Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 165(3): 379-390, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36815399

RESUMEN

Dietary lipids, particularly omega-3 polyunsaturated fatty acids, are speculated to impact behaviors linked to the dopaminergic system, such as movement and control of circadian rhythms. However, the ability to draw a direct link between dopaminergic omega-3 fatty acid metabolism and behavioral outcomes has been limited to the use of diet-based approaches, which are confounded by systemic effects. Here, neuronal lipid metabolism was targeted in a diet-independent manner by manipulation of long-chain acyl-CoA synthetase 6 (ACSL6) expression. ACSL6 performs the initial reaction for cellular fatty acid metabolism and prefers the omega-3 polyunsaturated fatty acid, docosahexaenoic acid (DHA). The loss of Acsl6 in mice (Acsl6-/- ) depletes neuronal membranes of DHA content and results in phenotypes linked to dopaminergic control, such as hyperlocomotion, impaired short-term spatial memory, and imbalances in dopamine neurochemistry. To investigate the role of dopaminergic ACSL6 on these outcomes, a dopaminergic neuron-specific ACSL6 knockout mouse was generated (Acsl6DA-/- ). Acsl6DA-/- mice demonstrated hyperlocomotion and imbalances in striatal dopamine neurochemistry. Circadian rhythms of both the Acsl6-/- and the Acsl6DA-/- mice were similar to control mice under basal conditions. However, upon light entrainment, a mimetic of jet lag, both the complete knockout of ACSL6 and the dopaminergic-neuron-specific loss of ACSL6 resulted in a longer recovery to entrainment compared to control mice. In conclusion, these data demonstrate that ACSL6 in dopaminergic neurons alters dopamine metabolism and regulation of light entrainment suggesting that DHA metabolism mediated by ACSL6 plays a role in dopamine neuron biology.


Asunto(s)
Neuronas Dopaminérgicas , Metabolismo de los Lípidos , Ratones , Animales , Neuronas Dopaminérgicas/metabolismo , Dopamina , Grasas de la Dieta , Dieta , Ratones Noqueados , Ácidos Docosahexaenoicos/metabolismo , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo
2.
Chem Res Toxicol ; 36(8): 1361-1373, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37421305

RESUMEN

Animal fat and iron-rich diets are risk factors for Parkinson's disease (PD). The heterocyclic aromatic amines (HAAs) harman and norharman are neurotoxicants formed in many foods and beverages, including cooked meats, suggesting a role for red meat in PD. The structurally related carcinogenic HAAs 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), 2-amino-3,8-dimethylmidazo[4,5-f]quinoxaline (MeIQx), and 2-amino-9H-pyrido[2,3-b]indole (AαC) also form in cooked meats. We investigated the cytotoxicity, DNA-damaging potential, and mitochondrial damage of HAAs and their genotoxic HONH-HAA metabolites in galactose-dependent SH-SY5Y cells, a human neuroblastoma cell line relevant for PD-related neurotoxicity. All HAAs and HONH-HAAs induced weak toxicity except HONH-PhIP, which was 1000-fold more potent than the other chemicals. HONH-PhIP DNA adduct formation occurred at 300-fold higher levels than adducts formed with HONH-MeIQx and HONH-AαC, assuming similar cellular uptake rates. PhIP-DNA adduct levels occurred at concentrations as low as 1 nM and were threefold or higher and more persistent in mitochondrial DNA than nuclear DNA. N-Acetyltransferases (NATs), sulfotransferases, and kinases catalyzed PhIP-DNA binding and converted HONH-PhIP to highly reactive ester intermediates. DNA binding assays with cytosolic, mitochondrial, and nuclear fractions of SH-SY5Y fortified with cofactors revealed that cytosolic AcCoA-dependent enzymes, including NAT1, mainly carried out HONH-PhIP bioactivation to form N-acetoxy-PhIP, which binds to DNA. Furthermore, HONH-PHIP and N-acetoxy-PhIP inhibited mitochondrial complex-I, -II, and -III activities in isolated SH-SY5Y mitochondria. Mitochondrial respiratory chain complex dysfunction and DNA damage are major mechanisms in PD pathogenesis. Our data support the possible role of PhIP in PD etiology.


Asunto(s)
Carcinógenos , Neuroblastoma , Animales , Humanos , Carcinógenos/metabolismo , Piridinas , Daño del ADN , Aminas/metabolismo , Carne/análisis
3.
Chem Res Toxicol ; 35(1): 59-72, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34990108

RESUMEN

Heterocyclic aromatic amines (HAAs) are mainly formed in the pyrolysis process during high-temperature cooking of meat. Meat consumption is very typical of the western diet, and the amount of meat consumption in the eastern countries is growing rapidly; HAAs represents widespread exposure. HAAs are classified as possible human carcinogens; numerous epidemiological studies have demonstrated regular consumption of meat with HAAs as risk factor for cancers. Specific HAAs have received major attention. For example, 2-amino-1-methyl-6-phenylimidazo[4,5-b] pyridine has been extensively studied as a genotoxicant and mutagen, with emergent literature on neurotoxicity. Harmane has been extensively studied for a role in essential tremors and potentially Parkinson's disease (PD). Harmane levels have been demonstrated to be elevated in blood and brain in essential tremor patients. Meat consumption has been implicated in the etiology of neurodegenerative diseases; however, the role of toxicants formed during meat preparation has not been studied. Epidemiological studies are currently examining the association between HAAs and risk of neurodegenerative diseases such as essential tremors and PD. Studies from our laboratory and others have provided strong evidence that HAA exposure produces PD and Alzheimer's disease-relevant neurotoxicity in cellular and animal models. In this review, we summarize and critically evaluate previous studies on HAA-induced neurotoxicity and the molecular basis of potential neurotoxic effects of HAAs. The available studies provide strong support for the premise that HAAs may impact neurological function and that addressing gaps in understanding of adverse neurological outcomes is critical to determine whether these compounds are modifiable risk factors.


Asunto(s)
Aminas/efectos adversos , Compuestos Heterocíclicos/efectos adversos , Enfermedades Neurodegenerativas/inducido químicamente , Animales , Humanos
4.
Chem Res Toxicol ; 35(8): 1312-1333, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35921496

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are a group of persistent environmental pollutants that are ubiquitously found in the environment and virtually in all living organisms, including humans. PFAS cross the blood-brain barrier and accumulate in the brain. Thus, PFAS are a likely risk for neurotoxicity. Studies that measured PFAS levels in the brains of humans, polar bears, and rats have demonstrated that some areas of the brain accumulate greater amounts of PFAS. Moreover, in humans, there is evidence that PFAS exposure is associated with attention-deficit/hyperactivity disorder (ADHD) in children and an increased cause of death from Parkinson's disease and Alzheimer's disease in elderly populations. Given possible links to neurological disease, critical analyses of possible mechanisms of neurotoxic action are necessary to advance the field. This paper critically reviews studies that investigated potential mechanistic causes for neurotoxicity including (1) a change in neurotransmitter levels, (2) dysfunction of synaptic calcium homeostasis, and (3) alteration of synaptic and neuronal protein expression and function. We found growing evidence that PFAS exposure causes neurotoxicity through the disruption of neurotransmission, particularly the dopamine and glutamate systems, which are implicated in age-related psychiatric illnesses and neurodegenerative diseases. Evaluated research has shown there are highly reproduced increased glutamate levels in the hippocampus and catecholamine levels in the hypothalamus and decreased dopamine in the whole brain after PFAS exposure. There are significant gaps in the literature relative to the assessment of the nigrostriatal system (striatum and ventral midbrain) among other regions associated with PFAS-associated neurologic dysfunction observed in humans. In conclusion, evidence suggests that PFAS may be neurotoxic and associated with chronic and age-related psychiatric illnesses and neurodegenerative diseases. Thus, it is imperative that future mechanistic studies assess the impact of PFAS and PFAS mixtures on the mechanism of neurotransmission and the consequential functional effects.


Asunto(s)
Contaminantes Ambientales , Fluorocarburos , Síndromes de Neurotoxicidad , Anciano , Animales , Niño , Dopamina/metabolismo , Contaminantes Ambientales/metabolismo , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Glutamatos , Humanos , Síndromes de Neurotoxicidad/metabolismo , Ratas , Transmisión Sináptica
5.
J Biochem Mol Toxicol ; 35(4): e22694, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33393683

RESUMEN

Alzheimer's disease (AD) is a major public health crisis due to devastating cognitive symptoms, a lack of curative treatments, and increasing prevalence. Most cases are sporadic (>95% of cases) after the age of 65 years, implicating an important role of environmental factors in disease pathogenesis. Environmental neurotoxicants have been implicated in neurodegenerative disorders including Parkinson's Disease and AD. Animal models of AD and in vitro studies have shed light on potential neuropathological mechanisms, yet the biochemical and molecular underpinnings of AD-relevant environmental neurotoxicity remain poorly understood. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is a potentially critical pathogenic target of environmentally induced neurotoxicity. BACE1 clearly has a critical role in AD pathophysiology: It is required for amyloid beta production and expression and activity of BACE1 are increased in the AD brain. Though the literature on BACE1 in response to environmental insults is limited, current studies, along with extensive AD neurobiology literature suggest that BACE1 deserves attention as an important neurotoxic target. Here, we critically review research on environmental neurotoxicants such as metals, pesticides, herbicides, fungicides, polyfluoroalkyl substances, heterocyclic aromatic amines, advanced glycation end products, and acrolein that modulate BACE1 and potential mechanisms of action. Though more research is needed to clearly understand whether BACE1 is a critical mediator of AD-relevant neurotoxicity, available reports provide convincing evidence that BACE1 is altered by environmental risk factors associated with AD pathology, implying that BACE1 inhibition and its use as a biomarker should be considered in AD management and research.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Ácido Aspártico Endopeptidasas/biosíntesis , Encéfalo/enzimología , Exposición a Riesgos Ambientales/efectos adversos , Regulación Enzimológica de la Expresión Génica , Síndromes de Neurotoxicidad , Neurotoxinas/toxicidad , Anciano , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Síndromes de Neurotoxicidad/enzimología , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/patología
6.
Proc Natl Acad Sci U S A ; 115(49): 12525-12530, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30401738

RESUMEN

Docosahexaenoic acid (DHA) is an omega-3 fatty acid that is highly abundant in the brain and confers protection against numerous neurological diseases, yet the fundamental mechanisms regulating the enrichment of DHA in the brain remain unknown. Here, we have discovered that a member of the long-chain acyl-CoA synthetase family, Acsl6, is required for the enrichment of DHA in the brain by generating an Acsl6-deficient mouse (Acsl6-/-). Acsl6 is highly enriched in the brain and lipid profiling of Acsl6-/- tissues reveals consistent reductions in DHA-containing lipids in tissues highly abundant with Acsl6. Acsl6-/- mice demonstrate motor impairments, altered glutamate metabolism, and increased astrogliosis and microglia activation. In response to a neuroinflammatory lipopolysaccharide injection, Acsl6-/- brains show similar increases in molecular and pathological indices of astrogliosis compared with controls. These data demonstrate that Acsl6 is a key mediator of neuroprotective DHA enrichment in the brain.


Asunto(s)
Encéfalo/enzimología , Coenzima A Ligasas/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Animales , Encéfalo/metabolismo , Coenzima A Ligasas/genética , Regulación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Microglía , Actividad Motora
7.
Toxicol Appl Pharmacol ; 377: 114623, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31195004

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are synthetic compounds that are a major public health concern due to widespread use, long environmental and biological half-lives, detection in most human plasma samples, and links to multiple adverse health outcomes. The literature suggests that some PFAS may be neurotoxic. However, there are major gaps in the literature with respect to how environmentally-relevant doses during development may influence the nervous system. To address this gap, we utilized a sentinel species, Northern leopard frogs (Lithobates pipiens) to determine the effects of developmental exposure to environmentally relevant perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) on major neurotransmitter systems. Frog larvae at Gosner stage 25 were exposed to 10, 100, or 1000 ppb PFOS or PFOA for 30 days before neurochemical analysis. High performance liquid chromatography (HPLC) with electrochemical detection or fluorescent detection assays was used to measure neurotransmitter levels, which were normalized to protein levels in each sample. Dopamine (DA) decreased significantly in the brains of frogs treated with PFOA (1000 ppb) and PFOS (100 and 1000 ppb). Significant increases in DA turnover also resulted from PFOA and PFOS treatment. Neither PFOS, nor PFOA produced detectable alterations in serotonin (nor its metabolite), norepinephrine, gamma-amino butyric acid (GABA), glutamate, or acetylcholine. PFAS body burdens showed that PFOS accumulated relative to dose, while PFOA did not. These data suggest that DArgic neurotransmission is selectively affected in developmentally exposed amphibians and that PFAS should be evaluated for a potential role in diseases that target the DA system.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Química Encefálica/efectos de los fármacos , Caprilatos/toxicidad , Dopamina/metabolismo , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Síndromes de Neurotoxicidad/metabolismo , Rana pipiens , Animales , Carga Corporal (Radioterapia) , Relación Dosis-Respuesta a Droga , Femenino , Larva , Neurotransmisores/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Transmisión Sináptica/efectos de los fármacos
8.
Neurobiol Dis ; 95: 238-49, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27452482

RESUMEN

Extensive convergent evidence collectively suggests that mitochondrial dysfunction is central to the pathogenesis of Parkinson's disease (PD). Recently, changes in the dynamic properties of mitochondria have been increasingly implicated as a key proximate mechanism underlying neurodegeneration. However, studies have been limited by the lack of a model in which mitochondria can be imaged directly and dynamically in dopaminergic neurons of the intact vertebrate CNS. We generated transgenic zebrafish in which mitochondria of dopaminergic neurons are labeled with a fluorescent reporter, and optimized methods allowing direct intravital imaging of CNS dopaminergic axons and measurement of mitochondrial transport in vivo. The proportion of mitochondria undergoing axonal transport in dopaminergic neurons decreased overall during development between 2days post-fertilization (dpf) and 5dpf, at which point the major period of growth and synaptogenesis of the relevant axonal projections is complete. Exposure to 0.5-1.0mM MPP(+) between 4 and 5dpf did not compromise zebrafish viability or cause detectable changes in the number or morphology of dopaminergic neurons, motor function or monoaminergic neurochemistry. However, 0.5mM MPP(+) caused a 300% increase in retrograde mitochondrial transport and a 30% decrease in anterograde transport. In contrast, exposure to higher concentrations of MPP(+) caused an overall reduction in mitochondrial transport. This is the first time mitochondrial transport has been observed directly in CNS dopaminergic neurons of a living vertebrate and quantified in a PD model in vivo. Our findings are compatible with a model in which damage at presynaptic dopaminergic terminals causes an early compensatory increase in retrograde transport of compromised mitochondria for degradation in the cell body. These data are important because manipulation of early pathogenic mechanisms might be a valid therapeutic approach to PD. The novel transgenic lines and methods we developed will be useful for future studies on mitochondrial dynamics in health and disease.


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Transporte Axonal/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Dinámicas Mitocondriales , Neuroimagen , Animales , Transporte Axonal/fisiología , Axones/patología , Muerte Celular/efectos de los fármacos , Sistema Nervioso Central/fisiopatología , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Intoxicación por MPTP/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Dinámicas Mitocondriales/fisiología , Enfermedad de Parkinson/metabolismo , Pez Cebra
9.
Neurobiol Dis ; 73: 356-65, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25447228

RESUMEN

Repeated seizures are often associated with development of refractory chronic epilepsy, the most common form of which is temporal lobe epilepsy. G-protein-coupled cannabinoid receptors (CB1 and CB2 receptors) regulate neuronal excitability and have been shown to mediate acute anticonvulsant effects of cannabinoids in animal models. However, the potential of cannabinoids to prevent chronic neuronal damage and development of epilepsy remains unexplored. We hypothesized that treatment with a CB receptor agonist after an episode of status epilepticus--but before development of spontaneous recurrent seizures--might prevent the development of functional changes that lead to chronic epilepsy. Using the rat pilocarpine model, a therapeutic approach was simulated by administering the CB agonist, WIN 55,212-2 after an episode of status epilepticus. Epileptic behavior was monitored during development of spontaneous recurrent seizures for up to 6 months. Histology, neurochemistry, redox status and NMDA receptor subunit expression were assessed at 6 months after pilocarpine-induced seizures. Sub-acute treatment with WIN 55,212-2 (for 15 days starting 24h after PILO injection) dramatically attenuated the severity, duration and frequency of spontaneous recurrent seizures. Further, in contrast to vehicle-treated animals, hippocampi from WIN 55,212-2-treated animals showed: normal thiol redox state, normal NR2A and NR2B subunit expression, preservation of GABAergic neurons and prevention of abnormal proliferation of GABAergic progenitors. This study shows for the first time that, after a known inciting event, treatment with a compound targeting CB receptors has the potential to prevent the epileptogenic events that result in chronic epileptic damage.


Asunto(s)
Benzoxazinas/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Epilepsia/prevención & control , Hipocampo/metabolismo , Morfolinas/farmacología , Naftalenos/farmacología , Animales , Enfermedad Crónica/prevención & control , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico
10.
J Neurochem ; 129(2): 315-27, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24266758

RESUMEN

α6ß2* nicotinic acetylcholine receptors (nAChRs)s in the ventral tegmental area to nucleus accumbens (NAc) pathway are implicated in the response to nicotine, and recent work suggests these receptors play a role in the rewarding action of ethanol. Here, we studied mice expressing gain-of-function α6ß2* nAChRs (α6L9'S mice) that are hypersensitive to nicotine and endogenous acetylcholine. Evoked extracellular dopamine (DA) levels were enhanced in α6L9'S NAc slices compared to control, non-transgenic (non-Tg) slices. Extracellular DA levels in both non-Tg and α6L9'S slices were further enhanced in the presence of GBR12909, suggesting intact DA transporter function in both mouse strains. Ongoing α6ß2* nAChR activation by acetylcholine plays a role in enhancing DA levels, as α-conotoxin MII completely abolished evoked DA release in α6L9'S slices and decreased spontaneous DA release from striatal synaptosomes. In HPLC experiments, α6L9'S NAc tissue contained significantly more DA, 3,4-dihydroxyphenylacetic acid, and homovanillic acid compared to non-Tg NAc tissue. Serotonin (5-HT), 5-hydroxyindoleacetic acid, and norepinephrine (NE) were unchanged in α6L9'S compared to non-Tg tissue. Western blot analysis revealed increased tyrosine hydroxylase expression in α6L9'S NAc. Overall, these results show that enhanced α6ß2* nAChR activity in NAc can stimulate DA production and lead to increased extracellular DA levels.


Asunto(s)
Dopamina/metabolismo , Receptores Nicotínicos/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Dopamina/biosíntesis , Inhibidores de Captación de Dopamina/farmacología , Fenómenos Electrofisiológicos , Espacio Extracelular/metabolismo , Mecamilamina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neostriado/metabolismo , Neurotransmisores/metabolismo , Antagonistas Nicotínicos/farmacología , Núcleo Accumbens/metabolismo , Técnicas de Placa-Clamp , Piperazinas/farmacología , Reacción en Cadena de la Polimerasa , Receptores Nicotínicos/genética , Recompensa
11.
Neurotoxicology ; 103: 266-287, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38964509

RESUMEN

Parkinson's disease (PD) is a debilitating neurodegenerative disease affecting millions of individuals worldwide. Hallmark features of PD pathology are the formation of Lewy bodies in neuromelanin-containing dopaminergic (DAergic) neurons of the substantia nigra pars compacta (SNpc), and the subsequent irreversible death of these neurons. Although genetic risk factors have been identified, around 90 % of PD cases are sporadic and likely caused by environmental exposures and gene-environment interaction. Mechanistic studies have identified a variety of chemical PD risk factors. PD neuropathology occurs throughout the brain and peripheral nervous system, but it is the loss of DAergic neurons in the SNpc that produce many of the cardinal motor symptoms. Toxicology studies have found specifically the DAergic neuron population of the SNpc exhibit heightened sensitivity to highly variable chemical insults (both in terms of chemical structure and mechanism of neurotoxic action). Thus, it has become clear that the inherent neurobiology of nigral DAergic neurons likely underlies much of this neurotoxic response to broad insults. This review focuses on inherent neurobiology of nigral DAergic neurons and how such neurobiology impacts the primary mechanism of neurotoxicity. While interactions with a variety of other cell types are important in disease pathogenesis, understanding how inherent DAergic biology contributes to selective sensitivity and primary mechanisms of neurotoxicity is critical to advancing the field. Specifically, key biological features of DAergic neurons that increase neurotoxicant susceptibility.

12.
J Biol Chem ; 287(5): 2971-83, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22128150

RESUMEN

α-Synuclein is strongly implicated in the pathogenesis of Parkinson disease. However, the normal functions of synucleins and how these relate to disease pathogenesis are uncertain. We characterized endogenous zebrafish synucleins in order to develop tractable models to elucidate the physiological roles of synucleins in neurons in vivo. Three zebrafish genes, sncb, sncg1, and sncg2 (encoding ß-, γ1-, and γ2-synucleins respectively), show extensive phylogenetic conservation with respect to their human paralogues. A zebrafish α-synuclein orthologue was not found. Abundant 1.45-kb sncb and 2.7-kb sncg1 mRNAs were detected in the CNS from early development through adulthood and showed overlapping but distinct expression patterns. Both transcripts were detected in catecholaminergic neurons throughout the CNS. Zebrafish lacking ß-, γ1-, or both synucleins during early development showed normal CNS and body morphology but exhibited decreased spontaneous motor activity that resolved as gene expression recovered. Zebrafish lacking both ß- and γ1-synucleins were more severely hypokinetic than animals lacking one or the other synuclein and showed delayed differentiation of dopaminergic neurons and reduced dopamine levels. Phenotypic abnormalities resulting from loss of endogenous zebrafish synucleins were rescued by expression of human α-synuclein. These data demonstrate that synucleins have essential phylogenetically conserved neuronal functions that regulate dopamine homeostasis and spontaneous motor behavior. Zebrafish models will allow further elucidation of the molecular physiology and pathophysiology of synucleins in vivo.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Actividad Motora/fisiología , Proteínas de Pez Cebra/metabolismo , Sinucleína beta/metabolismo , gamma-Sinucleína/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular/fisiología , Dopamina/genética , Humanos , Inmovilización , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/genética , Sinucleína beta/genética
13.
Neurobiol Dis ; 57: 38-46, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22776331

RESUMEN

Interactions between genetic factors and environmental exposures are thought to be major contributors to the etiology of Parkinson's disease. While such interactions are poorly defined and incompletely understood, recent epidemiological studies have identified specific interactions of potential importance to human PD. In this review, the most current data on gene-environment interactions in PD from human studies are critically discussed. Animal models have also highlighted the importance of genetic susceptibility to toxicant exposure and data of potential relevance to human PD are discussed. Goals and needs for the future of the field are proposed.


Asunto(s)
Interacción Gen-Ambiente , Enfermedad de Parkinson/etiología , Animales , Modelos Animales de Enfermedad , Humanos , Enfermedad de Parkinson/genética
14.
Toxicol Sci ; 191(1): 163-178, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36269219

RESUMEN

Organophosphate (OP) pesticides are widely used in agriculture. While acute cholinergic toxicity has been extensively studied, chronic effects on other neurons are less understood. Here, we demonstrated that the OP pesticide chlorpyrifos (CPF) and its oxon metabolite are dopaminergic neurotoxicants in Caenorhabditis elegans. CPF treatment led to inhibition of mitochondrial complex II, II + III, and V in rat liver mitochondria, while CPF-oxon did not (complex II + III and IV inhibition observed only at high doses). While the effect on C. elegans cholinergic behavior was mostly reversible with toxicant washout, dopamine-associated deficits persisted, suggesting dopaminergic neurotoxicity was irreversible. CPF reduced the mitochondrial content in a dose-dependent manner and the fat modulatory genes cyp-35A2 and cyp-35A3 were found to have a key role in CPF neurotoxicity. These findings were consistent with in vitro effects of CPF and CPF-oxon on nuclear receptor signaling and fatty acid/steroid metabolism observed in ToxCast assays. Two-way hierarchical analysis revealed in vitro effects on estrogen receptor, pregnane X receptor, and peroxisome proliferator-activated receptor gamma pathways as well as neurotoxicity of CPF, malathion, and diazinon, whereas these effects were not detected in malaoxon and diazoxon. Taken together, our study suggests that mitochondrial toxicity and metabolic effects of CPF, but not CPF-oxon, have a key role of CPF neurotoxicity in the low-dose, chronic exposure. Further mechanistic studies are needed to examine mitochondria as a common target for all OP pesticide parent compounds, because this has important implications on cumulative pesticide risk assessment.


Asunto(s)
Cloropirifos , Insecticidas , Plaguicidas , Ratas , Animales , Cloropirifos/toxicidad , Cloropirifos/metabolismo , Inhibidores de la Colinesterasa/toxicidad , Dopamina , Caenorhabditis elegans/metabolismo , Insecticidas/toxicidad
15.
Food Chem Toxicol ; 174: 113685, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36813153

RESUMEN

Parkinson's Disease (PD) and Alcohol Use Disorder (AUD) are disorders that involve similar dopaminergic neurobiological pathways and dysregulations in motivation- and reward-related behaviors. This study explored whether exposure to a PD-related neurotoxicant, paraquat (PQ), alters binge-like alcohol drinking and striatal monoamines in mice selectively bred for high alcohol preference (HAP), and whether these effects are sex-dependent. Previous studies found female mice are less susceptible to PD-related toxicants compared to male mice. Mice were treated with PQ or vehicle over 3 weeks (10 mg/kg, i.p. once per week) and binge-like alcohol [20% (v/v)] drinking was assessed. Mice were euthanized and brains were microdissected for monoamine analyses by high performance liquid chromatography with electrochemical detection (HPLC-ECD). PQ-treated HAP male mice showed significantly decreased binge-like alcohol drinking and ventral striatal 3,4-Dihydroxyphenylacetic acid (DOPAC) levels compared to vehicle-treated HAP mice. These effects were absent in female HAP mice. These findings suggest that male HAP mice may be more susceptible than female mice to PQ's disruptive effects on binge-like alcohol drinking and associated monoamine neurochemistry and may be relevant for understanding neurodegenerative processes implicated in PD and AUD.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas , Enfermedad de Parkinson , Ratones , Animales , Masculino , Femenino , Paraquat , Ratones Endogámicos C57BL , Consumo de Bebidas Alcohólicas , Etanol
16.
Biomedicines ; 11(10)2023 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-37893165

RESUMEN

Many potential immune therapeutic targets are similarly affected in adult-onset neurodegenerative diseases, such as Alzheimer's (AD) disease, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and frontotemporal dementia (FTD), as well as in a seemingly distinct Niemann-Pick type C disease with primarily juvenile onset. This strongly argues for an overlap in pathogenic mechanisms. The commonly researched immune targets include various immune cell subsets, such as microglia, peripheral macrophages, and regulatory T cells (Tregs); the complement system; and other soluble factors. In this review, we compare these neurodegenerative diseases from a clinical point of view and highlight common pathways and mechanisms of protein aggregation, neurodegeneration, and/or neuroinflammation that could potentially lead to shared treatment strategies for overlapping immune dysfunctions in these diseases. These approaches include but are not limited to immunisation, complement cascade blockade, microbiome regulation, inhibition of signal transduction, Treg boosting, and stem cell transplantation.

17.
Biomedicines ; 11(6)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37371694

RESUMEN

Proteinopathy and neuroinflammation are two main hallmarks of neurodegenerative diseases. They also represent rare common events in an exceptionally broad landscape of genetic, environmental, neuropathologic, and clinical heterogeneity present in patients. Here, we aim to recount the emerging trends in amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD) spectrum disorder. Our review will predominantly focus on neuroinflammation and systemic immune imbalance in ALS and FTD, which have recently been highlighted as novel therapeutic targets. A common mechanism of most ALS and ~50% of FTD patients is dysregulation of TAR DNA-binding protein 43 (TDP-43), an RNA/DNA-binding protein, which becomes depleted from the nucleus and forms cytoplasmic aggregates in neurons and glia. This, in turn, via both gain and loss of function events, alters a variety of TDP-43-mediated cellular events. Experimental attempts to target TDP-43 aggregates or manipulate crosstalk in the context of inflammation will be discussed. Targeting inflammation, and the immune system in general, is of particular interest because of the high plasticity of immune cells compared to neurons.

18.
Front Toxicol ; 4: 826488, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35373186

RESUMEN

Neurological hazard assessment of industrial and pesticidal chemicals demands a substantial amount of time and resources. Caenorhabditis elegans is an established model organism in developmental biology and neuroscience. It presents an ideal test system with relatively fewer neurons (302 in hermaphrodites) versus higher-order species, a transparent body, short lifespan, making it easier to perform neurotoxic assessment in a time and cost-effective manner. Yet, no regulatory testing guidelines have been developed for C. elegans in the field of developmental and adult neurotoxicity. Here, we describe a set of morphological and behavioral assessment protocols to examine neurotoxicity in C. elegans with relevance to cholinergic and dopaminergic systems. We discuss the homology of human genes and associated proteins in these two signaling pathways and evaluate the morphological and behavioral endpoints of C. elegans in the context of published adverse outcome pathways of neurodegenerative diseases. We conclude that C. elegans neurotoxicity testing will not only be instrumental to eliminating mammalian testing in neurological hazard assessment but also lead to new knowledge and mechanistic validation in the adverse outcome pathway framework.

19.
J Neurosci Res ; 88(2): 420-7, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19681169

RESUMEN

Parkinson's disease (PD) is characterized pathologically by progressive neurodegeneration of the nigrostriatal dopamine (DA) system. Currently, the cause of the disease is unknown, except for a small percentage of familial cases (<10% of total). The rat rotenone model reproduces many of the pathological features of the human disease, including apomorphine-responsive behavioral deficits, DA depletion, loss of striatal DA terminals and nigral dopaminergic neurons, and alpha-synuclein/polyubiquitin-positive cytoplasmic inclusions reminiscent of Lewy bodies. Therefore, this model is well-suited to examine potential neuroprotective agents. Melatonin is produced mainly by the pineal gland and is known primarily for regulating circadian rhythms. It also has potent free radical scavenging and antiinflammatory properties. Melatonin has been reported to be neuroprotective in the 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) models of PD. However, there are conflicting reports suggesting that melatonin does not provide neuroprotection in these models. Melatonin elicits significant functional changes in the nigrostriatal DA system that may affect 6-OHDA and MPTP entry into cells. Therefore, rotenone is an ideal model for assessing protection, because it does not rely on the dopamine transporter uptake to exert neurotoxicity. In this study, the neuroprotective potential of melatonin in the rotenone PD model was assessed. Melatonin potentiated striatal catecholamine depletion, striatal terminal loss, and nigral DA cell loss. Indeed, melatonin alone elicited alterations in striatal catecholamine content. Our findings indicate that melatonin is not neuroprotective in the rotenone model of PD and may exacerbate neurodegeneration.


Asunto(s)
Melatonina/efectos adversos , Fármacos Neuroprotectores/efectos adversos , Trastornos Parkinsonianos/fisiopatología , Animales , Peso Corporal/efectos de los fármacos , Recuento de Células , Muerte Celular/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/fisiología , Trastornos Parkinsonianos/inducido químicamente , Distribución Aleatoria , Ratas , Ratas Endogámicas Lew , Rotenona , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología , Sustancia Negra/fisiopatología , Análisis de Supervivencia , Tirosina 3-Monooxigenasa/metabolismo
20.
Toxicology ; 437: 152436, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32169473

RESUMEN

Alzheimer's disease (AD) is a public health crisis due to debilitating cognitive symptoms and lack of curative treatments, in the context of increasing prevalence. Thus, it is critical to identify modifiable risk factors. High levels of meat consumption may increase AD risk. Many toxins are formed during meat cooking such as heterocyclic aromatic amines (HAAs). Our prior studies have shown that HAAs produce dopaminergic neurotoxicity. Given the mechanistic and pathological overlap between AD and dopaminergic disorders we investigated whether exposure to 2-amino-1-methyl-6-phenylimidazo [4,5-b] pyridine (PhIP), a prevalent dietary HAA formed during high-temperature meat cooking, may produce AD-relevant neurotoxicity. Here, C57BL/6 mice were treated with 100 or 200 mg/kg PhIP for 8 h or 75 mg/kg for 4 weeks and 16 weeks. PhIP exposure for 8 h produced oxidative damage, and AD-relevant alterations in hippocampal synaptic proteins, Amyloid-beta precursor protein (APP), and ß-Site amyloid precursor protein cleaving enzyme 1 (BACE1). PhIP exposure for 4 weeks resulted in an increase in BACE1. PhIP exposure for 16 weeks resulted in increased hippocampal oxidative damage, APP, BACE1, Aß aggregation, and tau phosphorylation. Quantification of intracellular nitrotyrosine revealed oxidative damage in cholinergic neurons after 8 h, 4 weeks and 16 weeks of PhIP exposure. Our study demonstrates that increase in oxidative damage, APP and BACE1 might be a possible mechanism by which PhIP promotes Aß aggregation. Given many patients with AD or PD exhibit neuropathological overlap, our study suggests that HAA exposure should be further studied for roles in mediating pathogenic overlap.


Asunto(s)
Enfermedad de Alzheimer/patología , Contaminación de Alimentos , Hipocampo/patología , Imidazoles , Neuronas/patología , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Estrés Oxidativo , Fosforilación , Agregación Patológica de Proteínas , Factores de Tiempo , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA