1
|
Rosa YFP, Noé GG, Merlo MGO, Calixto RR, Vidigal APP, Silva BFD, Silva KBD, Coelho VF, Minassa VS, Sampaio KN, Beijamini V. Chlorpyrifos intermittent exposure enhances cardiovascular but not behavioural responses to contextual fear conditioning in adult rats: Possible involvement of brain oxidative-nitrosative stress. Behav Brain Res 2025; 479:115358. [PMID: 39603423 DOI: 10.1016/j.bbr.2024.115358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/18/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
Exposure to organophosphorus compounds (OPs) may cause psychiatric, neurologic, biochemical, and cardiovascular abnormalities. Neurotoxicity of OP compounds is primarily due to irreversibly inhibition of the acetylcholinesterase (AChE) enzyme both centrally and peripherally. Chlorpyrifos (CPF) is a widely used OP classified as moderately toxic. Previously, it has been shown that CPF administration, given every other day to adult rats, impairs spatial memory and prepulse inhibition associated with brain AChE inhibition. Our group also found that intermittent treatment with CPF, simulating occupational exposure, impairs the cardiorespiratory reflexes and causes cardiac hypertrophy. Thereby, we aimed to examine whether subchronic and intermittent administration of CPF would affect the behavioural (freezing) and cardiovascular (mean arterial pressure, MAP; heart rate, HR) responses elicited during contextual fear conditioning (CFC) and extinction. Wistar adult male rats were injected with sublethal and intermittent CPF doses (4 and 7 mg/kg) three times a week for one month. Two days after the last injection, a range of tests were performed to assess depression (sucrose preference), anxiety (elevated plus-maze, EPM), locomotion (open field, OF), and conditioned fear expression and extinction. Separate cohorts of animals were euthanized to measure plasma butyrylcholinesterase (BChE), erythrocyte AChE, brain AChE activity, and markers of oxidative-nitrosative stress. Intermittent CPF treatment did not affect sucrose preference. CPF (4 and 7 mg/kg) reduced open-arms exploration in the EPM, suggesting an anxiogenic effect. The higher dose of CPF decreased the total distance travelled in the OFT, suggesting motor impairment. After a seven-day CPF-free washout period, CPF (7 mg/kg) increased the tachycardic response without affecting freezing behaviour in the CFC extinction session. CPF 7 mg/kg decreased AChE activity in the hippocampus, pre-frontal cortex and brainstem 72 after the last administration whilst transiently increasing oxidative-nitrosative stress specifically in the brainstem. Overall, our results outlined the behavioural, autonomic and biochemical abnormalities caused by an intermittent dosing regimen of CPF that elicits brain AChE inhibition and brain oxidative-nitrosative stress. This paradigm might be valuable in further exploring long-term consequences and mechanisms of OP neurotoxicity as well as comprehensive therapeutic approaches.
Collapse
Affiliation(s)
- Yuri Fernandes Pereira Rosa
- Pharmaceutical Sciences Graduate Program, Health Sciences Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Gabriel Gavazza Noé
- Pharmaceutical Sciences Graduate Program, Health Sciences Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Maria Gabriela Oliveira Merlo
- Department of Pharmaceutical Sciences, Health Science Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Raphael Rizzo Calixto
- Department of Pharmaceutical Sciences, Health Science Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Anna Paula Perin Vidigal
- Pharmaceutical Sciences Graduate Program, Health Sciences Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Bruna Ferreira da Silva
- Department of Pharmaceutical Sciences, Health Science Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Kissylla Brisson da Silva
- Department of Pharmaceutical Sciences, Health Science Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Vitória Fosse Coelho
- Department of Pharmaceutical Sciences, Health Science Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Vítor Sampaio Minassa
- Pharmaceutical Sciences Graduate Program, Health Sciences Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Karla Nívea Sampaio
- Pharmaceutical Sciences Graduate Program, Health Sciences Centre, Federal University of Espírito Santo, Vitória, ES, Brazil; Department of Pharmaceutical Sciences, Health Science Centre, Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Vanessa Beijamini
- Pharmaceutical Sciences Graduate Program, Health Sciences Centre, Federal University of Espírito Santo, Vitória, ES, Brazil; Department of Pharmaceutical Sciences, Health Science Centre, Federal University of Espírito Santo, Vitória, ES, Brazil.
| |
Collapse
|
2
|
Urquizu E, Paratusic S, Goyenechea J, Gómez-Canela C, Fumàs B, Pubill D, Raldúa D, Camarasa J, Escubedo E, López-Arnau R. Acute Paraoxon-Induced Neurotoxicity in a Mouse Survival Model: Oxidative Stress, Dopaminergic System Alterations and Memory Deficits. Int J Mol Sci 2024; 25:12248. [PMID: 39596313 PMCID: PMC11594717 DOI: 10.3390/ijms252212248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The secondary neurotoxicity induced by severe organophosphorus (OP) poisoning, including paraoxon (POX), is associated with cognitive impairments in survivors, who, despite receiving appropriate emergency treatments, may still experience lasting neurological deficits. Thus, the present study provides a survival mouse model of acute and severe POX poisoning to examine secondary neurotoxicity. Swiss CD-1 male mice were injected with POX (4 mg/kg, s.c.) followed by atropine (4 mg/kg, i.p.), pralidoxime (2-PAM; Pyridine-2-aldoxime methochloride) (25 mg/kg, i.p., twice, 1 h apart) and diazepam (5 mg/kg, i.p.), resulting in a survival rate >90% and Racine score of 5-6. Our results demonstrated that the model showed increased lipid peroxidation, downregulation of antioxidant enzymes and astrogliosis in the mouse hippocampus (HP) and prefrontal cortex (PFC), brain areas involved in cognitive functions. Moreover, dopamine (DA) levels were reduced in the hp, but increased in the PFC. Furthermore, the survival mouse model of acute POX intoxication did not exhibit phenotypic manifestations of depression, anxiety or motor incoordination. However, our results demonstrated long-term recognition memory impairments, which are in accordance with the molecular and neurochemical effects observed. In conclusion, this mouse model can aid in researching POX exposure's effects on memory and developing potential countermeasures against the secondary neurotoxicity induced by severe OP poisoning.
Collapse
Affiliation(s)
- Edurne Urquizu
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; (E.U.)
| | - Selma Paratusic
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; (E.U.)
| | - Júlia Goyenechea
- Department of Analytical Chemistry and Applied (Chromatography Section), School of Engineering, Institut Químic de Sarrià—Universitat Ramon Llull, 08017 Barcelona, Spain
| | - Cristian Gómez-Canela
- Department of Analytical Chemistry and Applied (Chromatography Section), School of Engineering, Institut Químic de Sarrià—Universitat Ramon Llull, 08017 Barcelona, Spain
| | - Berta Fumàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; (E.U.)
| | - David Pubill
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; (E.U.)
| | - Demetrio Raldúa
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), 08034 Barcelona, Spain
| | - Jordi Camarasa
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; (E.U.)
| | - Elena Escubedo
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; (E.U.)
| | - Raúl López-Arnau
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain; (E.U.)
| |
Collapse
|
3
|
Tsai YH, González EA, Grodzki ACG, Bruun DA, Saito NH, Harvey DJ, Lein PJ. Acute intoxication with diisopropylfluorophosphate promotes cellular senescence in the adult male rat brain. FRONTIERS IN TOXICOLOGY 2024; 6:1360359. [PMID: 38745692 PMCID: PMC11091247 DOI: 10.3389/ftox.2024.1360359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/12/2024] [Indexed: 05/16/2024] Open
Abstract
Acute intoxication with high levels of organophosphate (OP) cholinesterase inhibitors can cause cholinergic crisis, which is associated with acute, life-threatening parasympathomimetic symptoms, respiratory depression and seizures that can rapidly progress to status epilepticus (SE). Clinical and experimental data demonstrate that individuals who survive these acute neurotoxic effects often develop significant chronic morbidity, including behavioral deficits. The pathogenic mechanism(s) that link acute OP intoxication to chronic neurological deficits remain speculative. Cellular senescence has been linked to behavioral deficits associated with aging and neurodegenerative disease, but whether acute OP intoxication triggers cellular senescence in the brain has not been investigated. Here, we test this hypothesis in a rat model of acute intoxication with the OP diisopropylfluorophosphate (DFP). Adult male Sprague-Dawley rats were administered DFP (4 mg/kg, s.c.). Control animals were administered an equal volume (300 µL) of sterile phosphate-buffered saline (s.c.). Both groups were subsequently injected with atropine sulfate (2 mg/kg, i.m.) and 2-pralidoxime (25 mg/kg, i.m.). DFP triggered seizure activity within minutes that rapidly progressed to SE, as determined using behavioral seizure criteria. Brains were collected from animals at 1, 3, and 6 months post-exposure for immunohistochemical analyses of p16, a biomarker of cellular senescence. While there was no immunohistochemical evidence of cellular senescence at 1-month post-exposure, at 3- and 6-months post-exposure, p16 immunoreactivity was significantly increased in the CA3 and dentate gyrus of the hippocampus, amygdala, piriform cortex and thalamus, but not the CA1 region of the hippocampus or the somatosensory cortex. Co-localization of p16 immunoreactivity with cell-specific biomarkers, specifically, NeuN, GFAP, S100β, IBA1 and CD31, revealed that p16 expression in the brain of DFP animals is neuron-specific. The spatial distribution of p16-immunopositive cells overlapped with expression of senescence associated β-galactosidase and with degenerating neurons identified by FluoroJade-C (FJC) staining. The co-occurrence of p16 and FJC was positively correlated. This study implicates cellular senescence as a novel pathogenic mechanism underlying the chronic neurological deficits observed in individuals who survive OP-induced cholinergic crisis.
Collapse
Affiliation(s)
- Yi-Hua Tsai
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Eduardo A. González
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Ana C. G. Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Donald A. Bruun
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Naomi H. Saito
- Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Danielle J. Harvey
- Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
4
|
Golime R, Singh N, Rajput A, Dp N, Lodhi VK. Chronic sub lethal nerve agent (Soman) exposure induced long-term neurobehavioral, histological, and biochemical alterations in rats. J Chem Neuroanat 2024; 136:102388. [PMID: 38182038 DOI: 10.1016/j.jchemneu.2024.102388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/01/2024] [Accepted: 01/01/2024] [Indexed: 01/07/2024]
Abstract
Organophosphorus (OP) pesticides and insecticides are used in agriculture and other industries can also cause adverse effects through environmental exposures in the people working in agricultural and pesticide industries. OP nerve agent exposures have been associated with delayed neurotoxic effects including sleep disorders, cognitive malfunctions, and brain damage in Gulf War victims, and Japanese victims of terrorist attacks with nerve agents. However, the mechanisms behind such prolonged adverse effects after chronic OP nerve agent's exposures in survivors are not well understood. In the present study, male Wistar rats were subcutaneously exposed to nerve agent soman (0.25XLD50) for 21 consecutive days to evaluate the neurobehavioral, neuropathological and biochemical alterations (oxidative stress and antioxidants levels). Neurobehavioral studies using Elevated Plus Maze (EPM), T-Maze, and rotarod tests revealed that chronic soman exposure produced alterations in behavioral functions including increased anxiety and reduction in working memory and neuromuscular strength. Biochemical studies showed that antioxidants enzyme (glutathione peroxidase (GPx), catalase (CAT), and superoxide dismutase (SOD) levels were reduced and oxidative stress (reduced glutathione (GSH) and lipid peroxidation levels (malondialdehyde (MDA)) were significantly increased in brain at 30 days in soman exposed rats as compared to control rats. Neuroselective fluorojade-c stain was used to examine the brain damage after chronic soman exposure. Results demonstrated that chronic soman exposure induced neurodegeneration as brain damage was detected at 30- and 90-days post exposure. The present study results suggest that chronic nerve agent exposures even at low doses may produce long-term adverse effects like neurobehavioral deficits in rats.
Collapse
Affiliation(s)
- RamaRao Golime
- Biomedical Verification Division, Defence Research and Development Establishment (DRDE), Jhansi road, Gwalior, MP, India.
| | - Naveen Singh
- Biomedical Verification Division, Defence Research and Development Establishment (DRDE), Jhansi road, Gwalior, MP, India
| | - Ankush Rajput
- Biomedical Verification Division, Defence Research and Development Establishment (DRDE), Jhansi road, Gwalior, MP, India
| | - Nagar Dp
- Pharmacology and Toxicology Division, DRDE, Jhansi Road, Gwalior, MP, India
| | - Vinod K Lodhi
- Process Technology Development Division, DRDE, Jhansi Road, Gwalior, MP, India
| |
Collapse
|
5
|
Neff MJ, Reddy DS. Long-Term Neuropsychiatric Developmental Defects after Neonatal Organophosphate Exposure: Mitigation by Synthetic Neurosteroids. J Pharmacol Exp Ther 2024; 388:451-468. [PMID: 37863488 PMCID: PMC10806574 DOI: 10.1124/jpet.123.001763] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 10/22/2023] Open
Abstract
Children are much more susceptible to the neurotoxic effects of organophosphate (OP) pesticides and nerve agents than adults. OP poisoning in children leads to acute seizures and neuropsychiatric sequela, including the development of long-term disabilities and cognitive impairments. Despite these risks, there are few chronic rodent models that use pediatric OP exposure for studying neurodevelopmental consequences and interventions. Here, we investigated the protective effect of the neurosteroid ganaxolone (GX) on the long-term developmental impact of neonatal exposure to the OP compound, diisopropyl-fluorophosphate (DFP). Pediatric postnatal day-28 rats were acutely exposed to DFP, and at 3 and 10 months after exposure, they were evaluated using a series of cognitive and behavioral tests with or without the postexposure treatment of GX. Analysis of the neuropathology was performed after 10 months. DFP-exposed animals displayed significant long-term deficits in mood, anxiety, depression, and aggressive traits. In spatial and nonspatial cognitive tests, they displayed striking impairments in learning and memory. Analysis of brain sections showed significant loss of neuronal nuclei antigen(+) principal neurons, parvalbumin(+) inhibitory interneurons, and neurogenesis, along with increased astrogliosis, microglial neuroinflammation, and mossy fiber sprouting. These detrimental neuropathological changes are consistent with behavioral dysfunctions. In the neurosteroid GX-treated cohort, behavioral and cognitive deficits were significantly reduced and were associated with strong protection against long-term neuroinflammation and neurodegeneration. In conclusion, this pediatric model replicates the salient features of children exposed to OPs, and the protective outcomes from neurosteroid intervention support the viability of developing this strategy for mitigating the long-term effects of acute OP exposure in children. SIGNIFICANCE STATEMENT: An estimated 3 million organophosphate exposures occur annually worldwide, with children comprising over 30% of all victims. Our understanding of the neurodevelopmental consequences in children exposed to organophosphates is limited. Here, we investigated the long-term impact of neonatal exposure to diisopropyl-fluorophosphate in pediatric rats. Neurosteroid treatment protected against major deficits in behavior and memory and was well correlated with neuropathological changes. Overall, this pediatric model is helpful to screen novel therapies to mitigate long-term developmental deficits of organophosphate exposure.
Collapse
Affiliation(s)
- Michael James Neff
- Department of Neuroscience and Experimental Therapeutics (M.J.N., D.S.R.) and Institute of Pharmacology and Neurotherapeutics (M.J.N., D.S.R.), School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics (M.J.N., D.S.R.) and Institute of Pharmacology and Neurotherapeutics (M.J.N., D.S.R.), School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
6
|
Blair RE, Hawkins E, Pinchbeck LR, DeLorenzo RJ, Deshpande LS. Chronic Epilepsy and Mossy Fiber Sprouting Following Organophosphate-Induced Status Epilepticus in Rats. J Pharmacol Exp Ther 2024; 388:325-332. [PMID: 37643794 PMCID: PMC10801751 DOI: 10.1124/jpet.123.001739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
Organophosphate (OP) compounds are highly toxic and include pesticides and chemical warfare nerve agents. OP exposure inhibits the acetylcholinesterase enzyme, causing cholinergic overstimulation that can evolve into status epilepticus (SE) and produce lethality. Furthermore, OP-induced SE survival is associated with mood and memory dysfunction and spontaneous recurrent seizures (SRS). In male Sprague-Dawley rats, we assessed hippocampal pathology and chronic SRS following SE induced by administration of OP agents paraoxon (2 mg/kg, s.c.), diisopropyl fluorophosphate (4 mg/kg, s.c.), or O-isopropyl methylphosphonofluoridate (GB; sarin) (2 mg/kg, s.c.), immediately followed by atropine and 2-PAM. At 1-hour post-OP-induced SE onset, midazolam was administered to control SE. Approximately 6 months after OP-induced SE, SRS were evaluated using video and electroencephalography monitoring. Histopathology was conducted using hematoxylin and eosin (H&E), while silver sulfide (Timm) staining was used to assess mossy fiber sprouting (MFS). Across all the OP agents, over 60% of rats that survived OP-induced SE developed chronic SRS. H&E staining revealed a significant hippocampal neuronal loss, while Timm staining revealed extensive MFS within the inner molecular region of the dentate gyrus. This study demonstrates that OP-induced SE is associated with hippocampal neuronal loss, extensive MFS, and the development of SRS, all hallmarks of chronic epilepsy. SIGNIFICANCE STATEMENT: Models of organophosphate (OP)-induced SE offer a unique resource to identify molecular mechanisms contributing to neuropathology and the development of chronic OP morbidities. These models could allow the screening of targeted therapeutics for efficacious treatment strategies for OP toxicities.
Collapse
Affiliation(s)
- Robert E Blair
- Departments of Neurology (R.E.B., E.H., R.J.D., L.S.D.) and Pharmacology and Toxicology (R.J.D., L.S.D.) School of Medicine, and Department of Biology, College of Humanities & Sciences (L.R.P.), Virginia Commonwealth University, Richmond, Virginia
| | - Elisa Hawkins
- Departments of Neurology (R.E.B., E.H., R.J.D., L.S.D.) and Pharmacology and Toxicology (R.J.D., L.S.D.) School of Medicine, and Department of Biology, College of Humanities & Sciences (L.R.P.), Virginia Commonwealth University, Richmond, Virginia
| | - Lauren R Pinchbeck
- Departments of Neurology (R.E.B., E.H., R.J.D., L.S.D.) and Pharmacology and Toxicology (R.J.D., L.S.D.) School of Medicine, and Department of Biology, College of Humanities & Sciences (L.R.P.), Virginia Commonwealth University, Richmond, Virginia
| | - Robert J DeLorenzo
- Departments of Neurology (R.E.B., E.H., R.J.D., L.S.D.) and Pharmacology and Toxicology (R.J.D., L.S.D.) School of Medicine, and Department of Biology, College of Humanities & Sciences (L.R.P.), Virginia Commonwealth University, Richmond, Virginia
| | - Laxmikant S Deshpande
- Departments of Neurology (R.E.B., E.H., R.J.D., L.S.D.) and Pharmacology and Toxicology (R.J.D., L.S.D.) School of Medicine, and Department of Biology, College of Humanities & Sciences (L.R.P.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
7
|
Almeida MF, Farizatto KLG, Almeida RS, Bahr BA. Lifestyle strategies to promote proteostasis and reduce the risk of Alzheimer's disease and other proteinopathies. Ageing Res Rev 2024; 93:102162. [PMID: 38070831 DOI: 10.1016/j.arr.2023.102162] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/31/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Unhealthy lifestyle choices, poor diet, and aging can have negative influences on cognition, gradually increasing the risk for mild cognitive impairment (MCI) and the continuum comprising early dementia. Aging is the greatest risk factor for age-related dementias such as Alzheimer's disease, and the aging process is known to be influenced by life events that can positively or negatively affect age-related diseases. Remarkably, life experiences that make the brain vulnerable to dementia, such as seizure episodes, neurotoxin exposures, metabolic disorders, and trauma-inducing events (e.g. traumatic injuries or mild neurotrauma from a fall or blast exposure), have been associated with negative effects on proteostasis and synaptic integrity. Functional compromise of the autophagy-lysosomal pathway, a major contributor to proteostasis, has been implicated in Alzheimer's disease, Parkinson's disease, obesity-related pathology, Huntington's disease, as well as in synaptic degeneration which is the best correlate of cognitive decline. Correspondingly, pharmacological and non-pharmacological strategies that positively modulate lysosomal proteases are recognized as synaptoprotective through degradative clearance of pathogenic proteins. Here, we discuss life-associated vulnerabilities that influence key hallmarks of brain aging and the increased burden of age-related dementias. Additionally, we discuss exercise and diet among the lifestyle strategies that regulate proteostasis as well as synaptic integrity, leading to evident prevention of cognitive deficits during brain aging in pre-clinical models.
Collapse
Affiliation(s)
- Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina - Pembroke, Pembroke, NC 28372, USA; Department of Biology, University of North Carolina - Pembroke, Pembroke, NC 28372, USA; Department of Biology & Marine Biology, and the Integrative, Comparative & Marine Biology Program, University of North Carolina - Wilmington, Wilmington, NC 28409, USA
| | - Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina - Pembroke, Pembroke, NC 28372, USA
| | - Renato S Almeida
- Department of Biosciences, University of Taubate, Taubate, SP 12020-270, Brazil
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina - Pembroke, Pembroke, NC 28372, USA; Department of Biology, University of North Carolina - Pembroke, Pembroke, NC 28372, USA.
| |
Collapse
|
8
|
Zou S, Wang Q, He Q, Liu G, Song J, Li J, Wang F, Huang Y, Hu Y, Zhou D, Lv Y, Zhu Y, Wang B, Zhang L. Brain-targeted nanoreactors prevent the development of organophosphate-induced delayed neurological damage. J Nanobiotechnology 2023; 21:256. [PMID: 37550745 PMCID: PMC10405429 DOI: 10.1186/s12951-023-02039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Organophosphate (OP)-induced delayed neurological damage is attributed to permanent neuropathological lesions caused by irreversible OP-neurocyte interactions, without potent brain-targeted etiological antidotes to date. The development of alternative therapies to achieve intracerebral OP detoxification is urgently needed. METHODS We designed a brain-targeted nanoreactor by integrating enzyme immobilization and biomimetic membrane camouflaging protocols with careful characterization, and then examined its blood-brain barrier (BBB) permeability both in vitro and in vivo. Subsequently, the oxidative stress parameters, neuroinflammatory factors, apoptotic proteins and histopathological changes were measured and neurobehavioral tests were performed. RESULTS The well-characterized nanoreactors exerted favourable BBB penetration capability both in vitro and in vivo, significantly inhibiting OP-induced intracerebral damage. At the cellular and tissue levels, nanoreactors obviously blocked oxidative stress, cellular apoptosis, inflammatory reactions and brain histopathological damage. Furthermore, nanoreactors radically prevented the occurrence of OP-induced delayed cognitive deficits and psychiatric abnormality. CONCLUSION The nanoreactors significantly prevented the development of OP-induced delayed neurological damage, suggesting a potential brain-targeted etiological strategy to attenuate OP-related delayed neurological and neurobehavioral disorders.
Collapse
Affiliation(s)
- Shuaijun Zou
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Qianqian Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Qian He
- The Third Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Guoyan Liu
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Juxingsi Song
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Jie Li
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Fan Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Yichao Huang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Yanan Hu
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Dayuan Zhou
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Yongfei Lv
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China
| | - Yuanjie Zhu
- Department of Marine Biological Injury and Dermatology, Naval Special Medical Centre, Naval Medical University, Shanghai, 200052, China.
| | - Beilei Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China.
| | - Liming Zhang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Centre, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
9
|
Countermeasures in organophosphorus intoxication: pitfalls and prospects. Trends Pharmacol Sci 2022; 43:593-606. [DOI: 10.1016/j.tips.2022.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 11/24/2022]
|
10
|
Andrew PM, Lein PJ. Neuroinflammation as a Therapeutic Target for Mitigating the Long-Term Consequences of Acute Organophosphate Intoxication. Front Pharmacol 2021; 12:674325. [PMID: 34054549 PMCID: PMC8153682 DOI: 10.3389/fphar.2021.674325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
Acute intoxication with organophosphates (OPs) can cause a potentially fatal cholinergic crisis characterized by peripheral parasympathomimetic symptoms and seizures that rapidly progress to status epilepticus (SE). While current therapeutic countermeasures for acute OP intoxication significantly improve the chances of survival when administered promptly, they are insufficient for protecting individuals from chronic neurologic outcomes such as cognitive deficits, affective disorders, and acquired epilepsy. Neuroinflammation is posited to contribute to the pathogenesis of these long-term neurologic sequelae. In this review, we summarize what is currently known regarding the progression of neuroinflammatory responses after acute OP intoxication, drawing parallels to other models of SE. We also discuss studies in which neuroinflammation was targeted following OP-induced SE, and explain possible reasons why such therapeutic interventions have inconsistently and only partially improved long-term outcomes. Finally, we suggest future directions for the development of therapeutic strategies that target neuroinflammation to mitigate the neurologic sequelae of acute OP intoxication.
Collapse
Affiliation(s)
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, United States
| |
Collapse
|
11
|
Pinto Savall AS, Fidelis EM, Quines CB, Bresolin L, Gervini V, Pinton S. Potential role of a newly AChE reactivator in the depressive-like behavior induced by malathion. Neurosci Lett 2021; 749:135697. [PMID: 33540058 DOI: 10.1016/j.neulet.2021.135697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 11/27/2022]
Abstract
AChE inhibition caused by exposure to organophosphate (OP) compounds is strongly related to behavioural disorders such as depression. Malathion is an OP that already has a relationship between its exposure and behavioural changes, although few data still have its effects in a longer exposure protocol. In addition, intoxication therapy is based on the use of atropine-oxime which still has its controversial efficacy depending on the type of compound. For this, (3Z)-5-Chloro-3-(hydroxyimino)indolin-2-one (Cℓ-HIN), a compound that has properties of isatin and oxime in its structure, have shown reactivating properties in the activity of AChE that have been added to antidepressant-like effects in rats exposed to malathion in acute protocol. In this sense, effects of Cℓ-HIN on the depressive-like behaviour and AChE activity were evaluated in a protocol of subchronic exposure to malathion in rats. Male wistar rats were co-treated with Cℓ-HIN [5 mg/kg, p.o.] and/or malathion [1 or 10 mg/kg, i.p] for 20 days. The exposure to both doses of malathion increased immobility time of rats on the forced swimming test (FST). Besides, malathion inhibited the AChE activity in the prefrontal cortex of rats, but any significant difference was observed in the hippocampus. Cℓ-HIN protected against increased immobility time in the FST of those rats exposed to a dose of 1 mg/kg of malathion. Similarly, Cℓ-HIN was able to reactivate AChE activity only in that group exposed to the lowest dose of malathion. Collectively, the results of this study suggest that Cℓ-HIN is an oxime capable of reactivating AChE inhibited and presents na antidepressant-like effect in cases of prolonged exposure to malathion.
Collapse
Affiliation(s)
- Anne Suély Pinto Savall
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, ZIP code 97500-970, RS, Brazil.
| | | | - Caroline Brandão Quines
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, ZIP code 97500-970, RS, Brazil.
| | - Leandro Bresolin
- Federal University of Rio Grande - Campus Carreiros, Rio Grande, ZIP code 96201-900, RS, Brazil.
| | - Vanessa Gervini
- Federal University of Rio Grande - Campus Carreiros, Rio Grande, ZIP code 96201-900, RS, Brazil.
| | - Simone Pinton
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, ZIP code 97500-970, RS, Brazil.
| |
Collapse
|
12
|
Fitzgerald PJ, Hale PJ, Ghimire A, Watson BO. Repurposing Cholinesterase Inhibitors as Antidepressants? Dose and Stress-Sensitivity May Be Critical to Opening Possibilities. Front Behav Neurosci 2021; 14:620119. [PMID: 33519395 PMCID: PMC7840590 DOI: 10.3389/fnbeh.2020.620119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
When stress becomes chronic it can trigger lasting brain and behavioral changes including Major Depressive Disorder (MDD). There is conflicting evidence regarding whether acetylcholinesterase inhibitors (AChEIs) may have antidepressant properties. In a recent publication, we demonstrated a strong dose-dependency of the effect of AChEIs on antidepressant-related behavior in the mouse forced swim test: whereas the AChEI donepezil indeed promotes depression-like behavior at a high dose, it has antidepressant-like properties at lower doses in the same experiment. Our data therefore suggest a Janus-faced dose-response curve for donepezil in depression-related behavior. In this review, we investigate the mood-related properties of AChEIs in greater detail, focusing on both human and rodent studies. In fact, while there have been many studies showing pro-depressant activity by AChEIs and this is a major concept in the field, a variety of other studies in both humans and rodents show antidepressant effects. Our study was one of the first to systematically vary dose to include very low concentrations while measuring behavioral effects, potentially explaining the apparent disparate findings in the field. The possibility of antidepressant roles for AChEIs in rodents may provide hope for new depression treatments. Importantly, MDD is a psychosocial stress-linked disorder, and in rodents, stress is a major experimental manipulation for studying depression mechanisms, so an important future direction will be to determine the extent to which these depression-related effects are stress-sensitive. In sum, gaining a greater understanding of the potentially therapeutic mood-related effects of low dose AChEIs, both in rodent models and in human subjects, should be a prioritized topic in ongoing translational research.
Collapse
Affiliation(s)
- Paul J Fitzgerald
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Pho J Hale
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Anjesh Ghimire
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Brendon O Watson
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
13
|
Dose- and time-related effects of acute diisopropylfluorophosphate intoxication on forced swim behavior and sucrose preference in rats. Neurotoxicology 2020; 82:82-88. [PMID: 33232745 DOI: 10.1016/j.neuro.2020.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 11/20/2022]
Abstract
Acute intoxication by organophosphorus anticholinesterases (OPs) has been associated with depression and other neuropsychiatric disorders. We previously reported that adult male rats treated with diisopropylfluorophosphate (2.5 mg/kg, sc) showed acute cholinergic signs followed by changes (increased immobility/decreased swimming) in the forced swim test (a measure of behavioral despair) for at least one month. This study was conducted to evaluate the further persistence of changes in the forced swim test out to 4 months and to compare responses in a sucrose preference test, a measure of anhedonia. Adult male rats were treated with vehicle (peanut oil, 1 mL/kg, sc) or DFP (2.0, 2.25 or 2.5 mg/kg) followed by sacrifice 4 h later for measurement of OP-sensitive serine hydrolases (cholinesterase [ChE], fatty acid amide hydrolase [FAAH], and monoacylglycerol lipase [MAGL]) in hippocampus. Additional rats were treated similarly and evaluated for functional signs of acute toxicity from 30 min to 6 days, and then motor activity, forced swim behavior and sucrose preference at approximately 1 week, 1 month and 4 months after dosing. All dosages of DFP elicited serine hydrolase inhibition (ChE, 92-96 %; FAAH, 46-63 %; MAGL, 26-33 %). Body weight was reduced in all DFP-treated groups during the first two weeks, and lethality was noted with the higher dosages. Involuntary movements were elicited in all DFP treatment groups during the first week, but both time of onset and rate of recovery were dose-related. There was a significant reduction in ambulation at one week after the highest dosage (2.5 mg/kg), but no other significant locomotor changes were noted. Immobility was increased and swimming was decreased in the forced swim test at all three time-points by 2.25 mg/kg DFP, and at 2 of 3 time-points by the other dosages. While length of water deprivation and time after DFP dosing affected sucrose preference, DFP treatment had no main effect. We conclude that the forced swim test (a measure of behavioral despair/coping mechanism for inescapable stress) is a robust and persistent neurobehavioral consequence of acute DFP intoxication while sucrose preference, a measure of anhedonia and a common symptom of major clinical depression, is not affected.
Collapse
|
14
|
Deshpande LS, Blair RE, Halquist M, Kosmider L, DeLorenzo RJ. Intramuscular atenolol and levetiracetam reduce mortality in a rat model of paraoxon-induced status epilepticus. Ann N Y Acad Sci 2020; 1480:219-232. [PMID: 32961584 DOI: 10.1111/nyas.14500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/01/2022]
Abstract
Organophosphorus (OP) compounds are chemical threat agents and are irreversible inhibitors of the enzyme acetylcholinesterase that lead to a hypercholinergic response that could include status epilepticus (SE). SE particularly targets the heart and brain and despite existing therapies, it is still associated with significant mortality and morbidity. Here, we investigated the effect of intramuscular (i.m.) adjunct therapy consisting of atenolol (AT) and levetiracetam (LV) when administered after paraoxon (POX)-induced SE. The combination therapy was administered twice daily for 2, 7, or 14 days. POX exposure in rats produced rapid SE onset that was treated with atropine, pralidoxime chloride, and midazolam. Here, AT + LV therapy produced significant reductions in POX SE mortality assessed at 30 days post-SE. AT + LV therapy exhibited muscle pathology inflammation scores that were not significantly different from saline-treated controls. Pharmacokinetic analyses revealed that the i.m. route achieved faster and stabler plasma therapeutic levels for both AT and LV under OP SE conditions compared with oral administrations. Our data provide evidence of the safety and efficacy of i.m. AT + LV therapy for reducing mortality following POX SE.
Collapse
Affiliation(s)
- Laxmikant S Deshpande
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia.,Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Robert E Blair
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Matthew Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Leon Kosmider
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Robert J DeLorenzo
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia.,Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
15
|
Belinskaia DA, Goncharov NV. Theoretical and Practical Aspects of Albumin Esterase Activity. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1068162020030036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
16
|
Phillips KF, Santos E, Blair RE, Deshpande LS. Targeting Intracellular Calcium Stores Alleviates Neurological Morbidities in a DFP-Based Rat Model of Gulf War Illness. Toxicol Sci 2020; 169:567-578. [PMID: 30859209 PMCID: PMC6542335 DOI: 10.1093/toxsci/kfz070] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gulf War Illness (GWI) is a chronic multi-symptom disorder afflicting the veterans of the First Gulf War, and includes neurological symptoms characterized by depression and memory deficits. Chronic exposure to organophosphates (OPs) is considered a leading cause for GWI, yet its pathobiology is not fully understood. We recently observed chronic elevations in neuronal Ca2+ levels ([Ca2+]i) in an OP-diisopropyl fluorophosphate (DFP)-based rat model for GWI. This study was aimed at identifying mechanisms underlying elevated [Ca2+]i in this DFP model and investigating whether their therapeutic targeting could improve GWI-like neurological morbidities. Male Sprague-Dawley rats (9 weeks) were exposed to DFP (0.5 mg/kg, s.c., 1×-daily for 5 days) and at 3 months postDFP exposure, behavior was assessed and rats were euthanized for protein estimations and ratiometric Fura-2 [Ca2+]i estimations in acutely dissociated hippocampal neurons. In DFP rats, a sustained elevation in intracellular Ca2+ levels occurred, and pharmacological blockade of Ca2+-induced Ca2+-release mechanisms significantly lowered elevated [Ca2+]i in DFP neurons. Significant reductions in the protein levels of the ryanodine receptor (RyR) stabilizing protein Calstabin2 were also noted. Such a posttranslational modification would render RyR “leaky” resulting in sustained DFP [Ca2+]i elevations. Antagonism of RyR with levetiracetam significantly lower elevated [Ca2+]i in DFP neurons and improved GWI-like behavioral symptoms. Since Ca2+ is a major second messenger molecule, such chronic increases in its levels could underlie pathological synaptic plasticity that expresses itself as GWI morbidities. Our studies show that treatment with drugs targeted at blocking intracellular Ca2+ release could be effective therapies for GWI neurological morbidities.
Collapse
Affiliation(s)
| | | | | | - Laxmikant S Deshpande
- Departments of Neurology.,Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298
| |
Collapse
|
17
|
Guignet M, Dhakal K, Flannery BM, Hobson BA, Zolkowska D, Dhir A, Bruun DA, Li S, Wahab A, Harvey DJ, Silverman JL, Rogawski MA, Lein PJ. Persistent behavior deficits, neuroinflammation, and oxidative stress in a rat model of acute organophosphate intoxication. Neurobiol Dis 2020; 133:104431. [PMID: 30905768 PMCID: PMC6754818 DOI: 10.1016/j.nbd.2019.03.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/05/2019] [Accepted: 03/20/2019] [Indexed: 01/24/2023] Open
Abstract
Current medical countermeasures for organophosphate (OP)-induced status epilepticus (SE) are not effective in preventing long-term morbidity and there is an urgent need for improved therapies. Rat models of acute intoxication with the OP, diisopropylfluorophosphate (DFP), are increasingly being used to evaluate therapeutic candidates for efficacy in mitigating the long-term neurologic effects associated with OP-induced SE. Many of these therapeutic candidates target neuroinflammation and oxidative stress because of their implication in the pathogenesis of persistent neurologic deficits associated with OP-induced SE. Critical to these efforts is the rigorous characterization of the rat DFP model with respect to outcomes associated with acute OP intoxication in humans, which include long-term electroencephalographic, neurobehavioral, and neuropathologic effects, and their temporal relationship to neuroinflammation and oxidative stress. To address these needs, we examined a range of outcomes at later times post-exposure than have previously been reported for this model. Adult male Sprague-Dawley rats were given pyridostigmine bromide (0.1 mg/kg, im) 30 min prior to administration of DFP (4 mg/kg, sc), which was immediately followed by atropine sulfate (2 mg/kg, im) and pralidoxime (25 mg/kg, im). This exposure paradigm triggered robust electroencephalographic and behavioral seizures that rapidly progressed to SE lasting several hours in 90% of exposed animals. Animals that survived DFP-induced SE (~70%) exhibited spontaneous recurrent seizures and hyperreactive responses to tactile stimuli over the first 2 months post-exposure. Performance in the elevated plus maze, open field, and Pavlovian fear conditioning tests indicated that acute DFP intoxication reduced anxiety-like behavior and impaired learning and memory at 1 and 2 months post-exposure in the absence of effects on general locomotor behavior. Immunohistochemical analyses revealed significantly increased expression of biomarkers of reactive astrogliosis, microglial activation and oxidative stress in multiple brain regions at 1 and 2 months post-DFP, although there was significant spatiotemporal heterogeneity across these endpoints. Collectively, these data largely support the relevance of the rat model of acute DFP intoxication as a model for acute OP intoxication in the human, and support the hypothesis that neuroinflammation and/or oxidative stress represent potential therapeutic targets for mitigating the long-term neurologic sequelae of acute OP intoxication.
Collapse
Affiliation(s)
- Michelle Guignet
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616 USA, , , , , ,
| | - Kiran Dhakal
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616 USA, , , , , ,
| | - Brenna M. Flannery
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616 USA, , , , , ,
| | - Brad A. Hobson
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616 USA, , , , , ,
| | - Dorota Zolkowska
- Department of Neurology, School of Medicine, University of California-Davis, 4860 Y Street, Sacramento, CA 95817 USA, , , ;
| | - Ashish Dhir
- Department of Neurology, School of Medicine, University of California-Davis, 4860 Y Street, Sacramento, CA 95817 USA, , , ;
| | - Donald A. Bruun
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616 USA, , , , , ,
| | - Shuyang Li
- Department of Public Health Sciences, University of California-Davis, One Shields Avenue, Davis, CA 95616 USA, ,
| | - Abdul Wahab
- Department of Neurology, School of Medicine, University of California-Davis, 4860 Y Street, Sacramento, CA 95817 USA, , , ;
| | - Danielle J. Harvey
- Department of Public Health Sciences, University of California-Davis, One Shields Avenue, Davis, CA 95616 USA, ,
| | - Jill L. Silverman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California-Davis, 2230 Stockton Boulevard, Sacramento, CA 95817 USA,
- MIND Institute, School of Medicine, University of California-Davis, 2825 50 Street, Sacramento, CA 95817 USA
| | - Michael A. Rogawski
- Department of Neurology, School of Medicine, University of California-Davis, 4860 Y Street, Sacramento, CA 95817 USA, , , ;
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616 USA, , , , , ,
- MIND Institute, School of Medicine, University of California-Davis, 2825 50 Street, Sacramento, CA 95817 USA
| |
Collapse
|
18
|
Robidillo CJT, Wandelt S, Dalangin R, Zhang L, Yu H, Meldrum A, Campbell RE, Veinot JGC. Ratiometric Detection of Nerve Agents by Coupling Complementary Properties of Silicon-Based Quantum Dots and Green Fluorescent Protein. ACS APPLIED MATERIALS & INTERFACES 2019; 11:33478-33488. [PMID: 31414591 DOI: 10.1021/acsami.9b10996] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ratiometric photoluminescent detection of the toxicologically potent organophosphate ester nerve agents paraoxon (PX) and parathion (PT) using the complementary optical and chemical properties of the long Stokes shift green fluorescent protein variant, mAmetrine1.2 (mAm), and red-emitting silicon-based quantum dots (SiQDs) is reported. PX and PT selectively quench SiQD photoluminescence (PL) through a dynamic quenching mechanism, thereby, facilitating the development of a ratiometric sensor platform that shows micromolar limits of detection for PX and PT and that is unaffected by the presence of common inorganic and organic interferents. As a part of the present study, we also demonstrate that the paper-based sensors derived from mAm and SiQDs detect PX and PT at concentrations as low as 5 μM using a readily available commercial color analysis smartphone "app". The ratiometric sensor reported herein can potentially be used for the convenient and rapid on-site detection and quantification of PX and PT in real-world samples.
Collapse
Affiliation(s)
- Christopher Jay T Robidillo
- Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
- Department of Physical Sciences and Mathematics , University of the Philippines Manila , P. Faura Street , Ermita, Manila 1000 , Philippines
| | - Sophia Wandelt
- Faculty of Chemistry and Pharmacy , Ludwig-Maximilians-Universität München , Munich 81377 , Germany
| | - Rochelin Dalangin
- Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
| | - Lijuan Zhang
- Department of Physics , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | - Haoyang Yu
- Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
| | - Alkiviathes Meldrum
- Department of Physics , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | - Robert E Campbell
- Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
- Department of Chemistry , The University of Tokyo , Tokyo 113-0033 , Japan
| | - Jonathan G C Veinot
- Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
| |
Collapse
|
19
|
Farizatto KLG, Almeida MF, Long RT, Bahr BA. Early Synaptic Alterations and Selective Adhesion Signaling in Hippocampal Dendritic Zones Following Organophosphate Exposure. Sci Rep 2019; 9:6532. [PMID: 31024077 PMCID: PMC6484076 DOI: 10.1038/s41598-019-42934-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/11/2019] [Indexed: 11/09/2022] Open
Abstract
Organophosphates account for many of the world's deadliest poisons. They inhibit acetylcholinesterase causing cholinergic crises that lead to seizures and death, while survivors commonly experience long-term neurological problems. Here, we treated brain explants with the organophosphate compound paraoxon and uncovered a unique mechanism of neurotoxicity. Paraoxon-exposed hippocampal slice cultures exhibited progressive declines in synaptophysin, synapsin II, and PSD-95, whereas reduction in GluR1 was slower and NeuN and Nissl staining showed no indications of neuronal damage. The distinctive synaptotoxicity was observed in dendritic zones of CA1 and dentate gyrus. Interestingly, declines in synapsin II dendritic labeling correlated with increased staining for β1 integrin, a component of adhesion receptors that regulate synapse maintenance and plasticity. The paraoxon-induced β1 integrin response was targeted to synapses, and the two-fold increase in β1 integrin was selective as other synaptic adhesion molecules were unchanged. Additionally, β1 integrin-cofilin signaling was triggered by the exposure and correlations were found between the extent of synaptic decline and the level of β1 integrin responses. These findings identified organophosphate-mediated early and lasting synaptotoxicity which can explain delayed neurological dysfunction later in life. They also suggest that the interplay between synaptotoxic events and compensatory adhesion responses influences neuronal fate in exposed individuals.
Collapse
Affiliation(s)
- Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ronald T Long
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Chemistry and Physics, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.
| |
Collapse
|
20
|
Deshpande LS, DeLorenzo RJ. Novel therapeutics for treating organophosphate-induced status epilepticus co-morbidities, based on changes in calcium homeostasis. Neurobiol Dis 2019; 133:104418. [PMID: 30872159 DOI: 10.1016/j.nbd.2019.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/26/2019] [Accepted: 03/09/2019] [Indexed: 11/24/2022] Open
Abstract
Organophosphate (OP) chemicals include pesticides such as parathion, and nerve gases such as sarin and soman and are considered major chemical threat agents. Acute OP exposure is associated with a cholinergic crisis and status epilepticus (SE). It is also known that the survivors of OP toxicity exhibit neurobehavioral deficits such as mood changes, depression, and memory impairment, and acquired epilepsy. Our research has focused on addressing the need to develop effective therapeutic agents that could be administered even after prolonged seizures and would prevent or lessen the chronic morbidity associated with OP-SE survival. We have developed rat survival models of OP pesticide metabolite paraoxon (POX) and nerve agent sarin surrogate diisopropyl fluorophosphate (DFP) induced SE that are being used to screen for medical countermeasures against an OP attack. Our research has focused on studying neuronal calcium (Ca2+) homeostatic mechanisms for identifying mechanisms and therapeutics for the expression of neurological morbidities associated with OP-SE survival. We have observed development of a "Ca2+ plateau" characterized by sustained elevations in neuronal Ca2+ levels in OP-SE surviving rats that coincided with the appearance of OP-SE chronic morbidities. These Ca2+ elevations had their origin in Ca2+ release from the intracellular stores such that blockade with antagonists like dantrolene, carisbamate, and levetiracetam lowered OP-SE mediated Ca2+ plateau and afforded significant neuroprotection. Since the Ca2+ plateau lasts for a prolonged period, our studies suggest that blocking it after the control of SE may represent a unique target for development of novel countermeasures to prevent long term Ca2+ mediated OP-SE neuropsychiatric comorbidities such as depression, anxiety, and acquired epilepsy (AE).
Collapse
Affiliation(s)
- Laxmikant S Deshpande
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA; Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Robert J DeLorenzo
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA; Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
21
|
Aroniadou-Anderjaska V, Figueiredo TH, Apland JP, Braga MF. Targeting the glutamatergic system to counteract organophosphate poisoning: A novel therapeutic strategy. Neurobiol Dis 2019; 133:104406. [PMID: 30798006 DOI: 10.1016/j.nbd.2019.02.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/23/2019] [Accepted: 02/20/2019] [Indexed: 12/15/2022] Open
Abstract
One of the devastating effects of acute exposure to organophosphates, like nerve agents, is the induction of severe and prolonged status epilepticus (SE), which can cause death, or brain damage if death is prevented. Seizures after exposure are initiated by muscarinic receptor hyperstimulation-after inhibition of acetylcholinesterase by the organophosphorus agent and subsequent elevation of acetylcholine-but they are reinforced and sustained by glutamatergic hyperexcitation, which is the primary cause of brain damage. Diazepam is the FDA-approved anticonvulsant for the treatment of nerve agent-induced SE, and its replacement by midazolam is currently under consideration. However, clinical data derived from the treatment of SE of any etiology, as well as studies on the control of nerve agent-induced SE in animal models, have indicated that diazepam and midazolam control seizures only temporarily, their antiseizure efficacy is reduced as the latency of treatment from the onset of SE increases, and their neuroprotective efficacy is limited or absent. Here, we review data on the discovery of a novel anticonvulsant and neuroprotectant, LY293558, an AMPA/GluK1 receptor antagonist. Treatment of soman-exposed immature, young-adult, and aged rats with LY293558, terminates SE with limited recurrence of seizures, significantly protects from brain damage, and prevents long-term behavioral deficits, even when LY293558 is administered 1 h post-exposure. More beneficial effects and complete neuroprotection is obtained when LY293558 administration is combined with caramiphen, which antagonizes NMDA receptors. Further efficacy studies may bring the LY293558 + caramiphen combination therapy on the pathway to approval for human use.
Collapse
Affiliation(s)
- Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| | - James P Apland
- Neuroscience Program, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, United States of America.
| | - Maria F Braga
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| |
Collapse
|
22
|
Guignet M, Lein PJ. Neuroinflammation in organophosphate-induced neurotoxicity. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
23
|
Anxiolytic activity of paraoxon is associated with alterations in rat brain glutamatergic system. Neurotoxicol Teratol 2018; 71:32-40. [PMID: 30576762 DOI: 10.1016/j.ntt.2018.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 01/18/2023]
Abstract
Exposure to organophosphate (OP) compounds leads to behavioral alterations. To determine whether paraoxon has effects on anxiety, anxiety-like behaviors were assessed in paraoxon-exposed rats. Protein expression of glutamate transporters has also been measured in hippocampus and prefrontal cortex. Three doses of paraoxon (0.3, 0.7, or 1 mg/kg) or corn oil (vehicle) were intraperitoneally injected to adult male rats. At 14 or 28 days after exposure, behavioral tests were done using elevated plus-maze (EPM) or open field tests. Thereafter, animals were sacrificed and both hippocampi and prefrontal cortices were extracted for cholinesterase assay and western blotting. Animals treated with convulsive doses of paraoxon (0.7 and 1 mg/kg) showed an increase in percentage of time spent in open arms and percentage of open arm entries in the EPM. In the open field test, an increase in the time spent in central area was observed in rats treated with the same doses of paraoxon. These effects of paraoxon were independent of any changes in locomotor activity. There was an increase in both astrocytic glutamate transporter proteins (GLAST and GLT-1) in the hippocampus of animals treated with 0.7 and 1 mg/kg of paraoxon. In the prefrontal cortex, protein levels of the GLAST and GLT-1 increased in 0.7 and decreased in 1 mg/kg groups. Only a significant decrease in EAAC1 protein was observed in the prefrontal cortex at 14 days following exposure to 1 mg/kg of paraoxon. Collectively, this study showed that exposure to convulsive doses of paraoxon induced anxiolytic-like behaviors in both behavioral tests. This effect may be attributed to alterations of glutamate transporter proteins in the rat hippocampus and prefrontal cortex.
Collapse
|
24
|
Dunn EN, Ferrara-Bowens TM, Chachich ME, Honnold CL, Rothwell CC, Hoard-Fruchey HM, Lesyna CA, Johnson EA, Cerasoli DM, McDonough JH, Cadieux CL. Evaluating mice lacking serum carboxylesterase as a behavioral model for nerve agent intoxication. Toxicol Mech Methods 2018; 28:563-572. [PMID: 29768075 DOI: 10.1080/15376516.2018.1476637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mice and other rodents are typically utilized for chemical warfare nerve agent research. Rodents have large amounts of carboxylesterase in their blood, while humans do not. Carboxylesterase nonspecifically binds to and detoxifies nerve agent. The presence of this natural bioscavenger makes mice and other rodents poor models for studies identifying therapeutics to treat humans exposed to nerve agents. To obviate this problem, a serum carboxylesterase knockout (Es1 KO) mouse was created. In this study, Es1 KO and wild type (WT) mice were assessed for differences in gene expression, nerve agent (soman; GD) median lethal dose (MLD) values, and behavior prior to and following nerve agent exposure. No expression differences were detected between Es1 KO and WT mice in more than 34 000 mouse genes tested. There was a significant difference between Es1 KO and WT mice in MLD values, as the MLD for GD-exposed WT mice was significantly higher than the MLD for GD-exposed Es1 KO mice. Behavioral assessments of Es1 KO and WT mice included an open field test, a zero maze, a Barnes maze, and a sucrose preference test (SPT). While sex differences were observed in various measures of these tests, overall, Es1 KO mice behaved similarly to WT mice. The two genotypes also showed virtually identical neuropathological changes following GD exposure. Es1 KO mice appear to have an enhanced susceptibility to GD toxicity while retaining all other behavioral and physiological responses to this nerve agent, making the Es1 KO mouse a more human-like model for nerve agent research.
Collapse
Affiliation(s)
- Emily N Dunn
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Teresa M Ferrara-Bowens
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Mark E Chachich
- b Department of Psychology , Towson University , Towson , MD , USA
| | - Cary L Honnold
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Cristin C Rothwell
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Heidi M Hoard-Fruchey
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Catherine A Lesyna
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Erik A Johnson
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Douglas M Cerasoli
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - John H McDonough
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - C Linn Cadieux
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| |
Collapse
|
25
|
Apland JP, Aroniadou-Anderjaska V, Figueiredo TH, Pidoplichko VI, Rossetti K, Braga MFM. Comparing the Antiseizure and Neuroprotective Efficacy of LY293558, Diazepam, Caramiphen, and LY293558-Caramiphen Combination against Soman in a Rat Model Relevant to the Pediatric Population. J Pharmacol Exp Ther 2018; 365:314-326. [PMID: 29467308 PMCID: PMC5878669 DOI: 10.1124/jpet.117.245969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
The currently Food and Drug Administration-approved anticonvulsant for the treatment of status epilepticus (SE) induced by nerve agents is the benzodiazepine diazepam; however, diazepam does not appear to offer neuroprotective benefits. This is of particular concern with respect to the protection of children because, in the developing brain, synaptic transmission mediated via GABAA receptors, the target of diazepam, is weak. In the present study, we exposed 21-day-old male rats to 1.2 × LD50 soman and compared the antiseizure, antilethality, and neuroprotective efficacy of diazepam (10 mg/kg), LY293558 (an AMPA/GluK1 receptor antagonist; 15 mg/kg), caramiphen (CRM, an antimuscarinic with NMDA receptor-antagonistic properties; 50 mg/kg), and LY293558 (15 mg/kg) + CRM (50 mg/kg), administered 1 hour after exposure. Diazepam, LY293558, and LY293558 + CRM, but not CRM alone, terminated SE; LY293558 + CRM treatment acted significantly faster and produced a survival rate greater than 85%. Thirty days after soman exposure, neurodegeneration in limbic regions was most severe in the CRM-treated group, minimal to severe-depending on the region-in the diazepam group, absent to moderate in the LY293558-treated group, and totally absent in the LY293558 + CRM group. Amygdala and hippocampal atrophy, a severe reduction in spontaneous inhibitory activity in the basolateral amygdala, and increased anxiety-like behavior in the open-field and acoustic startle response tests were present in the diazepam and CRM groups, whereas the LY293558 and LY293558 + CRM groups did not differ from controls. The combined administration of LY293558 and CRM, by blocking mainly AMPA, GluK1, and NMDA receptors, is a very effective anticonvulsant and neuroprotective therapy against soman in young rats.
Collapse
Affiliation(s)
- James P Apland
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Vassiliki Aroniadou-Anderjaska
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Taiza H Figueiredo
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Volodymyr I Pidoplichko
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Katia Rossetti
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Maria F M Braga
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
26
|
Farizatto KLG, Bahr BA. Paraoxon: An Anticholinesterase That Triggers an Excitotoxic Cascade of Oxidative Stress, Adhesion Responses, and Synaptic Compromise. ACTA ACUST UNITED AC 2017; 13:29-37. [PMID: 29805717 DOI: 10.19044/esj.2017.c1p4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The anticholinesterase paraoxon (Pxn) is an organophosphate (OP) and the active metabolite of the insecticide parathion. It potently inhibits the enzyme acetylcholinesterase and leads to enhanced glutamate release, diminished GABA uptake, oxidative damage, and neurodegeneration. The resulting increased levels of acetylcholine can trigger seizures and cause neuronal and excitotoxic damage in the brain. The brain susceptibility related to anticholinesterase toxins extends beyond potential brain damage and death from toxic levels of the agent. Asymptomatic low-level exposure to such toxins can also leave the brain vulnerable or even cause it to exhibit neurological problems later in life. The actions of Pxn and similar neurotoxins have been studied in order to examine the events associated with anticholinesterase toxicity in the brain. A recent study demonstrated that Pxn exposure initiates a pathogenic cascade involving seizure events and subsequent signs of damage including unique presynaptic vulnerability and associated behavioral deficits. In addition, Pxn-mediated synaptotoxicity is also associated with enhanced production of oxidative stress as well as integrin adhesion responses. These findings provide a better understanding of the molecular events involved in Pxn toxicity.
Collapse
Affiliation(s)
- Karen L G Farizatto
- Biotechnology Research and Training Center, William C. Friday Laboratory, University of North Carolina - Pembroke, Pembroke, North Carolina, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, William C. Friday Laboratory, University of North Carolina - Pembroke, Pembroke, North Carolina, USA
| |
Collapse
|
27
|
Differential expression of glutamate transporters in cerebral cortex of paraoxon-treated rats. Neurotoxicol Teratol 2017; 62:20-26. [PMID: 28603072 DOI: 10.1016/j.ntt.2017.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/12/2017] [Accepted: 06/07/2017] [Indexed: 01/05/2023]
Abstract
Glutamatergic system is involved in pathological effects of organophosphorus (OP) compounds. We aimed to determine in vivo effects of paraoxon, the bioactive metabolite of parathion, on the expression of glutamate transporters as well as Bax and Bcl2 in rat cerebral cortex. Male Wistar rats received an intraperitoneal (i.p.) injection of one of three doses of paraoxon (0.3, 0.7, or 1mg/kg) or corn oil as vehicle (1ml/kg). After 4 or 18h, cerebral cortices were dissected out and used for quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blot assays to measure mRNA and protein levels, respectively. The cortical glial glutamate transporters (GLAST and GLT-1) were up-regulated in animals treated with 0.7mg/kg of paraoxon, but down-regulated in 1mg/kg group. Neuronal glutamate transporter (EAAC1) was unchanged in 0.7mg/kg treated rats, while reduced in 1mg/kg group. No significant difference was found in the mRNA and protein expression of EAAC1 in animals intoxicated with 0.3mg/kg of paraoxon. Paraoxon (1mg/kg) resulted in an up-regulation of Bax and down-regulation of Bcl2 mRNA levels in the rat cerebral cortex. These results indicate that paraoxon can differentially regulate expression of glutamate transporters at mRNA and protein levels in the cerebral cortex. Changes in the expression of glutamate transporters are closely related to paraoxon-induced seizure activity.
Collapse
|
28
|
Qiao J, Rong L, Wang Z, Zhang M. Involvement of Akt/GSK3β/CREB signaling pathway on chronic omethoate induced depressive-like behavior and improvement effects of combined lithium chloride and astaxanthin treatment. Neurosci Lett 2017; 649:55-61. [PMID: 28366776 DOI: 10.1016/j.neulet.2017.03.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 03/26/2017] [Accepted: 03/28/2017] [Indexed: 11/25/2022]
Abstract
Chronic organophosphorus pesticides (OP) exposure is associated with an increased risk of depression, and there is an urgent need to find an effective treatment for the depressive-like symptoms caused by OP. The main purpose of this study was to investigate whether combined lithium chloride (LiCl) and astaxanthin (AST) treatment would manifest synergetic antidepressant effects on mice with chronic OP exposure, and to determine the role of the Akt/GSK3β/CREB signaling pathway. Our results showed that chronic omethoate exposure significantly increased immobility time in behavioral tests and induced neuron damage in HE staining. The expression of p-GSK3β, p-CREB, p-PI3K and p-Akt in hippocampus after OP exposure were significantly down-regulated, while the influences were reversed by LiCl and AST treatment. Moreover, the combined application of AST and LiCl had synergistic therapeutic effects compared to LiCl and AST treatment alone, the expression of p-GSK3β, p-CREB, p-PI3K and p-Akt after combined LiCl-AST treatment were significantly higher than that with single drug application. These results showed that the combination of LiCl and AST could efficiently ameliorate depressive-like behavior induced by omethoate, and Akt/GSK3β/CREB signaling pathway might be responsible for the neuroprotective effect.
Collapse
Affiliation(s)
- Jingwen Qiao
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China; Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Lixia Rong
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhaoyue Wang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Meizeng Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
29
|
Mohammadi M, Zare Z, Allah-Moradi E, Vaezi N, Valadan R, Tehrani M. Alterations in mRNA and protein expression of glutamate transporters in rat hippocampus after paraoxon exposure. Neurotoxicology 2016; 57:251-257. [PMID: 27769869 DOI: 10.1016/j.neuro.2016.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/15/2016] [Accepted: 10/15/2016] [Indexed: 01/30/2023]
Abstract
Organophosphates affect brain function through a variety of mechanisms beyond their shared role as cholinesterase inhibitors. The aim of the current study was to investigate the changes in the expression of glial (GLAST and GLT-1) and neuronal (EAAC1) glutamate transporters at mRNA and protein levels in paraoxon-treated rat hippocampus. Adult male Wistar rats were intraperitoneally treated with either vehicle (corn oil) or one of three dosages of paraoxon (0.3, 0.7 or 1mg/kg). After 4 or 18h, both hippocampi of each rat were collected to detect mRNA and protein expression of glutamate transporters using the quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blotting, respectively. Animals treated with 0.3mg/kg paraoxon showed no difference in mRNA and protein levels of the glutamate transporters when compared with control group. At 4h after exposure with 0.7 and 1mg/kg paraoxon, the expression of GLAST and GLT-1 increased at mRNA and protein levels and remained elevated after 18h. No difference in the expression of EAAC1 at mRNA and protein levels was observed in any paraoxon-treated groups compared with the control group. This study showed an increased expression of glial (GLAST and GLT-1), but not neuronal (EAAC1) glutamate transporters, in adult rat hippocampus following administration of convulsive dosages of paraoxon. These suggest a protective and compensatory adaptation for effective uptake of glutamate in hippocampus induced by paraoxon and thus attenuating seizure activity.
Collapse
Affiliation(s)
- Moslem Mohammadi
- Department of Physiology & Pharmacology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zohreh Zare
- Department of Anatomical Sciences, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Esmaeil Allah-Moradi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narges Vaezi
- Department of Toxicology and Pharmacology, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Valadan
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohsen Tehrani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
30
|
Deshpande LS, Blair RE, Phillips KF, DeLorenzo RJ. Role of the calcium plateau in neuronal injury and behavioral morbidities following organophosphate intoxication. Ann N Y Acad Sci 2016; 1374:176-83. [PMID: 27327161 DOI: 10.1111/nyas.13122] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 12/28/2022]
Abstract
Organophosphate (OP) chemicals include nerve agents and pesticides, and there is a growing concern of OP-based chemical attacks against civilians. Current antidotes are essential in limiting immediate mortality associated with OP exposure. However, further research is needed to identify the molecular mechanisms underlying long-term neurological deficits following survival of OP toxicity in order to develop effective therapeutics. We have developed rat survival models of OP-induced status epilepticus (SE) that mimic chronic mortality and morbidity following OP intoxication. We have observed significant elevations in hippocampal calcium levels after OP SE that persisted for weeks following initial survival. Drugs inhibiting intracellular calcium-induced calcium release, such as dantrolene, levetiracetam, and carisbamate, lowered OP SE-mediated protracted calcium elevations. Given the critical role of calcium signaling in modulating behavior and cell death mechanisms, drugs targeted at preventing the development of the calcium plateau could enhance neuroprotection, help reduce morbidity, and improve outcomes following survival of OP SE.
Collapse
Affiliation(s)
- Laxmikant S Deshpande
- Departments of Neurology, Virginia Commonwealth University, Richmond, Virginia.,Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Robert E Blair
- Departments of Neurology, Virginia Commonwealth University, Richmond, Virginia
| | - Kristin F Phillips
- Departments of Neurology, Virginia Commonwealth University, Richmond, Virginia
| | - Robert J DeLorenzo
- Departments of Neurology, Virginia Commonwealth University, Richmond, Virginia.,Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
31
|
Pharmacological blockade of the calcium plateau provides neuroprotection following organophosphate paraoxon induced status epilepticus in rats. Neurotoxicol Teratol 2016; 56:81-86. [PMID: 27224207 DOI: 10.1016/j.ntt.2016.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/05/2016] [Accepted: 05/16/2016] [Indexed: 12/25/2022]
Abstract
Organophosphate (OP) compounds which include nerve agents and pesticides are considered chemical threat agents. Currently approved antidotes are crucial in limiting OP mediated acute mortality. However, survivors of lethal OP exposure exhibit delayed neuronal injury and chronic behavioral morbidities. In this study, we investigated neuroprotective capabilities of dantrolene and carisbamate in a rat survival model of paraoxon (POX) induced status epilepticus (SE). Significant elevations in hippocampal calcium levels were observed 48-h post POX SE survival, and treatment with dantrolene (10mg/kg, i.m.) and carisbamate (90mg/kg, i.m.) lowered these protracted calcium elevations. POX SE induced delayed neuronal injury as characterized by Fluoro Jade C labeling was observed in critical brain areas including the dentate gyrus, parietal cortex, amygdala, and thalamus. Dantrolene and carisbamate treatment provided significant neuroprotection against delayed neuronal damage in these brain regions when administered one-hour after POX-SE. These results indicate that dantrolene or carisbamate could be effective adjuvant therapies to the existing countermeasures to reduce neuronal injury and behavioral morbidities post OP SE survival.
Collapse
|
32
|
Phillips KF, Deshpande LS. Repeated low-dose organophosphate DFP exposure leads to the development of depression and cognitive impairment in a rat model of Gulf War Illness. Neurotoxicology 2015; 52:127-33. [PMID: 26619911 DOI: 10.1016/j.neuro.2015.11.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/20/2015] [Accepted: 11/22/2015] [Indexed: 02/02/2023]
Abstract
Approximately 175,000-250,000 of the returning veterans from the 1991 Persian Gulf War exhibit chronic multi-symptom illnesses that includes neurologic co-morbidities such as depression, anxiety and cognitive impairments. Amongst a host of causative factors, exposure to low levels of the nerve agent Sarin has been strongly implicated for expression of Gulf War Illness (GWI). Nerve agents similar to pesticides are organophosphate (OP) compounds. There is evidence from civilian population that exposure to OPs such as in agricultural workers and nerve agents such as the survivors and first-responders of the Tokyo subway Sarin gas attack suffer from chronic neurological problems similar to GWI symptoms. Given this unique chemical profile, OPs are ideal to study the effects of nerve agents and develop models of GWI in civilian laboratories. In this study, we used repeated low-dose exposure to OP agent diisopropyl fluorophosphate (DFP) over a 5-day period to approximate the duration and level of Sarin exposure during the Persian Gulf War. We tested the rats at 3-months post DFP exposure. Using a battery of behavioral assays, we observed the presence of symptoms of chronic depression, anxiety and memory problems as characterized by increased immobility time in the Forced Swim Test, anhedonia in the Sucrose Preference Test, anxiety in the Elevated Plus Maze, and spatial memory impairments in the Object Location Test, respectively. Chronic low dose DFP exposure was also associated with hippocampal neuronal damage as characterized by the presence of Fluoro-Jade staining. Given that OP exposure is considered a leading cause of GWI related morbidities, this animal model will be ideally suited to study underlying molecular mechanisms for the expression of GWI neurological symptoms and identify drugs for the effective treatment of GWIs.
Collapse
Affiliation(s)
- Kristin F Phillips
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | |
Collapse
|