1
|
Krucik DDR, Cook P, Cathey M, Meegan JM, Gomez FM, Bonn WV, Le-Bert C. Adult-onset epilepsy and hippocampal pathology in a California sea lion (Zalophus californianus): A case study of suspected in utero exposure to domoic acid. Neurotoxicology 2023; 96:13-18. [PMID: 36871906 DOI: 10.1016/j.neuro.2023.02.010] [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: 11/10/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
Domoic acid (DA) is a naturally occurring marine neurotoxin produced by Pseudo-nitzschia diatoms. Adult California sea lions (Zalophus californianus) can experience multiple post-exposure syndromes, including acute toxicosis and chronic epilepsy. Additionally, a delayed-onset epileptic syndrome is proposed for California sea lions (CSL) exposed in utero. This brief report explores a case of a CSL developing adult-onset epilepsy with progressive hippocampal neuropathology. Initial brain magnetic resonance imaging (MRI) and hippocampal volumetric analyses relative to brain size were normal. Approximately 7 years later, MRI studies to evaluate a newly developed epileptic syndrome demonstrated unilateral hippocampal atrophy. While other causes of unilateral hippocampal atrophy cannot be completely excluded, this case may represent in vivo evidence of adult-onset epileptiform DA toxicosis in a CSL. By estimating in utero DA exposure time period, and extrapolating from studies conducted on laboratory species, this case provides circumstantial evidence for a neurodevelopmental explanation correlating in utero exposure to adult-onset disease. Evidence of delayed disease development secondary to gestational exposure to naturally occurring DA has broad implications for marine mammal medicine and public health.
Collapse
Affiliation(s)
- David D R Krucik
- Cummings School of Veterinary Medicine at Tufts University, Grafton, MA 01581, USA
| | - Peter Cook
- New College of Florida, Sarasota, FL 34243, USA
| | | | | | | | | | - Carolina Le-Bert
- US Navy Marine Mammal Program, Naval Information Warfare Center Pacific, San Diego, CA 92152, USA.
| |
Collapse
|
2
|
Simeone C, Fauquier D, Skidmore J, Cook P, Colegrove K, Gulland F, Dennison S, Rowles TK. Clinical signs and mortality of non-released stranded California sea lions housed in display facilities: the suspected role of prior exposure to algal toxins. Vet Rec 2019; 185:304. [PMID: 31427410 PMCID: PMC6817987 DOI: 10.1136/vr.105371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/28/2019] [Accepted: 07/09/2019] [Indexed: 01/19/2023]
Abstract
Stranded California sea lions considered unable to survive in the wild are often placed in public display facilities. Exposure to the biotoxin domoic acid (DA) is a common cause of stranding, and chronic effects are observed long after initial exposure. Medical records for 171 sea lions placed in US institutions between 2000 and 2016 were reviewed, including results from clinical examinations, histopathology, behavioural testing and advanced imaging. There was a statistically significant increase in neurological disease detected in neonates (24%) compared with other age classes (11%). Sixty per cent of all neurological cases died during the study period. In the 11 neurological neonate cases, six died (55%) and five are still alive with three of five developing epilepsy during placement. Of the six neurological neonate cases that died, one was attributed to DA toxicosis, one to seizures and four to acute unexplained neurological disease. This survey suggests delayed neurological disease can develop in sea lions after stranding as neonates. These data coupled with stranding records and epidemiological data on DA-producing algal blooms suggest further research into effects of neonatal exposure to DA on risk of neurological disease in later life is warranted. California sea lions offer a natural model of DA exposure to study such effects.
Collapse
Affiliation(s)
- Claire Simeone
- The Marine Mammal Center, Sausalito, California, USA .,Office of Protected Resources, National Marine Fisheries Service, Silver Spring, Maryland, USA
| | - Deborah Fauquier
- Office of Protected Resources, National Marine Fisheries Service, Silver Spring, Maryland, USA
| | - Jennifer Skidmore
- Office of Protected Resources, National Marine Fisheries Service, Silver Spring, Maryland, USA
| | - Peter Cook
- New College of Florida, Sarasota, Florida, USA
| | - Kathleen Colegrove
- Zoological Pathology Program, University of Illinois at Urbana-Champaign College of Veterinary Medicine, Urbana, Illinois, USA
| | - Frances Gulland
- The Marine Mammal Center, Sausalito, California, USA.,Wildlife Health Center, University of California-Davis, Davis, California, USA
| | | | - Teresa K Rowles
- Office of Protected Resources, National Marine Fisheries Service, Silver Spring, Maryland, USA
| |
Collapse
|
3
|
Jing J, Petroff R, Shum S, Crouthamel B, Topletz AR, Grant KS, Burbacher TM, Isoherranen N. Toxicokinetics and Physiologically Based Pharmacokinetic Modeling of the Shellfish Toxin Domoic Acid in Nonhuman Primates. Drug Metab Dispos 2018; 46:155-165. [PMID: 29150543 PMCID: PMC5776359 DOI: 10.1124/dmd.117.078485] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/13/2017] [Indexed: 12/16/2022] Open
Abstract
Domoic acid (DA), a neurotoxin, is produced by marine algae and has caused toxications worldwide in animals and humans. However, the toxicokinetics of DA have not been fully evaluated, and information is missing on the disposition of DA following oral exposures at doses that are considered safe for human consumption. In this study, toxicokinetics of DA were investigated in cynomolgus monkeys following single doses of 5 µg/kg DA intravenously, 0.075 mg/kg DA orally, and 0.15 mg/kg DA orally. After intravenous dosing, DA had a systemic clearance of 124 ± 71 (ml/h)/kg, volume of distribution at steady state of 131 ± 71 ml/kg and elimination half-life of 1.2 ± 1.1 hours. However, following oral dosing, the average terminal half-life of DA was 11.3 ± 2.4 hours, indicating that DA disposition follows flip-flop kinetics with slow, rate-limiting absorption. The absorption of DA was low after oral dosing with absolute bioavailability of 6% ± 4%. The renal clearance of DA was variable [21-152 (ml/h)/kg] with 42% ± 11% of the intravenous DA dose recovered in urine. A physiologically based pharmacokinetic model was developed for DA in monkeys and humans that replicated the flip-flop kinetics observed after oral administration and allowed simulation of urinary excretion and brain and kidney distribution of DA following intravenous and oral dosing. This study is the first to characterize DA disposition at exposure levels close to the current estimated tolerable daily intake and to mechanistically model DA disposition in a model species, providing important information of the toxicokinetics of DA for human safety assessment.
Collapse
Affiliation(s)
- Jing Jing
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Rebekah Petroff
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Sara Shum
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Brenda Crouthamel
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Ariel R Topletz
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Kimberly S Grant
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Thomas M Burbacher
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Nina Isoherranen
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| |
Collapse
|
4
|
Mills BD, Pearce HL, Khan O, Jarrett BR, Fair DA, Lahvis GP. Prenatal domoic acid exposure disrupts mouse pro-social behavior and functional connectivity MRI. Behav Brain Res 2016; 308:14-23. [PMID: 27050322 PMCID: PMC4918767 DOI: 10.1016/j.bbr.2016.03.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/18/2016] [Accepted: 03/22/2016] [Indexed: 12/29/2022]
Abstract
Domoic acid (DA) is a toxin produced by marine algae and known primarily for its role in isolated outbreaks of Amnestic Shellfish Poisoning and for the damage it inflicts on marine mammals, particularly California sea lions. Lethal effects of DA are often preceded by seizures and coma. Exposure to DA during development can result in subtle and highly persistent effects on brain development and include behavioral changes that resemble diagnostic features of schizophrenia and anomalies in social behavior we believe are relevant to autism spectrum disorder (ASD). To more fully examine this hypothesis, we chose to examine adolescent mice exposed in utero to DA for endpoints relevant to ASD, specifically changes in social behavior and network structure, the latter measured by resting state functional connectivity (rs-fcMRI). We found that male offspring exposed in utero to DA expressed reproducible declines in social interaction and atypical patterns of functional connectivity in the anterior cingulate, a region of the default mode network that is critical for social functioning. We also found disruptions in global topology in regions involved in the processing of reward, social, and sensory experiences. Finally, we found that DA exposed males expressed a pattern of local over-connectivity. These anomalies in brain connectivity bear resemblance to connectivity patterns in ASD and help validate DA-exposed mice as a model of this mental disability.
Collapse
Affiliation(s)
- Brian D Mills
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States
| | - Hadley L Pearce
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States
| | - Omar Khan
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States
| | - Ben R Jarrett
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States
| | - Damien A Fair
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; Department of Psychiatry, Oregon Health & Science University, Portland, OR 97239, United States; Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, United States
| | - Garet P Lahvis
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|
5
|
Funk JA, Janech MG, Dillon JC, Bissler JJ, Siroky BJ, Bell PD. Characterization of renal toxicity in mice administered the marine biotoxin domoic Acid. J Am Soc Nephrol 2014; 25:1187-97. [PMID: 24511141 PMCID: PMC4033377 DOI: 10.1681/asn.2013080836] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/24/2013] [Indexed: 11/03/2022] Open
Abstract
Domoic acid (DA), an excitatory amino acid produced by diatoms belonging to the genus Pseudo-nitzschia, is a glutamate analog responsible for the neurologic condition referred to as amnesic shellfish poisoning. To date, the renal effects of DA have been underappreciated, although renal filtration is the primary route of systemic elimination and the kidney expresses ionotropic glutamate receptors. To characterize the renal effects of DA, we administered either a neurotoxic dose of DA or doses below the recognized limit of toxicity to adult Sv128/Black Swiss mice. DA preferentially accumulated in the kidney and elicited marked renal vascular and tubular damage consistent with acute tubular necrosis, apoptosis, and renal tubular cell desquamation, with toxic vacuolization and mitochondrial swelling as hallmarks of the cellular damage. Doses≥0.1 mg/kg DA elevated the renal injury biomarkers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin, and doses≥0.005 mg/kg induced the early response genes c-fos and junb. Coadministration of DA with the broad spectrum excitatory amino acid antagonist kynurenic acid inhibited induction of c-fos, junb, and neutrophil gelatinase-associated lipocalin. These findings suggest that the kidney may be susceptible to excitotoxic agonists, and renal effects should be considered when examining glutamate receptor activation. Additionally, these results indicate that DA is a potent nephrotoxicant, and potential renal toxicity may require consideration when determining safe levels for human exposure.
Collapse
Affiliation(s)
- Jason A Funk
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Michael G Janech
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Joshua C Dillon
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - John J Bissler
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
| | - Brian J Siroky
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
6
|
Costa LG, Giordano G, Faustman EM. Domoic acid as a developmental neurotoxin. Neurotoxicology 2010; 31:409-23. [PMID: 20471419 PMCID: PMC2934754 DOI: 10.1016/j.neuro.2010.05.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 05/04/2010] [Accepted: 05/05/2010] [Indexed: 11/21/2022]
Abstract
Domoic acid (DomA) is an excitatory amino acid which can accumulate in shellfish and finfish under certain environmental conditions. DomA is a potent neurotoxin. In humans and in non-human primates, oral exposure to a few mg/kg DomA elicits gastrointestinal effects, while slightly higher doses cause neurological symptoms, seizures, memory impairment, and limbic system degeneration. In rodents, which appear to be less sensitive than humans or non-human primates, oral doses cause behavioral abnormalities (e.g. hindlimb scratching), followed by seizures and hippocampal degeneration. Similar effects are also seen in other species (from sea lions to zebrafish), indicating that DomA exerts similar neurotoxic effects across species. The neurotoxicity of DomA is ascribed to its ability to interact and activate the AMPA/KA receptors, a subfamily of receptors for the neuroexcitatory neurotransmitter glutamate. Studies exploring the neurotoxic effects of DomA on the developing nervous system indicate that DomA elicits similar behavioral, biochemical and morphological effects as in adult animals. However, most importantly, developmental neurotoxicity is seen at doses of DomA that are one to two orders of magnitude lower than those exerting neurotoxicity in adults. This difference may be due to toxicokinetic and/or toxicodynamic differences. Estimated safe doses may be exceeded in adults by high consumption of shellfish contaminated with DomA at the current limit of 20 microg/g. Given the potential higher susceptibility of the young to DomA neurotoxicity, additional studies investigating exposure to, and effects of this neurotoxin during brain development are warranted.
Collapse
Affiliation(s)
- Lucio G Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA.
| | | | | |
Collapse
|
7
|
Jeffery B, Barlow T, Moizer K, Paul S, Boyle C. Amnesic shellfish poison. Food Chem Toxicol 2004; 42:545-57. [PMID: 15019178 DOI: 10.1016/j.fct.2003.11.010] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2002] [Accepted: 11/07/2003] [Indexed: 11/22/2022]
Abstract
Amnesic shellfish poisoning (ASP) is caused by consumption of shellfish that have accumulated domoic acid, a neurotoxin produced by some strains of phytoplankton. The neurotoxic properties of domoic acid result in neuronal degeneration and necrosis in specific regions of the hippocampus. A serious outbreak of ASP occurred in Canada in 1987 and involved 150 reported cases, 19 hospitalisations and 4 deaths after consumption of contaminated mussels. Symptoms ranged from gastrointestinal disturbances, to neurotoxic effects such as hallucinations, memory loss and coma. Monitoring programmes are in place in numerous countries worldwide and closures of shellfish harvesting areas occur when domoic acid concentrations exceed regulatory limits. This paper reviews the chemistry, sources, metabolism and toxicology of domoic acid as well as human case reports of ASP and discusses a possible mechanism of toxicity.
Collapse
Affiliation(s)
- B Jeffery
- Food Standards Agency, Aviation House, 125 Kingsway, London WC2B 6NH, UK.
| | | | | | | | | |
Collapse
|
8
|
Scallet AC, Kowalke PK, Rountree RL, Thorn BT, Binienda ZK. Electroencephalographic, behavioral, and c-fos responses to acute domoic acid exposure. Neurotoxicol Teratol 2004; 26:331-42. [PMID: 15019966 DOI: 10.1016/j.ntt.2003.10.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2003] [Revised: 08/12/2003] [Accepted: 10/03/2003] [Indexed: 11/17/2022]
Abstract
Domoic acid, a potent excitotoxic analogue of glutamate and kainate, may cause seizures, amnesia, and sometimes death in humans consuming contaminated shellfish. Continuous behavioral observations and recordings of the electrocorticogram (ECoG, via bipolar, epidural electrodes) were obtained from nonanesthetized rats for 2 h after intraperitoneal injection with either saline, 2.2, or 4.4 mg/kg of domoic acid. Rats were then sacrificed for c-fos immunohistochemistry. Fast Fourier transformation (FFT) of the ECoG data to obtain the voltage as a function of frequency indicated that the lower frequency bands (theta, 4.75-6.75 Hz and delta, 1.25-4.50 Hz) were the first to respond, with a significant elevation by 30 min after the high dose of domoic acid. The lower dose of domoic acid also caused a significant elevation of ECoG voltage, but not until later in the session. Sixty minutes after dosing, the behavioral biomarkers of "ear scratching" and "rearing, praying" (RP) seizures became significantly elevated in the high-dose rats. The low-dose rats showed no significant alterations in behavior at any time during the session. In postmortem brains obtained immediately after the sessions, c-fos was activated in the anterior olfactory nucleus by both the low and high doses of domoic acid. However, only the high dose increased c-fos immunoreactivity in the hippocampus, affecting both the granule and pyramidal neurons. These data indicate that electroencephalographic and c-fos responses can be obtained at a dose of domoic acid that fails to activate the behavioral response most commonly used as a bioassay for this marine toxin: ear scratching with the ipsilateral foot.
Collapse
Affiliation(s)
- Andrew C Scallet
- Division of Neurotoxicology, National Center for Toxicological Research, USFDA, 3900 NCTR Drive, Jefferson, AR 72079, USA.
| | | | | | | | | |
Collapse
|
9
|
Binienda ZK, Scallet AC, Schmued LC, Ali SF. Ibogaine neurotoxicity assessment: electrophysiological, neurochemical, and neurohistological methods. THE ALKALOIDS. CHEMISTRY AND BIOLOGY 2002; 56:193-210. [PMID: 11705108 DOI: 10.1016/s0099-9598(01)56014-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Affiliation(s)
- Z K Binienda
- Division of Neurotoxicology, FDA/National Center for Toxicological Research, Jefferson, AR 72079-9502, USA
| | | | | | | |
Collapse
|
10
|
Mayer AM. The marine toxin domoic acid may affect the developing brain by activation of neonatal brain microglia and subsequent neurotoxic mediator generation. Med Hypotheses 2000; 54:837-41. [PMID: 10859697 DOI: 10.1054/mehy.1999.0962] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Amnesic shellfish poisoning, one of the shellfish poisoning syndromes, is caused by the marine diatom toxin domoic acid (DOM). While in adult rats, mice, monkeys and humans DOM poorly penetrates the blood-brain barrier, DOM has been shown to be very toxic to fetal in newborn mice, because the blood-brain barrier is incomplete during neurodevelopment. This fact may explain why neonates show a higher sensitivity to neurotoxins like DOM as compared to adult animals. Mechanistic studies on DOM's neurotoxicity have mainly concentrated on the investigation of DOM's effect on neuronal tissue. Recent studies have shown that glia is also involved in DOM's neurotoxicity to the adult as well as the developing nervous system. The scientific literature strongly supports the hypothesis that the microglia may play a critical role in mediating DOM's neurotoxic effects. However, the effect of DOM on microglia has not been systematically investigated. The literature supporting our hypothesis is presented and discussed.
Collapse
Affiliation(s)
- A M Mayer
- Department of Pharmacology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, USA.
| |
Collapse
|
11
|
Vreugdenhil E, Datson N, Engels B, de Jong J, van Koningsbruggen S, Schaaf M, de Kloet ER. Kainate-elicited seizures induce mRNA encoding a CaMK-related peptide: A putative modulator of kinase activity in rat hippocampus. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1097-4695(199904)39:1<41::aid-neu4>3.0.co;2-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Nagatomo I, Akasaki Y, Uchida M, Tominaga M, Kuchiiwa S, Nakagawa S, Takigawa M. Kainic and domoic acids differentially affect NADPH-diaphorase neurons in the mouse hippocampal formation. Brain Res Bull 1999; 48:277-82. [PMID: 10229334 DOI: 10.1016/s0361-9230(98)00171-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigated changes in numbers of nitric-oxide-producing cells in the hippocampal formation, striatum, and temporal cortex of mice 24 h after intraperitoneal administration of kainic acid (5, 10, 15, and 20 mg/kg) or domoic acid (1, 2, and 4 mg/kg). Nitric-oxide-producing cells were demonstrated histochemically by staining for nicotinamide adenine dinucleotide phosphate diaphorase. Nicotinamide adenine dinucleotide phosphate-diaphorase-positive neurons in the dentate gyrus and the subiculum did not change in number following administration of kainic acid or domoic acid at any dose. Positive neurons in the CA3 region of mice treated with kainic acid or domoic acid at any dose were significantly fewer than in controls. Although the numbers of positive neurons in the CA1/CA2 regions did not differ from those of controls at any of the four doses of kainic acid, positive cells in the CA1/CA2 were significantly more numerous than in controls at any dose of domoic acid. Although no significant differences in the numbers of positive neurons in the striatum were apparent between controls and any of the four doses of kainic acid, domoic acid significantly decreased the numbers of such cells. These results suggest that systemically administered kainic acid and domoic acid affect differentially nitric-oxide-producing cells in the hippocampal formation.
Collapse
Affiliation(s)
- I Nagatomo
- Department of Neuropsychiatry, Faculty of Medicine, Kagoshima University, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Kumar V, Goguen DM, Guido ME, Rusak B. Melatonin does not influence the expression of c-fos in the suprachiasmatic nucleus of rats and hamsters. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1997; 52:242-8. [PMID: 9495545 DOI: 10.1016/s0169-328x(97)00260-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have assessed whether melatonin can induce c-fos expression at various circadian phases, and whether melatonin can inhibit photically induced c-fos expression in the suprachiasmatic nucleus (SCN) in both rats and Syrian hamsters. Subcutaneous administration of melatonin at a dose of 100 microg/kg neither induced expression of Fos, the protein product of the c-fos proto-oncogene, nor inhibited the expression of Fos-like immunoreactivity (Fos-lir) induced by a light pulse in the SCN of rats and hamsters. In situ hybridization studies also demonstrated the absence of induction by acute melatonin treatments of c-fos mRNA in the SCN. Taken together, these results demonstrate that melatonin effects on SCN cells involve signal transduction pathways that do not include regulation of c-fos gene expression.
Collapse
Affiliation(s)
- V Kumar
- Department of Psychology, Dalhousie University, Halifax, NS, Canada
| | | | | | | |
Collapse
|
14
|
Scallet AC, Ye X. Excitotoxic mechanisms of neurodegeneration in transmissible spongiform encephalopathies. Ann N Y Acad Sci 1997; 825:194-205. [PMID: 9369987 DOI: 10.1111/j.1749-6632.1997.tb48429.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Endogenous excitatory amino acids (EAAs) such as glutamic or aspartic acids have been proposed to mediate the brain damage to EAA receptor-rich brain sites that is caused by a variety of external toxic agents (glutamic acid, domoic acid, kainic acid, ibogaine, trimethyltin (TMT), 3-nitropropionic acid (3-NPA)), as well as from such naturally-occurring age-related neurodegenerative diseases as Alzheimer's disease, Huntington's chorea, and Parkinson's disease. Sites often damaged include the hypothalamus (glutamate), the hippocampal and neocortical pyramidal neurons (domoic acid), the cerebellar Purkinje neurons (ibogaine) and the corpus striatum (3-NPA, amphetamine). The excitotoxic damage occurs to neuronal cell bodies and their dendrites, resulting in a characteristics appearance of pyknotic neurons surrounded by their vacuolated, swollen dendrites. Axons passing through the region that lack EAA receptors are completely spared. However, astrocytes with swollen perikarya and nuclei (Alzheimer's type II "reactive" astrocytes) are often observed in the vicinity of the lesions. Animal and human "Prion Diseases" or "Transmissible Spongiform Encephalopathies" (TSEs) result (after a period of months to years) in a neurodegenerative picture characterized by pyknotic neurons surrounded by vacuoles with numerous reactive astrocytes in the vicinity of the damage. In addition, amyloid deposits composed of a protease-resistant protein (PrPSc) characteristic of the particular host species with the disease are found near the degenerating neurons. By using different strains of the scrapies TSE agent to inoculate hamsters and mice, reproducible models of hypothalamic, hippocampal, or cerebellar damage resulting in the appropriate functional deficits may be obtained. Because of the close similarity in the appearance, localization, and functional consequences from TSE neuropathology compared to some of the well-known EAA syndromes, we propose that excitotoxic mechanisms may play a role in the pathogenesis of TSE neurodegenerative diseases. The similarity in pathogenesis of the neurodegenerative processes in excitotoxicity compared to TSE diseases also implies that neuroprotective strategies against excitotoxicity may also be effective against TSEs.
Collapse
Affiliation(s)
- A C Scallet
- Division of Neurotoxicology, Food and Drug Administration, Jefferson, Arkansas 72079-9502, USA.
| | | |
Collapse
|
15
|
Appel NM, Rapoport SI, O'Callaghan JP, Bell JM, Freed LM. Sequelae of parenteral domoic acid administration in rats: comparison of effects on different metabolic markers in brain. Brain Res 1997; 754:55-64. [PMID: 9134959 DOI: 10.1016/s0006-8993(97)00042-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Parenterally administered domoic acid, a structural analog of the excitatory amino acids glutamic acid and kainic acid, has specific effects on brain histology in rats, as measured using different anatomic markers. Domoic acid-induced convulsions affects limbic structures such as hippocampus and entorhinal cortex, and different anatomic markers can detect these neurotoxic effects to varying degrees. Here we report effects of domoic acid administration on quantitative indicators of brain metabolism and gliosis. Domoic acid, 2.25 mg/kg i.p., caused stereotyped behavior and convulsions in approximately 60% of rats which received it. Six to eight days after domoic acid or vehicle administration, the animals were processed to measure regional brain incorporation of the long-chain fatty acids [1-(14)C]arachidonic acid ([14C]AA) and [9,10-(3)H]palmitic acid ([3H]PA), or regional cerebral glucose utilization (rCMRglc) using 2-[1-(14)C]deoxy-D-glucose, by quantitative autoradiography. Others rats were processed to measure brain glial fibrillary acidic protein (GFAP) by enzyme-linked immunosorbent assay. Domoic acid increased GFAP in the anterior portion of cerebral cortex, the caudate putamen and thalamus compared with vehicle. However, in rats that convulsed after domoic acid GFAP was significantly increased throughout the cerebral cortex, as well as in the hippocampus, septum, caudate putamen, and thalamus. Domoic acid, in the absence of convulsions, decreased relative [14C]AA incorporation in the claustrum and pyramidal cell layer of the hippocampus compared with vehicle-injected controls. In the presence of convulsions, relative [14C]AA incorporation was decreased in hippocampus regions CA1 and CA2. Uptake of [3H]PA into brain was unaffected. Relative rCMRglc decreased in entorhinal cortex following domoic acid administration with or without convulsions. These results suggest that acute domoic acid exposure affects discrete brain circuits by inducing convulsions, and that domoic acid-induced convulsions cause chronic effects on brain function that are reflected in altered fatty acid metabolism and gliosis.
Collapse
Affiliation(s)
- N M Appel
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Laurel, MD 20708-2476, USA.
| | | | | | | | | |
Collapse
|
16
|
Appel NM, Rapoport SI, O'Callaghan JP. Sequelae of parenteral domoic acid administration in rats: comparison of effects on different anatomical markers in brain. Synapse 1997; 25:350-8. [PMID: 9097394 DOI: 10.1002/(sici)1098-2396(199704)25:4<350::aid-syn6>3.0.co;2-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Brain damage following administration of domoic acid, a structural analog of the excitatory amino acids glutamic acid and kainic acid, was compared using different anatomic markers in adult rats. Seven days after administration of domoic acid (2.25 mg/kg i.p.) or vehicle, brains were collected and sectioned and stained to visualize Nissl substance using thionin, argyrophilia using a cupric silver staining method, astroglia using immunohistochemistry to detect glial fibrillary acidic protein-like immunoreactivity (GFAP-ir), and activated microglia using lectin histochemistry to detect Griffonia simplicifolia I-B4 isolectin (GSI-B4) binding in adjacent sections. In approximately 60% of rats to which it was administered, domoic acid caused stereotyped behavior within 60 min, followed by convulsions within 2-3 h. Brains of domoic acid-administered rats that did not manifest stereotyped behavior or convulsions did not differ from brains from vehicle-administered controls. In animals that had manifested stereotyped behavior and convulsions, Nissl staining was mostly unremarkable in brain sections. In contrast, there was intense argyrophilia in anterior olfactory nucleus, CA1 hippocampus, lateral septum, parietal (layer IV), piriform, and entorhinal cortices, ventral posterolateral thalamus, and amygdala. This pattern was reminiscent of that seen in postmortem specimens from humans who consumed domoic acid-tainted mussels and in experimental animals after kainic acid administration. Adjacent sections displayed astrogliosis, evidenced by increased GFAP-ir, which was more diffuse than the argyrophilic reaction. Activated microglia were revealed using GSI-B4 histochemistry. These data suggest activation of discrete brain circuits in rats that convulse following domoic acid administration and subsequent pathological alterations. The data strongly suggest that neuropathology following domoic acid occurs only in animals manifesting domoic acid-induced sterotypy and convulsions. The data do not rule out more insidious damage in behaviorally normal rats that receive domoic acid.
Collapse
Affiliation(s)
- N M Appel
- Division of Applied Pharmacology Research, Food and Drug Administration, Laurel, Maryland 20708, USA.
| | | | | |
Collapse
|
17
|
Guido ME, Rusak B, Robertson HA. Expression of fosB mRNA in the hamster suprachiasmatic nucleus is induced at only selected circadian phases. Brain Res 1996; 739:132-8. [PMID: 8955933 DOI: 10.1016/s0006-8993(96)00816-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We have studied the expression of fosB mRNA in the suprachiasmatic nucleus (SCN) of hamsters by in situ hybridization using oligonucleotides with sequences complementary to the C-terminal of the fosB mRNA sequence. In animals exposed for 48 h to darkness, there was little or no background expression in SCN cells of fosB mRNA at any circadian phase. Light pulses (30 min) were able to induce fosB expression only during the subjective night. Transcripts of fosB increased rapidly to peak by the end of a 30-min light pulse. Light-induced increases gradually declined in darkness, but levels were still elevated for up to 150 min after the light pulse. Induction in response to a light pulse was largely restricted to the ventrolateral portion of the nucleus which receives the heaviest retinal projection. The temporal and anatomical pattern of fosB mRNA expression in the hamster SCN therefore resembles that reported previously for other immediate-early genes, such as c-fos.
Collapse
Affiliation(s)
- M E Guido
- Department of Psychology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | |
Collapse
|
18
|
Guido ME, Rusak B, Robertson HA. Spontaneous circadian and light-induced expression of junB mRNA in the hamster suprachiasmatic nucleus. Brain Res 1996; 732:215-22. [PMID: 8891287 DOI: 10.1016/0006-8993(96)00526-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We examined both spontaneous and light-evoked expression of junB mRNA in the hamster suprachiasmatic nucleus (SCN), an endogenous circadian pacemaker. junB was expressed in the SCN in response to a light pulse during the subjective night and early subjective day as well as spontaneously during the transition from subjective night to subjective day. Light-evoked expression was strongest in the ventral SCN, while spontaneous expression was stronger in the dorsal SCN. Spontaneous expression began around subjective dawn and persisted for at least 4 h into the subjective day. The expression of junB mRNA may play a role in both phase-shifting responses to light and in spontaneous oscillation of the SCN pacemaker.
Collapse
Affiliation(s)
- M E Guido
- Department of Psychology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | | | |
Collapse
|
19
|
Scallet AC, Binienda Z, Caputo FA, Hall S, Paule MG, Rountree RL, Schmued L, Sobotka T, Slikker W. Domoic acid-treated cynomolgus monkeys (M. fascicularis): effects of dose on hippocampal neuronal and terminal degeneration. Brain Res 1993; 627:307-13. [PMID: 8298975 DOI: 10.1016/0006-8993(93)90335-k] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Domoic acid is a tricarboxylic amino acid (structurally related to kainic acid and glutamic acid) that is found in the environment as a contaminant of some seafood. To determine the nature of any neurological damage caused by domoate, as well as the minimum neurotoxic dose, juvenile and adult monkeys were dosed intravenously with domoate at one of a range of doses from 0.25 to 4 mg/kg. When animals were perfused one week later, histochemical staining using a silver method to reveal degenerating axons and cell bodies showed two distinct types of hippocampal lesions. One lesion, termed 'Type A', was a small focal area of silver grains restricted to CA2 stratum lucidum, the site of greatest kainic acid receptor concentration in the brain. Type A lesions occurred over a dose range of 0.5 to 2.0 mg/kg in juvenile animals and 0.5 to 1.0 mg/kg in adult animals. No mortality occurred in any of the juvenile monkeys, but one juvenile animal that received 4.0 mg/kg sustained a second type of lesion, termed 'Type B', characterized by widespread damage to pyramidal neurons and axon terminals of CA4, CA3, CA2, CA1, and subiculum subfields of the hippocampus. Doses of more than 1.0 mg/kg in the adult monkeys either proved lethal or resulted in Type B lesions. Induction of c-fos protein had occurred in the hippocampal dentate gyrus and CA1 regions of moribund animals perfused within hours of their initial dose.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- A C Scallet
- Division of Neurotoxicology, FDA/National Center for Toxicological Research, Jefferson, AR 72079
| | | | | | | | | | | | | | | | | |
Collapse
|