Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Toxicol Appl Pharmacol ; 256(3): 348-59, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21840330

ABSTRACT

Organophosphorus-ester induced delayed neurotoxicity (OPIDN) is a neurodegenerative disorder characterized by ataxia progressing to paralysis with a concomitant central and peripheral, distal axonapathy. Diisopropylphosphorofluoridate (DFP) produces OPIDN in the chicken that results in mild ataxia in 7-14 days and severe paralysis as the disease progresses with a single dose. White leghorn layer hens were treated with DFP (1.7 mg/kg, sc) after prophylactic treatment with atropine (1mg/kg, sc) in normal saline and eserine (1mg/kg, sc) in dimethyl sulfoxide. Control groups were treated with vehicle propylene glycol (0.1 ml/kg, sc), atropine in normal saline and eserine in dimethyl sulfoxide. The hens were euthanized at different time points such as 1, 2, 5, 10 and 20 days, and the tissues from cerebrum, midbrain, cerebellum, brainstem and spinal cord were quickly dissected and frozen for mRNA (northern) studies. Northern blots were probed with BCL2, GADD45, beta actin, and 28S RNA to investigate their expression pattern. Another set of hens was treated for a series of time points and perfused with phosphate buffered saline and fixative for histological studies. Various staining protocols such as Hematoxylin and Eosin (H&E); Sevier-Munger; Cresyl echt Violet for Nissl substance; and Gallocynin stain for Nissl granules were used to assess various patterns of cell death and degenerative changes. Complex cell death mechanisms may be involved in the neuronal and axonal degeneration. These data indicate altered and differential mRNA expressions of BCL2 (anti apoptotic gene) and GADD45 (DNA damage inducible gene) in various tissues. Increased cell death and other degenerative changes noted in the susceptible regions (spinal cord and cerebellum) than the resistant region (cerebrum), may indicate complex molecular pathways via altered BCL2 and GADD45 gene expression, causing the homeostatic imbalance between cell survival and cell death mechanisms. Semi quantitative analysis revealed that the order of severity of damage declines from the spino-cerebellar, ventral, and dorsal tract respectively, suggesting neuroanatomical specificity. Thus, early activation of cell death and cell survival processes may play significant role in the clinical progression and syndromic clinical feature presentation of OPIDN.


Subject(s)
Cell Death/drug effects , Cell Survival/drug effects , Neurotoxicity Syndromes/metabolism , Organophosphorus Compounds/toxicity , Animals , Apoptosis/drug effects , Atropine/pharmacology , Blotting, Northern , Brain/drug effects , Brain/pathology , Cell Death/physiology , Cell Survival/physiology , Chickens , Cholinesterase Inhibitors/toxicity , Female , Isoflurophate/toxicity , Neurotoxicity Syndromes/pathology , Physostigmine/pharmacology , Spinal Cord/drug effects , Spinal Cord/pathology , Time Factors
2.
Gene Ther ; 13(17): 1281-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16672983

ABSTRACT

The deficiency of glucose-6-phosphatase (G6Pase) underlies life-threatening hypoglycemia and growth retardation in glycogen storage disease type Ia (GSD-Ia). An adeno-associated virus (AAV) vector encoding G6Pase was pseudotyped as AAV8 and administered to 2-week-old GSD-Ia mice (n = 9). Median survival was prolonged to 7 months following vector administration, in contrast to untreated GSD-Ia mice that survived for only 2 weeks. Although GSD-Ia mice were initially growth-retarded, treated mice increased fourfold in weight to normal size. Blood glucose was partially corrected by 2 weeks following treatment, whereas blood cholesterol normalized. Glucose-6-phosphatase activity was partially corrected to 25% of the normal level at 7 months of age in treated mice, and blood glucose during fasting remained lower in treated, affected mice than in normal mice. Glycogen storage was partially corrected in the liver by 2 weeks following treatment, but reaccumulated to pre-treatment levels by 7 months old (m.o.). Vector genome DNA decreased between 3 days and 3 weeks in the liver following vector administration, mainly through the loss of single-stranded genomes; however, double-stranded vector genomes were more stable. Although CD8+ lymphocytic infiltrates were present in the liver, partial biochemical correction was sustained at 7 m.o. The development of efficacious AAV vector-mediated gene therapy could significantly reduce the impact of long-term complications in GSD-Ia, including hypoglycemia, hyperlipidemia and growth failure.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Glucose-6-Phosphatase/analysis , Glucose-6-Phosphatase/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glycogen/analysis , Glycogen Storage Disease Type I/enzymology , Glycogen Storage Disease Type I/immunology , Immunohistochemistry , Injections, Intravenous , Kidney/chemistry , Kidney/enzymology , Kidney/immunology , Liver/chemistry , Liver/enzymology , Liver/immunology , Mice , Mice, Knockout , Models, Animal , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transduction, Genetic/methods
3.
Neurochem Res ; 26(1): 43-50, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11358281

ABSTRACT

Diisopropyl phosphorofluoridate (DFP) produces organophosphorus-ester induced delayed neurotoxicity (OPIDN) in the hen, human and other sensitive species. We studied the effect of single dose of DFP (1.7 mg/kg/s.c.) on the expression of alpha tubulin which is one of the major sub-unit of tubulin polymers that constitute an important constituent of cellular architecture. The hens were sacrificed at different time points i.e. 1, 2, 5, 10, and 20 days. Total RNA was extracted from the following brain regions: cerebrum, cerebellum, and brainstem as well as spinal cord. Northern blots prepared using standard protocols were hybridized with alpha tubulin as well as with beta-actin and 28S RNA cDNA (controls) probes. The results indicate a differential/spatial/temporal regulation of alpha tubulin levels which may be the result of perturbed microtubule dynamics not only in the axons but also in perikarya of neurons in the CNS of DFP treated hens. In the highly susceptible tissues like brainstem and spinal cord the initial down-regulation of mRNA levels could be attributed to DFP induced stress response resulting in inhibited cell metabolism and or cell injury/cell death. Increase in levels of mRNA at 5 days and thereafter coincided with increased tubulin transport which may be due to increased phosphorylation of tubulins in both axons and perikarya and other intraaxonal changes resulting in impaired axonal transport. DFP induced decreased rate of tubulin polymerization resulting in increased levels of free tubulin monomers may be involved in the altered alpha tubulin mRNA expression at different time points by autoregulatory circuits. Cerebellum being the less susceptible tissue showed only a moderate decline at day 2, while the alpha tubulin remained at near control levels at day 1. Delayed down-regulation may be due to the co-ordinated up or down-regulation of different sub-types of alpha and beta tubulins as well as the differential response of specialised cell types in cerebellum. Continuous overexpression of alpha tubulin in cerebrum from the beginning may be its effective protective strategy to safeguard itself from neurotoxicity. Differential expression pattern observed could be due to the differential susceptibility and variability in the rate of axonal transport of different regions besides the tubulin heterogenity of CNS. Hence our results indicte differential expression of alpha tubulin is either one of the reasons for the development of OPIDN or the result of progressive changes taking place during OPIDN.


Subject(s)
Central Nervous System/drug effects , Central Nervous System/metabolism , Isoflurophate/toxicity , RNA, Messenger/metabolism , Tubulin/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain Stem/drug effects , Brain Stem/metabolism , Chickens , Female , RNA, Ribosomal, 28S/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Time Factors
4.
Toxicol Sci ; 60(2): 305-14, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11248143

ABSTRACT

Since their return from Persian Gulf War (PGW), many veterans have complained of symptoms including muscle and joint pain, ataxia, chronic fatigue, headache, and difficulty with concentration. The causes of the symptoms remain unknown. Because these veterans were exposed to a combination of chemicals including pyridostigmine bromide (PB), DEET, and permethrin, we investigated the effects of these agents, alone and in combination, on the sensorimotor behavior and central cholinergic system of rats. Male Sprague-Dawley rats (200-250 gm) were treated with DEET (40 mg/kg, dermal) or permethrin (0.13 mg/kg, dermal), alone and in combination with PB (1.3 mg/kg, oral, last 15 days only), for 45 days. Sensorimotor ability was assessed by a battery of behavioral tests that included beam-walk score, beam-walk time, incline plane performance, and forepaw grip on days 30 and 45 following the treatment. On day 45 the animals were sacrificed, and plasma and CNS cholinesterase, and brain choline acetyl transferase, muscarinic and nicotinic acetylcholine receptors were evaluated. Animals treated with PB, alone or in combination with DEET and permethrin, showed a significant deficit in beam-walk score as well as beam-walk time as compared with controls. Treatment with either DEET or permethrin, alone or in combination with each other, did not have a significant effect on beam-walk score. All chemicals, alone or in combination, resulted in a significant impairment in incline plane testing on days 30 and 45 following treatment. Treatment with PB, DEET, or permethrin alone did not have any inhibitory effect on plasma or brain cholinesterase activities, except that PB alone caused moderate inhibition in midbrain acetylcholinesterase (AChE) activity. Treatment with permethrin alone caused significant increase in cortical and cerebellar AChE activity. A combination of DEET and permethrin or PB and DEET led to significant decrease in AChE activity in brainstem and midbrain and brainstem, respectively. A significant decrease in brainstem AChE activity was observed following combined exposure to PB and permethrin. Coexposure with PB, DEET, and permethrin resulted in significant inhibition in AChE in brainstem and midbrain. No effect was observed on choline acetyl transferase activity in brainstem or cortex, except combined exposure to PB, DEET, and permethrin caused a slight but significant increase in cortical choline acetyltransferase activity. Treatment with PB, DEET, and permethrin alone caused a significant increase in ligand binding for m2 muscarinic acetylcholine receptor (mAChR) in the cortex. Coexposure to PB, DEET, and permethrin did not have any effect over that of PB-induced increase in ligand binding. There was no significant change in ligand binding for nicotinic acetylcholine receptor (nAChR) associated with treatment with the chemical alone; a combination of PB and DEET or coexposure with PB, DEET, and permethrin caused a significant increase in nAChR ligand binding in the cortex. Thus, these results suggest that exposure to physiologically relevant doses of PB, DEET, and permethrin, alone or in combination, leads to neurobehavioral deficits and region-specific alterations in AChE and acetylcholine receptors.


Subject(s)
DEET/toxicity , Psychomotor Performance/drug effects , Pyrethrins/toxicity , Pyridostigmine Bromide/toxicity , Acetylcholinesterase/metabolism , Administration, Cutaneous , Administration, Oral , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/enzymology , Butyrylcholinesterase/metabolism , Choline O-Acetyltransferase/metabolism , DEET/administration & dosage , Drug Interactions , Male , Permethrin , Psychomotor Performance/physiology , Pyrethrins/administration & dosage , Pyridostigmine Bromide/administration & dosage , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M2 , Receptors, Muscarinic/metabolism , Receptors, Nicotinic/metabolism
5.
Neurochem Res ; 25(6): 809-16, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10943999

ABSTRACT

Diisopropyl phophorofluoridate (DFP) produces organophosphorus-ester induced delayed neurotoxicity (OPIDN) in the hen, human and other sensitive species. We studied the effect of DFP admimistration (1.7 mg/kg/s.c.) on the expression of Intermediate Filament (IF) proteins: Glial Fibrillary Acidic Protein (GFAP) and vimentin which are known indicators of neurotoxicity and astroglial pathology. The hens were sacrificed at different time points i.e. 1,2,5,10 and 20 days. Total RNA was extracted from the following brain regions: cerebrum, cerebellum, and brainstem as well as spinal cord. Northern blots prepared using standard protocols were hybridized with GFAP and vimentin as well as beta-actin and 18S RNA cDNA (controls) probes. The results indicate a differential/spatial/temporal regulation of GFAP and vimentin levels which may be due to the result of disruption of glial-neuronal network. The GFAP transcript levels reached near control levels (88% and 95%) at 20 days post DFP treatment after an initial down-regulation (60% and 73%) in highly susceptible tissues like spinal cord and brainstem respectively. However vimentin transcript levels remained down-regulated (61% and 53%) at 20 days after an early reduced levels(47% and 55%) for spinal cord and brainstem respectively. This may be due to the astroglial pathology resulting in neuronal alterations or vice-versa. In cerebellum (less susceptile tissue) GFAP levels were moderately down-regulated at 1,2 and 5 days and reached near control values at 10 and 20 days. Vimentin was rapidly reinduced (128%) in cerebellum at 5 days and remained at the same level at 10 days and then returned to control values at 20 days after an initial down-regulation at 1 and 2 days. Thus these alterations were less drastic in cerebellum as indicated by initial susceptibility followed by rapid recovery. On the other hand both GFAP and vimentin levels were upregulated from 2 days onwards in the non-susceptible tissue cerebrum, implying protective mechanisms from the beginning. Hence the DFP induced astroglial pathology as indicated by the complex expression profile of GFAP and vimentin mRNA levels may be playing an important role in the delayed degeneration of axons or is the result of progressive degeneration of axons in OPIDN.


Subject(s)
Central Nervous System/drug effects , Glial Fibrillary Acidic Protein/genetics , Isoflurophate/toxicity , RNA, Messenger/genetics , Vimentin/genetics , Animals , Central Nervous System/metabolism , Chickens , Female , Gene Expression Regulation/drug effects
6.
Neurochem Res ; 25(3): 327-34, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10761975

ABSTRACT

A single dose of diisopropyl phosphorofluoridate (DFP), an organophosphorus ester, produces delayed neurotoxicity (OPIDN) in hen. DFP produces mild ataxia in hens in 7-14 days, which develops into severe ataxia or paralysis as the disease progresses. Since, OPIDN is associated with alteration in the expression of several proteins (e.g., Ca2+/calmodulin-dependent protein kinase II (CaM kinase II) alpha-subunit, tau, tubulin, neurofilament (NF) protein, vimentin, GFAP) as well as their mRNAs (e.g., NF, CaM kinase II alpha-subunit), we determined the effect of a single dose of DFP on the expression of one of the best known immediate-early gene (IEG), c-fos. C-fos expression was measured by Northern hybridization in cerebrum, cerebellum, brainstem, midbrain, spinal cord, and the sciatic nerves of hens at 0.5 hr, 1 hr, 2 hr, 1 day, 5 days, 10 days, and 20 days after a single 1.7 mg/kg, sc. injection of DFP. All the tissues (cerebrum, 52%; cerebellum, 55%; brainstem, 49%; midbrain, 23%; spinal cord, 80%; sciatic nerve, 157%) showed significant increase in c-fos expression in 30 min and this elevated level persisted at least up to 2 hr. Expressions of beta-actin mRNA and 18S RNA were used as internal controls. The significant increase in c-fos expression in DFP-treated hens suggests that c-fos may be one of the IEGs involved in the development of OPIDN.


Subject(s)
Central Nervous System/metabolism , Isoflurophate/toxicity , Peripheral Nervous System/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/biosynthesis , Animals , Blotting, Northern , Brain Stem/metabolism , Cerebellum/metabolism , Cerebral Cortex/metabolism , Chickens , In Vitro Techniques , Male , Mesencephalon/metabolism , Organ Specificity , Sciatic Nerve/metabolism , Spinal Cord/metabolism
7.
Neurochem Res ; 25(12): 1579-86, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11152387

ABSTRACT

Diisopropyl phosphorofluoridate (DFP) produces organophosphorus-ester induced delayed neurotoxicity (OPIDN) in the hen, human and other sensitive species. We studied the effect of a single dose of DFP (1.7 mg/kg/sc) on the expression of c-jun, which is one of the heterodimerizing ITFs (Inducible Transcriptional Factors) of the AP-1 family. The hens were sacrificed at different time points ie 0.25, .0.50, 1 and 2 hrs. Total RNA was extracted from the following brain regions: cerebrum, cerebellum, brainstem, midbrain and as well as spinal cord. Northern blots prepared using standard protocols were hybridized with c-jun as well as b-actin and 18S RNA cDNA (control) probes. The results indicate differential regulation of c-jun levels which may be due to the activation of both cholinergic and non-cholinergic pathways of CNS, besides changing roles of c-jun (as mediator of degeneration or regeneration) depending on heterodimerization with other ITFs. In the highly susceptible tissues like brainstem and spinal cord c-jun transcript levels increased at 15 minutes and continued to increase gradually till it reached the maximum at 2 hrs. Overall spinal cord showed the maximum levels of c-jun induction (207%) at 2 hrs time point of all the CNS tissues. The enhancement of cholinergic transmisson by the inhibition of cholinestrase may be responsible for the gradual increase mediated by neural and vascular factors. In contrast, less susceptible tissue, cerebellum showed almost immediate induction to high level of (179%) at 15 minutes and the levels stayed more or less the same until it peaked to 185% at 2 hrs. Relatively low abundance of cholinergic neurons and high number of sensitized specialized cell types like Bergman glia and Purkinje cells may be responsible for the immediate higher induction. Non-susceptible tissue cerebrum did not show any changes in the c-jun levels. In midbrain the induction pattern was very similar to that of brainstem. This differential induction pattern of c-jun encomposing the differences in the quantity and time course was directly proportionate to the degree of susceptibility and cellular heterogeneity of different regions of CNS. The significant increase in c-jun levels along with our earlier observation on the increased c-fos levels indicate that AP-1 family of genes may be one of the IEGs involved in the long term changes which eventually lead to OPIDN.


Subject(s)
Gene Expression Regulation/drug effects , Isoflurophate/pharmacology , Neurotoxins/pharmacology , Proto-Oncogene Proteins c-jun/genetics , Animals , Brain/metabolism , Brain Stem/metabolism , Cerebellum/metabolism , Chickens , Female , Mesencephalon/metabolism , RNA, Messenger/metabolism , Spinal Cord/metabolism
8.
Genomics ; 45(2): 279-89, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9344650

ABSTRACT

The sensing of extracellular calcium is a general paradigm for regulating diverse cellular functions in many tissues. A calcium-sensing receptor (Casr) belonging to the metabotropic glutamate family of G-protein-coupled receptors (GPCR) that transduces the effects of extracellular calcium in the parathyroid gland as well as other tissues has been identified. The diversity of GPCR families and the recent finding of calcium sensing in cells lacking the known Casr suggest the existence of additional receptors related to Casr. By polymerase chain reaction (PCR) amplification and screening of genomic libraries, we have identified multiple Casr-related sequences (Casr-rs) in the mouse. Using primers designed to regions of the first and third intracellular loops of Casr, we initially PCR amplified a 497-bp Casr-related sequence (Casr-rs1) with high homology to Casr. The deduced protein sequence of Casr-rs1 is 63% similar and 40% identical to Casr over the available transmembrane region. We screened a mouse genomic library with a Casr-rs1 probe and identified two additional Casr-related sequences (Casr-rs2 and Casr-rs3). In the predicted transmembrane domain, Casr-rs2 and Casr-rs3 are 95% identical to Casr-rs1. We mapped Casr-rs1 to mouse Chromosome (Chr) 7 by interspecific backcross analysis, whereas the known Casr localizes to mouse Chr 16. By fluorescence in situ hybridization, Casr-rs2 also localized to mouse Chr 7 and Casr-rs3 mapped to mouse Chr 4. We were able to distinquish Casr-rs1 from Casr-rs2 by PCR using specific primers, suggesting that they are distinct genes clustered on Chr 7. By RT-PCR, we identified additional Casr-rs transcripts in mouse kidney, brain, testis, embryo, and MC3T3-E1 osteoblasts, but not in lung or liver. The homologous sequence in mouse kidney, embryo, and MC3T3-E1 osteoblasts, designated Casr-rs4, has a deduced amino acid sequence that is 100% similar and 97% identical to that of Casr-rs1. The sequence amplified from mouse brain, Casr-rs5, has a deduced protein sequence that is 96% similar and 92% identical to that of Casr-rs1. Our findings establish the existence of a novel multimembered family of Casr-related sequences in the mouse which may encode receptors that transduce responses to diverse extracellular cations.


Subject(s)
Receptors, Cell Surface/genetics , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , Chromosome Mapping , Crosses, Genetic , DNA/genetics , DNA Primers/genetics , Female , GTP-Binding Proteins/metabolism , Genomic Library , In Situ Hybridization, Fluorescence , Male , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Receptors, Calcium-Sensing , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL