Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Neurodegener Dis ; 11(1): 49-58, 2013.
Article in English | MEDLINE | ID: mdl-22627004

ABSTRACT

BACKGROUND: Inhibition of mitochondrial function occurs in many neurodegenerative diseases, and inhibitors of mitochondrial complexes I and II are used to model them. The complex II inhibitor, 3-nitroproprionic acid (3-NPA), kills the striatal neurons susceptible in Huntington's disease. The complex I inhibitor N-methyl-4-phenylpyridium (MPP(+)) and 6-hydroxydopamine (6-OHDA) are used to model Parkinson's disease. Zinc (Zn(2+)) accumulates after 3-NPA, 6-OHDA and MPP(+) in situ or in vivo. OBJECTIVE: We will investigate the role of Zn(2+) neurotoxicity in 3-NPA, 6-OHDA and MPP(+). METHODS: Murine striatal/midbrain tyrosine hydroxylase positive, or near-pure cortical neuronal cultures, or animals were exposed to 3-NPA or MPP(+) and 6-OHDA with or without neuroprotective compounds. Intracellular zinc ([Zn(2+)](i)), nicotinamide adenine dinucleotide (NAD(+)), NADH, glycolytic intermediates and neurotoxicity were measured. RESULTS: We showed that compounds or genetics which restore NAD(+) and attenuate Zn(2+) neurotoxicity (pyruvate, nicotinamide, NAD(+), increased NAD(+) synthesis, sirtuin inhibition or Zn(2+) chelation) attenuated the neuronal death induced by these toxins. The increase in [Zn(2+)](i) preceded a reduction in the NAD(+)/NADH ratio that caused a reversible glycolytic inhibition. Pyruvate, nicotinamide and NAD(+) reversed the reductions in the NAD(+)/NADH ratio, glycolysis and neuronal death after challenge with 3-NPA, 6-OHDA or MPP(+), as was previously shown for exogenous Zn(2+). To test efficacy in vivo, we injected 3-NPA into the striatum of rats and systemically into mice, with or without pyruvate. We observed early striatal Zn(2+) fluorescence, and pyruvate significantly attenuated the 3-NPA-induced lesion and restored behavioral scores. CONCLUSIONS: Together, these studies suggest that Zn(2+) accumulation caused by MPP(+) and 3-NPA is a novel preventable mechanism of the resultant neurotoxicity.


Subject(s)
Huntington Disease/drug therapy , Huntington Disease/metabolism , Parkinson Disease/metabolism , Zinc/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Carrier Proteins , Cation Transport Proteins , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dihydroxyacetone Phosphate/metabolism , Disease Models, Animal , Drug Interactions , Embryo, Mammalian , Fructose-Bisphosphatase/metabolism , Humans , Huntington Disease/chemically induced , Huntington Disease/pathology , Male , Matrix Metalloproteinase 16/deficiency , Membrane Proteins/deficiency , Membrane Transport Proteins , Mental Disorders/chemically induced , Mental Disorders/prevention & control , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mice, Transgenic , NAD/metabolism , NAD/therapeutic use , Neurons/drug effects , Niacinamide/therapeutic use , Nitro Compounds/toxicity , Oxidopamine/toxicity , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/pathology , Propionates/toxicity , Pyruvic Acid/therapeutic use , Rats , Rats, Long-Evans , Tyrosine 3-Monooxygenase/metabolism
2.
J Nutr ; 142(12): 2119-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23096014

ABSTRACT

Pancreatic zinc (Zn(2+)) concentrations are linked to diabetes and pancreatic dysfunction, but Zn(2+) is also required for insulin processing and packaging. Zn(2+) released with insulin increases ß-cell pancreatic death after streptozotocin toxin exposure in vitro and in vivo. Triosephosphate accumulation, caused by NAD(+) loss and glycolytic enzyme dysfunction, occur in type-1 diabetics (T1DM) and animal models. We previously showed these mechanisms are also involved in Zn(2+) neurotoxicity and are attenuated by nicotinamide- or pyruvate-induced restoration of NAD(+) concentrations, Zn(2+) restriction, or inhibition of Sir2 proteins. We tested the hypothesis that similar Zn(2+)- and NAD(+)-mediated mechanisms are involved in ß-cell toxicity in models of ongoing T1DM using mouse insulinoma cells, islets, and nonobese diabetic (NOD) mice. Zn(2+), streptozotocin, and cytokines caused NAD(+) loss and death in insulinoma cells and islets, which were attenuated by Zn(2+) restriction, pyruvate, nicotinamide, NAD(+), and inhibitors of Sir2 proteins. We measured diabetes incidence and mortality in NOD mice and demonstrated that pyruvate supplementation, or genetic or dietary Zn(2+) reduction, attenuated these measures. T-lymphocyte infiltration, punctate Zn(2+) staining, and ß-cell loss increased with time in islets of NOD mice. Dietary Zn(2+) restriction or Zn(2+) transporter 5 knockout reduced pancreatic Zn(2+) staining and increased ß-cell mass, glucose homeostasis, and survival in NOD mice, whereas Zn(2+) supplementation had the opposite effects. Pancreatic Zn(2+) reduction or NAD(+) restoration (pyruvate or nicotinamide supplementation) are suggested as novel targets for attenuating T1DM.


Subject(s)
Carrier Proteins/physiology , Insulinoma/pathology , Islets of Langerhans/pathology , Pancreatic Neoplasms/pathology , Pyruvic Acid/administration & dosage , Zinc/toxicity , Animals , Benzamides/pharmacology , Calcium Channel Blockers/pharmacology , Cell Line, Tumor , Diabetes Mellitus, Experimental/prevention & control , Dietary Supplements , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , NAD/metabolism , Naphthols/pharmacology , Reactive Oxygen Species/metabolism , Streptozocin/toxicity , Zinc/administration & dosage
3.
Eur J Neurosci ; 32(6): 894-904, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20722716

ABSTRACT

Trophic deprivation-mediated neuronal death is important during development, after acute brain or nerve trauma, and in neurodegeneration. Serum deprivation (SD) approximates trophic deprivation in vitro, and an in vivo model is provided by neuronal death in the mouse dorsal lateral geniculate nucleus (LGNd) after ablation of the visual cortex (VCA). Oxidant-induced intracellular Zn(2+) release ([Zn(2+) ](i) ) from metallothionein-3 (MT-III), mitochondria or 'protein Zn(2+) ', was implicated in trophic deprivation neurotoxicity. We have previously shown that neurotoxicity of extracellular Zn(2+) required entry, increased [Zn(2+) ](i) , and reduction of NAD(+) and ATP levels causing inhibition of glycolysis and cellular metabolism. Exogenous NAD(+) and sirtuin inhibition attenuated Zn(2+) neurotoxicity. Here we show that: (1) Zn(2+) is released intracellularly after oxidant and SD injuries, and that sensitivity to these injuries is proportional to neuronal Zn(2+) content; (2) NAD(+) loss is involved - restoration of NAD(+) using exogenous NAD(+) , pyruvate or nicotinamide attenuated these injuries, and potentiation of NAD(+) loss potentiated injury; (3) neurons from genetically modified mouse strains which reduce intracellular Zn(2+) content (MT-III knockout), reduce NAD(+) catabolism (PARP-1 knockout) or increase expression of an NAD(+) synthetic enzyme (Wld(s) ) each had attenuated SD and oxidant neurotoxicities; (4) sirtuin inhibitors attenuated and sirtuin activators potentiated these neurotoxicities; (5) visual cortex ablation (VCA) induces Zn(2+) staining and death only in ipsilateral LGNd neurons, and a 1 mg/kg Zn(2+) diet attenuated injury; and finally (6) NAD(+) synthesis and levels are involved given that LGNd neuronal death after VCA was dramatically reduced in Wld(s) animals, and by intraperitoneal pyruvate or nicotinamide. Zn(2+) toxicity is involved in serum and trophic deprivation-induced neuronal death.


Subject(s)
NAD/deficiency , Neurons/metabolism , Oxidative Stress/physiology , Serum Albumin/deficiency , Zinc/metabolism , Animals , Cations, Divalent/metabolism , Cell Death/physiology , Cells, Cultured , Geniculate Bodies/metabolism , Geniculate Bodies/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Sirtuin 1/physiology
4.
Eur J Neurosci ; 24(8): 2169-76, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17042794

ABSTRACT

Zinc neurotoxicity has been demonstrated in ischemic, seizure, hypoglycemic, and trauma-induced neuronal death where Zn(2+) is thought to be synaptically released and taken up in neighbouring neurons, reaching toxic concentrations. We previously demonstrated that toxicity of extracellular Zn(2+) depended on entry, elevation in intracellular free Zn(2+) ([Zn(2+)](i)), a reduction in NAD(+) and ATP levels, and dysfunction of glycolysis and cellular metabolism. We suggested that PARP-1 activation alone can not explain this loss of neuronal NAD(+). NAD(+) was recently demonstrated to permeate neurons and glia, and we have now shown that exogenous NAD(+) can reduce Zn(2+) neurotoxicity, and 3-acetylpyridine, which generates inactive NAD(+), potentiated Zn(2+) neurotoxicity. Sirtinol and 2-hydroxynaphthaldehyde, inhibitors of the sirtuin pathway (SIRT proteins are NAD(+)-catabolic protein deacetylases), attenuated both acute and chronic Zn(2+) neurotoxicity. Resveratrol and fisetin (sirtuin activators) potentiated NAD(+) loss and Zn(2+) neurotoxicities. Furthermore, neuronal cultures derived from the Wld(s) mouse, which overexpress the NAD(+) synthetic enzyme nicotinamide mononucleotide adenyl transferase (NMNAT-1), had reduced sensitivity to Zn(2+) neurotoxicity. Finally, nicotinamide was demonstrated to attenuate CA1 neuronal death after 10 min of global ischemia in rat even if administered 1 h after the insult. Together with previous data, these results further implicate NAD(+) levels in Zn(2+) neurotoxicity.


Subject(s)
NAD/metabolism , Neurotoxicity Syndromes/metabolism , Sirtuins/metabolism , Zinc/toxicity , Aldehydes/pharmacology , Animals , Antioxidants/pharmacology , Brain Ischemia/pathology , Cells, Cultured , Flavonoids/pharmacology , Flavonols , Ion Channels/physiology , Male , Mitochondria/metabolism , Naphthalenes/pharmacology , Neural Conduction/physiology , Neurotoxicity Syndromes/pathology , Neurotoxins/toxicity , Niacinamide/pharmacology , Pyridines/toxicity , Rats , Rats, Long-Evans , Resveratrol , Signal Transduction/drug effects , Sirtuins/antagonists & inhibitors , Stilbenes/pharmacology , Transcriptional Activation/physiology
5.
J Neurochem ; 91(4): 996-1006, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15525353

ABSTRACT

We report here that exposure to low concentrations of proteasome inhibitors (e.g. 10-100 nm MG-132, 0.1-3 nm epoxomicin or 10-30 nm clasto-lactacystin beta-lactone) resulted in an enhancement, rather than an inhibition, of proteasome activity in cultured neocortical neurons. Size-fractionation chromatography confirmed that the enhanced peptide cleavage activity was associated with proteasome-sized complexes. This sub toxic exposure reduced neuronal death caused by subsequent exposure to oxidative stress (100-200 microm H(2)O(2) for 30 min, 24-h exposure to 100 microm paraquat or 7.5 microm menadione), but did not alter vulnerability to excitotoxicity (5-min exposure to 30-100 microm NMDA or 24 exposure to 12 microm NMDA). Sub toxic proteasome inhibitor exposure caused an increase in levels of proteasome core subunit proteins and mRNAs, but not in levels of potentially cytoprotective heat shock proteins (hsp70, hsp90 and hsp40). The neuroprotective effects of proteasome inhibitor pre-treatment were blocked by coapplication of proteasome inhibitors during the oxidative insult. These findings support a model in which sublethal proteasome inhibition induces neurons to increase proteasome activity and promotes resistance to oxidative injury and suggests that enhancement of proteasome activity is a potential therapeutic target for diseases in which oxidative stress has been implicated.


Subject(s)
Enzyme Inhibitors/pharmacology , Neurons/drug effects , Neurons/enzymology , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Cytoprotection/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Mice , Neuroprotective Agents/pharmacology , Oxidants/toxicity , Oxidative Stress/physiology , Proteasome Endopeptidase Complex/genetics , RNA, Messenger/metabolism
6.
Eur J Neurosci ; 18(6): 1402-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14511320

ABSTRACT

We have previously suggested that zinc-induced neuronal death may be mediated in part by inhibition of the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH), secondary to depletion of the essential cosubstrate NAD+. Given convergent evidence implicating the NAD+-catabolizing enzyme, poly ADP ribosyl polymerase (PARP) in mediating ATP depletion and neuronal death after excitotoxic and ischemic insults, we tested the specific hypothesis that the neuronal death induced by exposure to toxic levels of extracellular zinc might be partly mediated by PARP. PARP was activated in cultured mouse cortical astrocytes after a toxic acute Zn2+ exposure (350 microm Zn2+ for 15 min), but not in cortical neurons or glia after exposure to a toxic chronic Zn2+ exposure (40 microm Zn2+ for 1-4 h), an exposure sufficient to deplete NAD+ and ATP levels. Furthermore, the neurotoxicity induced by acute, but not chronic, Zn2+ exposure was reduced in mixed neuronal-glial cultures prepared from mutant mice lacking the PARP gene. These data suggest PARP activation may contribute to more fulminant forms of Zn2+-induced neuronal death.


Subject(s)
Ethidium/analogs & derivatives , Neurons/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Zinc/toxicity , Animals , Blotting, Western , Cell Death/drug effects , Cells, Cultured , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule/veterinary , Embryo, Mammalian , Ethidium/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Neocortex/cytology , Neocortex/drug effects , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/enzymology , Neurons/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Reactive Oxygen Species/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...