)
Apoptosis/drug effects , Methylmercury Compounds/toxicity , Pituitary Gland/drug effects , Polychlorinated Biphenyls/toxicity , Animals , Calcium Signaling/drug effects , Caspases/physiology , Cell Line , Mice , Mitochondria/drug effects , Mitochondria/physiology , Pituitary Gland/pathology , Reactive Oxygen Species/metabolism
9.
Pediatr Nephrol ; 21(3): 382-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16388391

ABSTRACT

Urinary tract infections (UTIs) are often caused by Escherichia coli (E. coli). Previous studies have demonstrated that up-regulation of heme oxygenase-1 (HO-1) may trigger a survival mechanism against renal cell death induced by E. coli toxins. The present study analyses the role of carbon monoxide (CO), an end product of HO-1, in the survival mechanism. Moreover, we identified hemolysin as a putative pro-apoptotic toxin in the E. coli supernatant. Tubular cells were incubated with CO in the presence or absence of E. coli toxins. Uropathogenic or transformants of non-pathogenic strains expressing hemolysin were used. We found that the survival pathway during E. coli infection might be activated by HO-1-derived production of CO. The protection by CO was also associated with up-regulation of p21 protein expression. Furthermore, we found that in children with pyelonephritis, all the E. coli strains expressing hemolysin induced apoptosis. In E. coli strains not expressing hemolysin, only 45% of the strains could induce apoptosis. In conclusion, generation of CO elicited by HO-1 could promote survival signaling in renal cells. Hemolysin is one of the secreted toxins that are involved in inducing apoptosis during UTI.


Subject(s)
Apoptosis/drug effects , Bacterial Toxins/metabolism , Carbon Monoxide/pharmacology , Escherichia coli Infections/microbiology , Escherichia coli/metabolism , Kidney Tubules, Proximal/pathology , Pyelonephritis/microbiology , Animals , Child , Heme Oxygenase-1/metabolism , Hemolysin Proteins/pharmacology , Humans , Kidney Tubules, Proximal/microbiology , LLC-PK1 Cells , Pyelonephritis/pathology , Rats , Swine , Up-Regulation
10.
Toxicology ; 207(3): 391-9, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15664267

ABSTRACT

The environmental man-made pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has profound and deleterious effects on the endocrine system, and the pituitary gland is among TCDD endocrine target organs. In the present study, we have investigated the effects of TCDD (1 pM, 0.1 nM and 1 nM) on the AtT-20 pituitary cell line. TCDD induces cell death, with morphological and biochemical changes indicating the occurrence of both apoptotic and necrotic cell death. Exposed cells exhibited apoptotic features including DNA condensation, activation of caspase-3 and exposure of phosphatidylserine (PS) on the outer plasma membrane. Concomitantly, cells with necrotic morphology such as cell swelling and plasma membrane damage were also present. The relative level of Fas ligand mRNA was increased after TCDD exposure, as well as Fas and Fas ligand protein levels detected by Western blotting and immunocytochemistry. Taken together, the results suggest that TCDD induces both necrosis and apoptosis in the pituitary AtT-20 cells and that the Fas/FasL system plays a critical role in inducing necrotic cell death rather than apoptosis (supported by the Swedish Research Council).


Subject(s)
Apoptosis/drug effects , Environmental Pollutants/toxicity , Pituitary Gland/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Nucleus/drug effects , Cell Nucleus/pathology , Dose-Response Relationship, Drug , Fas Ligand Protein , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Necrosis , Pituitary Gland/metabolism , Pituitary Gland/pathology , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , fas Receptor
11.
Brain Res ; 933(1): 12-22, 2002 Apr 12.
Article in English | MEDLINE | ID: mdl-11929631

ABSTRACT

Neurobehavioral changes have been described in workers occupationally exposed to styrene vapors. Alterations of neurotransmitters and loss of neurons have been observed in brains of styrene-exposed rats. However, the mechanisms of neuronal damage are not yet clearly understood. We have characterized the cellular alterations induced by the main reactive intermediate of styrene metabolism, styrene 7,8-oxide (SO) in the human neuroblastoma SK-N-MC cell line and primary culture of rat cerebellar granule cells (CGC). SK-N-MC cells exposed to SO (0.3-1 mM) displayed apoptotic morphology, together with chromatin condensation and DNA cleavage into high molecular weight fragments of regular size. These features were accompanied by the activation of class II caspases, as detected with the DEVD assay, by following the cleavage of the caspase-substrate poly (ADP-ribose) polymerase (PARP) and by detection of the active fragment of caspase-3. Pre-incubation of the cells with the caspase inhibitor z-VAD-fmk reduced the cellular damage induced by SO, suggesting that caspases play an important role in SO toxicity. Increased proteolysis by class II caspases was detected also in primary culture of CGC exposed to SO. In addition, the presence of the 150-kDa cleavage product of alpha-fodrin suggests a possible activation of calpains in SK-N-MC cells. Moreover, SO did not affect the level of expression of the p53 protein, even though it is known to cause DNA damage. The identified intracellular pathways affected by SO exposure provides end-points that can be used in future studies for the evaluation of the neurotoxic effect of styrene in vivo.


Subject(s)
Caspases/metabolism , DNA Fragmentation , Epoxy Compounds/pharmacology , Neurons/drug effects , Neurons/physiology , Carrier Proteins/chemistry , Cell Membrane Permeability , Cells, Cultured , Coloring Agents , Culture Media, Conditioned/chemistry , Enzyme Activation , Epoxy Compounds/analysis , Humans , Immunoblotting , Microfilament Proteins/chemistry , Neurons/cytology , Osmolar Concentration , Staining and Labeling , Trypan Blue , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...