Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Cell ; 186(26): 5690-5704.e20, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38101407

RESUMEN

The maturation of genomic surveillance in the past decade has enabled tracking of the emergence and spread of epidemics at an unprecedented level. During the COVID-19 pandemic, for example, genomic data revealed that local epidemics varied considerably in the frequency of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lineage importation and persistence, likely due to a combination of COVID-19 restrictions and changing connectivity. Here, we show that local COVID-19 epidemics are driven by regional transmission, including across international boundaries, but can become increasingly connected to distant locations following the relaxation of public health interventions. By integrating genomic, mobility, and epidemiological data, we find abundant transmission occurring between both adjacent and distant locations, supported by dynamic mobility patterns. We find that changing connectivity significantly influences local COVID-19 incidence. Our findings demonstrate a complex meaning of "local" when investigating connected epidemics and emphasize the importance of collaborative interventions for pandemic prevention and mitigation.


Asunto(s)
COVID-19 , Humanos , COVID-19/epidemiología , COVID-19/transmisión , COVID-19/virología , Genómica , Pandemias/prevención & control , Salud Pública , SARS-CoV-2/genética , Control de Infecciones , Geografía
2.
Artículo en Inglés | MEDLINE | ID: mdl-37336825

RESUMEN

Fatigue has been characterized as a post COVID-19 condition known to persist months after SARS-CoV-2 infection. COVID-19 has been reported to be associated with impaired cognitive function, including disorders in attention, memory, information processing, and executive functions. The objective of this study was to determine if post-COVID fatigue, manifested as tiredness while performing low-intensity physical activity, has a detrimental effect on neuropsychological performance, to achieve this, we randomly selected 20 participants with post-COVID fatigue and 20 SARS-CoV-2 negative age-matched controls from a database of 360 residents of Tijuana, Baja California in a cross-sectional study design. All 40 participants responded to a health survey, along with a neuropsychological assessment test via telephone call. Statistical analysis was performed using a multiple linear regression model including the following independent variables: study condition (post-COVID fatigue or negative control), sex, age, years of education, hypertension, asthma, administration of supplemental oxygen during COVID-19 recovery, and the hour at which the evaluation started. Significant regression analysis was obtained for all global parameters of the assessment, including BANFE-2 score (p = 0.021, R2 Adj. = 0.263), NEUROPSI score (p = 0.008, R2 Adj. = 0.319), and total errors (p = 0.021, R2 Adj. = 0.263), with significant regression coefficients for study condition on two global parameters, BANFE-2 score (p = 0.028, ß = - 0.371) and NEUROPSI score (p = 0.010, ß = -0.428). These findings suggest that the presence of post-COVID fatigue is a factor associated with a decrease in neuropsychological performance.

3.
Brain Behav Immun ; 88: 920-924, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32276028

RESUMEN

The human genes for interleukin 13 (IL-13) and its receptor alpha 1 (IL-13Rα1) are in chromosomal regions associated with Parkinson's disease (PD). The interaction of IL-13 with its receptor increases the susceptibility of mouse dopaminergic neurons to oxidative stress. We identified two rare single SNPs in IL13 and IL13RA1 and measured their cytotoxic effects. rs148077750 is a missense leucine to proline substitution in IL13. It was found in individuals with early onset PD and no other known monogenic forms of the disease and is significantly linked with PD (Fisher's exact test: p-value = 0.01, odds ratio = 14.2). rs145868092 is a leucine to phenylalanine substitution in IL13RA1 affecting a residue critical for IL-13 binding. Both mutations increased the cytotoxic activity of IL-13 on human SH-SY5Y neurons exposed to sublethal doses of hydrogen peroxide, t-butyl hydroperoxide or RLS3, an inducer of ferroptosis. Our data show that both rs148077750 and rs145868092 conferred a gain-of-function that may increase the risk of developing PD.


Asunto(s)
Interleucina-13 , Enfermedad de Parkinson , Animales , Interleucina-13/genética , Subunidad alfa1 del Receptor de Interleucina-13/genética , Ratones , Estrés Oxidativo/genética , Enfermedad de Parkinson/genética , Polimorfismo de Nucleótido Simple
4.
Alcohol Clin Exp Res ; 44(7): 1378-1388, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32424852

RESUMEN

BACKGROUND: Alcohol use is on the rise among women in the United States which is especially concerning since women who drink have a higher risk of alcohol-related problems. Orexin (hypocretin) receptor antagonists may have some therapeutic value for alcohol-induced insomnia; however, the use of this class of drugs following female adolescent binge drinking is limited. The current study will address whether adolescent intermittent ethanol (AIE) in female rats can result in lasting changes in sleep pathology and whether orexin-targeted treatment can alleviate these deficits. METHODS: Following a 5-week AIE vapor model, young adult rats were evaluated on waking event-related oscillations (EROs) and EEG sleep. Subsequently, AIE rats were treated with orexin receptor 2 (OX2 R) antagonist (MK-1064; 10, 20mg/kg) to test for modifications in sleep pathology and waking ERO. RESULTS: Female AIE rats exhibited lasting changes in sleep compared to controls. This was demonstrated by increased fragmentation of slow wave sleep (SWS) and rapid eye movement sleep, as well as reductions in delta and theta power during SWS. There was no impact of AIE on waking EROs. Acute MK-1064 hastened SWS onset and increased the number of SWS episodes, without increasing sleep fragmentation in AIE and controls. While treatment with MK-1064 did not impact sleep EEG spectra, waking ERO energy was increased in delta, theta, and beta frequency bands. CONCLUSIONS: These results demonstrate that AIE can produce lasting changes in sleep in female rats, highly similar to what we previously found in males. Additionally, while the OX2 R antagonist promoted sleep in both alcohol-exposed and unexposed rats, it did not reverse most of the alcohol-induced disruptions in sleep. Thus, OX2 R antagonism may serve as a potential therapeutic strategy for the treatment of insomnia, but not the specific signs of alcohol-induced insomnia.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas , Ondas Encefálicas/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Antagonistas de los Receptores de Orexina/farmacología , Trastornos del Inicio y del Mantenimiento del Sueño , Sueño/efectos de los fármacos , Animales , Ritmo Delta/efectos de los fármacos , Modelos Animales de Enfermedad , Electroencefalografía , Femenino , Receptores de Orexina , Ratas , Privación de Sueño , Sueño REM/efectos de los fármacos , Sueño de Onda Lenta/efectos de los fármacos , Ritmo Teta/efectos de los fármacos , Consumo de Alcohol en Menores , Vigilia/efectos de los fármacos
5.
Int J Hyperthermia ; 37(1): 1368-1382, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33307890

RESUMEN

Hyperthermia is a potentially lethal side-effect of Methamphetamine (Meth), a stimulant drug. Activation of non-shivering thermogenesis in brown adipose tissue (BAT) is partly responsible for Meth-induced rise in temperature, with contributing sympathetic neurotransmitters, such as norepinephrine (NE), and reactive oxygen species (ROS). However, the mechanisms controlling the development of a molecular thermogenic program in brown adipocytes (BA) following Meth are unknown. We hypothesize that Meth and NE affect BAT cells, BA and macrophages, to modify their physiology and interactions, with consequences to thermogenic genes. We also hypothesize that ROS play a critical role in signaling transcription of thermogenic genes and their regulatory components. Using primary BA and macrophage cultures, we measured Meth and NE interference with physiological and phenotypic measures that are relevant to thermogenesis in BAT. Meth caused both BA and macrophages to decrease mitochondrial maximal capacity and increase ROS. In BA, the thermogenic genes UCP1, PPARγ, PGC1α and GADD45γ were transcriptionally increased by Meth in a ROS-dependent manner. In macrophages, Meth increased oxidative stress response and caused a predominance of M2 subset markers. BA transcriptional changes in response to Meth and NE were significantly controlled by macrophages. The results suggest that BA and macrophages respond to Meth and NE, with effects on mitochondrial functions and transcription of genes involved in thermogenesis. ROS-dependent signals in BA and cellular interactions between BA and macrophages synergize to regulate the BAT environment and control critical pathways leading to Meth-hyperthermia.


Asunto(s)
Adipocitos Marrones , Metanfetamina , Tejido Adiposo Pardo , Macrófagos , Metanfetamina/efectos adversos , Termogénesis
6.
Addict Biol ; 25(2): e12732, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-30884076

RESUMEN

Alcohol exposure typically begins in adolescence, and frequent binge drinking has been associated with health risk behaviors including alcohol use disorders (AUDs). Few studies have documented the effects of a history of adolescent binge drinking on neurophysiological consequences in young adulthood. Synchrony of phase (phase locking (PL)) of event-related oscillations (EROs) within and between different brain areas reflects communication exchange between neural networks and is a sensitive measure of adolescent development in both rats and humans, and thus may be a good translational measure of the potential harmful effects of alcohol exposure during adolescence. In this study, EROs were collected from 1041 young adults of Mexican American and American Indian ancestry (age 18-30 years) with and without a history of adolescent binge drinking (five drinks for boys and four for girls per occasion at least once per month) and in 74 young adult rats with and without a history of 5 weeks of adolescent alcohol vapor exposure. PL of theta and beta frequencies between frontal and parietal cortex were estimated using an auditory-oddball paradigm in the rats and a visual facial expression paradigm in the humans. Significantly lower PL between frontal and parietal cortices in the theta frequencies was seen in both the humans and the rats with a history of adolescent alcohol exposure as compared with their controls. These findings suggest that alcohol exposure during adolescence may result in decreases in synchrony between cortical neuronal networks, suggesting a developmental delay, in young adult humans and in rats.


Asunto(s)
Alcoholismo/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Electroencefalografía/métodos , Etanol/farmacología , Consumo de Alcohol en Menores/estadística & datos numéricos , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Americanos Mexicanos , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Adulto Joven , Indio Americano o Nativo de Alaska
7.
Proc Natl Acad Sci U S A ; 114(36): 9731-9736, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28827363

RESUMEN

When food resources are scarce, endothermic animals can lower core body temperature (Tb). This phenomenon is believed to be part of an adaptive mechanism that may have evolved to conserve energy until more food becomes available. Here, we found in the mouse that the insulin-like growth factor 1 receptor (IGF-1R) controls this response in the central nervous system. Pharmacological or genetic inhibition of IGF-1R enhanced the reduction of temperature and of energy expenditure during calorie restriction. Full blockade of IGF-1R affected female and male mice similarly. In contrast, genetic IGF-1R dosage was effective only in females, where it also induced transient and estrus-specific hypothermia in animals fed ad libitum. These effects were regulated in the brain, as only central, not peripheral, pharmacological activation of IGF-1R prevented hypothermia during calorie restriction. Targeted IGF-1R knockout selectively in forebrain neurons revealed that IGF signaling also modulates calorie restriction-dependent Tb regulation in regions rostral of the canonical hypothalamic nuclei involved in controlling body temperature. In aggregate, these data identify central IGF-1R as a mediator of the integration of nutrient and temperature homeostasis. They also show that calorie restriction, IGF-1R signaling, and body temperature, three of the main regulators of metabolism, aging, and longevity, are components of the same pathway.


Asunto(s)
Restricción Calórica/efectos adversos , Hipotermia/fisiopatología , Receptor IGF Tipo 1/fisiología , Envejecimiento/fisiología , Animales , Metabolismo Energético/fisiología , Femenino , Dosificación de Gen , Homeostasis/fisiología , Hipotermia/etiología , Hipotermia/prevención & control , Longevidad/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Podofilotoxina/análogos & derivados , Podofilotoxina/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/genética , Caracteres Sexuales , Transducción de Señal/fisiología
8.
Alcohol Clin Exp Res ; 43(12): 2547-2558, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31589333

RESUMEN

BACKGROUND: Adolescence is a critical period for neural development, and alcohol exposure during adolescence can lead to an elevated risk for health consequences as well as alcohol use disorders. Clinical and experimental data suggest that chronic alcohol exposure may produce immunomodulatory effects that can lead to the activation of pro-inflammatory cytokine pathways as well as microglial markers. The present study evaluated, in brain and blood, the effects of adolescent alcohol exposure and withdrawal on microglia and on the most representative pro- and anti-inflammatory cytokines and major chemokines that can contribute to the establishing of a neuroinflammatory environment. METHODS: Wistar rats (males, n = 96) were exposed to ethanol (EtOH) vapors, or air control, for 5 weeks over adolescence (PD22-PD58). Brains and blood samples were collected at 3 time points: (i) after 35 days of vapor/air exposure (PD58); (ii) after 1 day of withdrawal (PD59), and (iii) 28 days after withdrawal (PD86). The ionized calcium-binding adapter molecule 1 (Iba-1) was used to index microglial activation, and cytokine/chemokine responses were analyzed using magnetic bead panels. RESULTS: After 35 days of adolescent vapor exposure, a significant increase in Iba-1 immunoreactivity was seen in amygdala, frontal cortex, hippocampus, and substantia nigra. However, Iba-1 density returned to control levels at both 1 day and 28 days of withdrawal except in the hippocampus where Iba-1 density was significantly lower than controls. In serum, adolescent EtOH exposure induced a reduction in IL-13 and an increase in fractalkine at day 35. After 1 day of withdrawal, IL-18 was reduced, and IP-10 was elevated, whereas both IP-10 and IL-10 were elevated at 28 days following withdrawal. In the frontal cortex, adolescent EtOH exposure induced an increase in IL-1ß at day 35, and 28 days of withdrawal, and IL-10 was increased after 28 days of withdrawal. CONCLUSION: These data demonstrate that EtOH exposure during adolescence produces significant microglial activation; however, inflammatory markers seen in the blood appear to differ from those observed in the brain.


Asunto(s)
Encéfalo/metabolismo , Citocinas/metabolismo , Etanol/efectos adversos , Síndrome de Abstinencia a Sustancias/metabolismo , Factores de Edad , Animales , Proteínas de Unión al Calcio/metabolismo , Citocinas/sangre , Masculino , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Ratas , Síndrome de Abstinencia a Sustancias/sangre , Factores de Tiempo
9.
Proc Natl Acad Sci U S A ; 113(1): 26-33, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26668358

RESUMEN

Diacylglycerol lipases (DAGLα and DAGLß) convert diacylglycerol to the endocannabinoid 2-arachidonoylglycerol. Our understanding of DAGL function has been hindered by a lack of chemical probes that can perturb these enzymes in vivo. Here, we report a set of centrally active DAGL inhibitors and a structurally related control probe and their use, in combination with chemical proteomics and lipidomics, to determine the impact of acute DAGL blockade on brain lipid networks in mice. Within 2 h, DAGL inhibition produced a striking reorganization of bioactive lipids, including elevations in DAGs and reductions in endocannabinoids and eicosanoids. We also found that DAGLα is a short half-life protein, and the inactivation of DAGLs disrupts cannabinoid receptor-dependent synaptic plasticity and impairs neuroinflammatory responses, including lipopolysaccharide-induced anapyrexia. These findings illuminate the highly interconnected and dynamic nature of lipid signaling pathways in the brain and the central role that DAGL enzymes play in regulating this network.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Encéfalo/efectos de los fármacos , Diglicéridos/metabolismo , Endocannabinoides/metabolismo , Inhibidores Enzimáticos/farmacología , Glicéridos/metabolismo , Lipoproteína Lipasa/antagonistas & inhibidores , Plasticidad Neuronal/efectos de los fármacos , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Inhibidores Enzimáticos/química , Lipoproteína Lipasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Cannabinoides/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Alcohol Clin Exp Res ; 42(3): 624-633, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29286538

RESUMEN

BACKGROUND: Disturbances in sleep architecture, especially reductions in slow-wave sleep (SWS), are symptoms commonly observed in individuals with alcohol use disorders. Recent clinical trials have demonstrated that the anticonvulsant and analgesic drug gabapentin may have therapeutic value in normalizing sleep quality in recovering alcoholics. However, the brain mechanisms underlying this improvement in sleep following gabapentin treatment remain unknown. METHODS: In this study, adult Wistar rats were exposed to 8 weeks of chronic intermittent ethanol [EtOH] vapor (blood EtOH concentrations averaged 128.2 ± 17.4 mg/dl) or control conditions and then withdrawn. Sleep electroencephalograms [EEGs] and event-related oscillations (EROs) were evaluated at baseline prior to EtOH exposure and 24 hours following EtOH withdrawal. Four weeks following EtOH withdrawal the effects of saline and 2 doses of gabapentin (30, 120 mg/kg), on EROs and sleep EEGs, were evaluated. RESULTS: As compared to baseline, 24 hours following alcohol withdrawal SWS became fragmented as indexed by a significant increase in the number and a decrease in the duration of SWS episodes. Compared to controls, the EtOH-exposed group had more ERO energy in the beta frequency band in the parietal cortex. Gabapentin induced a dose-dependent decrease in the latency to the first SWS episode, and a reduction in sleep fragmentation. Gabapentin also produced a dose-dependent increase in ERO energy in the control group that was significantly attenuated in the EtOH-exposed group in the theta, and beta frequency bands. CONCLUSIONS: Taken together, these studies suggest that gabapentin can reverse some of the alcohol-induced sleep and EEG deficits but does not eliminate all of the enduring brain effects of EtOH exposure.


Asunto(s)
Ondas Encefálicas/efectos de los fármacos , Encéfalo/efectos de los fármacos , Depresores del Sistema Nervioso Central/efectos adversos , Etanol/efectos adversos , Antagonistas de Aminoácidos Excitadores/farmacología , Gabapentina/farmacología , Sueño/efectos de los fármacos , Animales , Encéfalo/fisiopatología , Ondas Encefálicas/fisiología , Depresores del Sistema Nervioso Central/administración & dosificación , Electroencefalografía , Etanol/administración & dosificación , Masculino , Ratas , Ratas Wistar , Sueño/fisiología , Sueño de Onda Lenta/efectos de los fármacos , Sueño de Onda Lenta/fisiología , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología
11.
J Neurosci ; 36(18): 5170-80, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27147667

RESUMEN

UNLABELLED: The proinflammatory cytokine IL-18 has central anorexigenic effects and was proposed to contribute to loss of appetite observed during sickness. Here we tested in the mouse the hypothesis that IL-18 can decrease food intake by acting on neurons of the bed nucleus of the stria terminalis (BST), a component of extended amygdala recently shown to influence feeding via its projections to the lateral hypothalamus (LH). We found that both subunits of the heterodimeric IL-18 receptor are highly expressed in the BST and that local injection of recombinant IL-18 (50 ng/ml) significantly reduced c-fos activation and food intake for at least 6 h. Electrophysiological experiments performed in BST brain slices demonstrated that IL-18 strongly reduces the excitatory input on BST neurons through a presynaptic mechanism. The effects of IL-18 are cell-specific and were observed in Type III but not in Type I/II neurons. Interestingly, IL-18-sensitve Type III neurons were recorded in the juxtacapsular BST, a region that contains BST-LH projecting neurons. Reducing the excitatory input on Type III GABAergic neurons, IL-18 can increase the firing of glutamatergic LH neurons through a disinhibitory mechanism. Imbalance between excitatory and inhibitory activity in the LH can induce changes in food intake. Effects of IL-18 were mediated by the IL-18R because they were absent in neurons from animals null for IL-18Rα (Il18ra(-/-)), which lack functional IL-18 receptors. In conclusion, our data show that IL-18 may inhibit feeding by inhibiting the activity of BST Type III GABAergic neurons. SIGNIFICANCE STATEMENT: Loss of appetite during sickness is a common and often debilitating phenomenon. Although proinflammatory cytokines are recognized as mediators of these anorexigenic effects, their mechanism and sites of action remain poorly understood. Here we show that interleukin 18, an anorexigenic cytokine, can act on neurons of the bed nucleus of the stria terminalis to reduce food intake via the IL-18 receptor. The findings identify a site and a mode of action that indicate targets for the treatment of cachexia or other eating disorders.


Asunto(s)
Conducta Alimentaria/fisiología , Interleucina-18/fisiología , Núcleos Septales/fisiología , Animales , Fenómenos Electrofisiológicos/fisiología , Área Hipotalámica Lateral/fisiología , Interleucina-18/biosíntesis , Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Proteínas Recombinantes/farmacología , Sinapsis/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología
12.
J Neuroinflammation ; 14(1): 88, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28427412

RESUMEN

BACKGROUND: The majority of Parkinson's disease (PD) cases are sporadic and idiopathic suggesting that this neurodegenerative disorder is the result of both environmental and genetic factors. Stress and neuroinflammation are among the factors being investigated for their possible contributions to PD. Experiments in rodents showed that severe chronic stress can reduce the number of dopaminergic neurons in the substantia nigra pars compacta (SNc); the same cells that are lost in PD. These actions are at least in part mediated by increased oxidative stress. Here, we tested the hypothesis that the interleukin-13 receptor alpha 1 (IL-13Rα1), a cytokine receptor whose activation increases the vulnerability of dopaminergic neurons to oxidative damage, participates in the stress-dependent damage of these neurons. METHODS: Mice were subject to daily sessions of 8 h (acute) stress for 16 weeks (5 days a week), a procedure previously showed to induce loss of dopaminergic neurons in the SNc. The source and the kinetics of interleukin-13 (IL-13), the endogenous ligand of IL-13Rα1, were evaluated 0, 1, 3, 6, and 8 h and at 16 weeks of stress. Identification of IL-13 producing cell-type was performed by immunofluorescent and by in situ hybridization experiments. Markers of oxidative stress, microglia activation, and the number of dopaminergic neurons in IL-13Rα1 knock-out animals (Il13ra1 Y/ - ) and their wild-type littermates (Il13ra1 Y/+ ) were evaluated at 16 weeks of stress and at 20 weeks, following a 4 week non-stressed period and compared to non-stressed mice. RESULTS: IL-13 was expressed in microglial cells within the SN and in a fraction of the tyrosine hydroxylase-positive neurons in the SNc. IL-13 levels were elevated during daily stress and peaked at 6 h. 16 weeks of chronic restraint stress significantly reduced the number of SNc dopaminergic neurons in Il13ra1 Y/+ mice. Neuronal loss at 16 weeks was significantly lower in Il13ra1 Y/- mice. However, the loss of dopaminergic neurons measured at 20 weeks, after 4 weeks of non-stress following the 16 weeks of stress, was similar in Il13ra1 Y/+ and Il13ra1 Y/- mice. CONCLUSIONS: IL-13, a cytokine previously demonstrated to increase the susceptibility of SNc dopaminergic neurons to oxidative stress, is elevated in the SN by restraint stress. Lack of IL-13Rα1 did not prevent nor halted but delayed neuronal loss in the mouse model of chronic restraint stress. IL-13/IL-13Rα1 may represent a target to reduce the rate of DA neuronal loss that can occur during severe chronic restraint stress.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Subunidad alfa1 del Receptor de Interleucina-13/deficiencia , Estrés Oxidativo/fisiología , Estrés Psicológico/metabolismo , Animales , Recuento de Células/métodos , Neuronas Dopaminérgicas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Psicológico/patología , Sustancia Negra/metabolismo , Sustancia Negra/patología
13.
FASEB J ; 30(1): 81-92, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26304220

RESUMEN

Brown adipocytes (BAs) are specialized for adaptive thermogenesis and, upon sympathetic stimulation, activate mitochondrial uncoupling protein (UCP)-1 and oxidize fatty acids to generate heat. The capacity for brown adipose tissue (BAT) to protect against obesity and metabolic disease is recognized, yet information about which signals activate BA, besides ß3-adrenergic receptor stimulation, is limited. Using single-cell transcriptomics, we confirmed the presence of mRNAs encoding traditional BAT markers (i.e., UCP1, expressed in 100% of BAs Adrb3, expressed in <50% of BAs) in mouse and have shown single-cell variability (>1000-fold) in their expression at both the mRNA and protein levels. We further identified mRNAs encoding novel markers, orphan GPCRs, and many receptors that bind the classic neurotransmitters, neuropeptides, chemokines, cytokines, and hormones. The transcriptome variability between BAs suggests a much larger range of responsiveness of BAT than previously recognized and that not all BAs function identically. We examined the in vivo functional expression of 12 selected receptors by microinjecting agonists into live mouse BAT and analyzing the metabolic response. In this manner, we expanded the number of known receptors on BAs at least 25-fold, while showing that the expression of classic BA markers is more complex and variable than previously thought.


Asunto(s)
Adipocitos Marrones/citología , Tejido Adiposo Pardo/metabolismo , Homeostasis/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Tejido Adiposo Pardo/citología , Animales , Canales Iónicos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Obesidad/metabolismo , Termogénesis/fisiología , Transcriptoma
14.
J Biol Chem ; 288(15): 10722-35, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23457303

RESUMEN

Sirt1 is a NAD(+)-dependent class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, emerging evidence suggests that neuronal Sirt1 activity plays a role in the central regulation of energy balance and glucose metabolism. To assess this idea, we generated Sirt1 neuron-specific knockout (SINKO) mice. On both standard chow and HFD, SINKO mice were more insulin sensitive than Sirt1(f/f) mice. Thus, SINKO mice had lower fasting insulin levels, improved glucose tolerance and insulin tolerance, and enhanced systemic insulin sensitivity during hyperinsulinemic euglycemic clamp studies. Hypothalamic insulin sensitivity of SINKO mice was also increased over controls, as assessed by hypothalamic activation of PI3K, phosphorylation of Akt and FoxO1 following systemic insulin injection. Intracerebroventricular injection of insulin led to a greater systemic effect to improve glucose tolerance and insulin sensitivity in SINKO mice compared with controls. In line with the in vivo results, insulin-induced AKT and FoxO1 phosphorylation were potentiated by inhibition of Sirt1 in a cultured hypothalamic cell line. Mechanistically, this effect was traced to a reduced effect of Sirt1 to directly deacetylate and repress IRS-1 function. The enhanced central insulin signaling in SINKO mice was accompanied by increased insulin receptor signal transduction in liver, muscle, and adipose tissue. In summary, we conclude that neuronal Sirt1 negatively regulates hypothalamic insulin signaling, leading to systemic insulin resistance. Interventions that reduce neuronal Sirt1 activity have the potential to improve systemic insulin action and limit weight gain on an obesigenic diet.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sirtuina 1/metabolismo , Animales , Células Cultivadas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Glucosa/genética , Glucosa/metabolismo , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Insulina/genética , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Especificidad de Órganos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirtuina 1/genética
15.
Brain Behav Immun ; 40: 85-94, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24603356

RESUMEN

Interleukin (IL)-18 is a cytokine previously demonstrated to participate in neuroinflammatory processes. Since the components of the IL-18 receptor complex are expressed in neurons throughout the brain, IL-18 is also believed to directly influence neuronal function. Here we tested this hypothesis on mouse hippocampal neurons by measuring the effects of IL-18 on three pathways previously shown to be regulated by this cytokine in non-neuronal cells: the MAPK pathways, p38 and ERK1/2 MAPKs, STAT3 and NF-κB. Experiments were carried out in vitro using the immortalized hippocampal neuronal line HT-22 or in vivo following i.c.v. injection with recombinant mouse IL-18. We showed that IL-18 did not activate NF-κB in HT-22 cells whereas it induced a rapid (within 15min) activation of the MAPK pathways. Moreover, we demonstrated that IL-18 treatment enhanced P-STAT3 (Tyr705)/STAT3 ratio in the nucleus of HT-22 cells after 30-60min of exposure. A similar increase in P-STAT3 (Tyr705)/STAT3 ratio was observed in the whole hippocampus one hour after i.c.v. injection. These data demonstrate that IL-18 can act directly on neuronal cells affecting the STAT3 pathway; therefore, possibly regulating the expression of specific genes within the hippocampus. This effect may help to explain some of the IL-18-induced effects on synaptic plasticity and functionality within the hippocampal system.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/metabolismo , Interleucina-18/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Células Cultivadas , Hipocampo/efectos de los fármacos , Interleucina-18/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-18/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal
16.
J Immunol ; 189(12): 5498-502, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23169588

RESUMEN

Inflammation and its mediators, including cytokines and reactive oxygen species, are thought to contribute to neurodegeneration. In the mouse brain, we found that IL-13Rα1 was expressed in the dopaminergic (DA) neurons of the substantia nigra pars compacta, which are preferentially lost in human Parkinson's disease. Mice deficient for Il13ra1 exhibited resistance to loss of DA neurons in a model of chronic peripheral inflammation using bacterial LPS. IL-13, as well as IL-4, potentiated the cytotoxic effects of t-butyl hydroperoxide and hydrogen peroxide on mouse DA MN9D cells. Collectively, our data indicate that expression of IL-13Rα1 on DA neurons can increase their susceptibility to oxidative stress-mediated damage, thereby contributing to their preferential loss. In humans, Il13ra1 lies on the X chromosome within the PARK12 locus of susceptibility to Parkinson's disease, suggesting that IL-13Rα1 may have a role in the pathogenesis of this neurodegenerative disease.


Asunto(s)
Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/metabolismo , Subunidad alfa1 del Receptor de Interleucina-13/biosíntesis , Lipopolisacáridos/toxicidad , Estrés Oxidativo/inmunología , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Enfermedad Crónica , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Predisposición Genética a la Enfermedad/etiología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Subunidad alfa1 del Receptor de Interleucina-13/deficiencia , Subunidad alfa1 del Receptor de Interleucina-13/genética , Lipopolisacáridos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
17.
bioRxiv ; 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38979362

RESUMEN

Neurons dynamically regulate their proteome in response to sensory input, a key process underlying experience-dependent plasticity. We characterized the visual experience-dependent nascent proteome within a brief, defined time window after stimulation using an optimized metabolic labeling approach. Visual experience induced cell type-specific and age-dependent alterations in the nascent proteome, including proteostasis-related processes. We identified Emerin as the top activity-induced candidate plasticity protein and demonstrated that its rapid activity-induced synthesis is transcription-independent. In contrast to its nuclear localization and function in myocytes, activity-induced neuronal Emerin is abundant in the endoplasmic reticulum and broadly inhibits protein synthesis, including translation regulators and synaptic proteins. Downregulating Emerin shifted the dendritic spine population from predominantly mushroom morphology to filopodia and decreased network connectivity. In mice, decreased Emerin reduced visual response magnitude and impaired visual information processing. Our findings support an experience-dependent feed-forward role for Emerin in temporally gating neuronal plasticity by negatively regulating translation.

18.
Proc Natl Acad Sci U S A ; 107(34): 15229-34, 2010 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-20660766

RESUMEN

Galanin receptors type 1 (GalR1) and/or type 2 (GalR2) represent unique pharmacological targets for treatment of seizures and epilepsy. Previous studies have shown that the endogenous peptide ligand galanin exerts powerful anticonvulsant effect through activation of these two G protein-coupled receptors, which are highly expressed in the temporal lobe of rodent brain. Here we report the characterization of a putative GalR2-positive allosteric modulator CYM2503. CYM2503 potentiated the galanin-stimulated IP1 accumulation in HEK293 cells stably expressing GalR2 receptor, whereas it exhibited no detectable affinity for the (125)I galanin-binding site of GalR2 receptor, an effect consistent with that of a positive allosteric modulator. In the rat Li-pilocarpine status epilepticus model, CYM2503, injected intraperitoneally, increased the latency to first electrographic seizure and the latency to first stage 3 behavioral seizure, decreased the latency to the establishment of status epilepticus, and dramatically decreased the mortality. In a Li-pilocarpine seizure model in mice, CYM2503 increased the latency to first electrographic seizure and decreased the total time in seizure. CYM2503 also attenuated electroshock-induced seizures in mice. Thus, CYM2503 provides a starting point for the development of anticonvulsant therapy using the galanin R2 receptor as target.


Asunto(s)
Anticonvulsivantes/farmacología , Carbamatos/farmacología , Dipéptidos/farmacología , Quinolonas/farmacología , Receptor de Galanina Tipo 2/agonistas , Regulación Alostérica , Animales , Línea Celular , Modelos Animales de Enfermedad , Electrochoque , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Pilocarpina/toxicidad , Ratas , Ratas Wistar , Receptor de Galanina Tipo 1/metabolismo , Receptor de Galanina Tipo 2/metabolismo , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Transducción de Señal/efectos de los fármacos , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico
19.
J Biol Chem ; 286(17): 14983-90, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21330367

RESUMEN

The objective is to investigate the role of insulin-like growth factor 1 (IGF-1) in the regulation of core body temperature. Sequencing cDNA libraries from individual warm-sensitive neurons from the preoptic area (POA) of the hypothalamus, a region involved in the central control of thermoregulation, identified neurons that express both IGF-1 receptor (IGF-1R) and insulin receptor transcripts. The effects of administration of IGF-1 into the POA was measured by radiotelemetry monitoring of core temperature, brown adipose tissue (BAT) temperature, metabolic assessment, and imaging of BAT by positron emission tomography of 2-[(18)F]fluoro-2-deoxyglucose uptake combined with computed tomography. IGF-1 injection into the POA caused dose-dependent hyperthermia that could be blocked by pretreatment with the IGF-1R tyrosine kinase inhibitor, PQ401. The IGF-1-evoked hyperthermia involved activation of brown adipose tissue and was accompanied by a switch from glycolysis to fatty acid oxidation as a source of energy as shown by lowered respiratory exchange ratio. Transgenic mice that lack neuronal insulin receptor expression in the brain (NIRKO mice) were unable to mount the full hyperthermic response to IGF-1, suggesting that the IGF-1 mediated hyperthermia is partly dependent on expression of functional neuronal insulin receptors. These data indicate a novel thermoregulatory role for both IGF-1R and neuronal insulin receptors in IGF-1 activation of BAT and hyperthermia. These central effects of IGF-1 signaling may play a role in regulation of metabolic rate, aging, and the risk of developing type 2 diabetes.


Asunto(s)
Fiebre/etiología , Hipotálamo Anterior/química , Factor I del Crecimiento Similar a la Insulina/fisiología , Receptor de Insulina/fisiología , Animales , Regulación de la Temperatura Corporal , Encéfalo/metabolismo , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones , Ratones Transgénicos , Receptor IGF Tipo 1 , Transducción de Señal
20.
J Neurosci ; 30(12): 4369-81, 2010 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-20335473

RESUMEN

The preoptic area/anterior hypothalamus, a region that contains neurons that control thermoregulation, is the main locus at which histamine affects body temperature. Here we report that histamine reduced the spontaneous firing rate of GABAergic preoptic neurons by activating H3 subtype histamine receptors. This effect involved a decrease in the level of phosphorylation of the extracellular signal-regulated kinase and was not dependent on synaptic activity. Furthermore, a population of non-GABAergic neurons was depolarized, and their firing rate was enhanced by histamine acting at H1 subtype receptors. In our experiments, activation of the H1R receptors was linked to the PLC pathway and Ca(2+) release from intracellular stores. This depolarization persisted in TTX or when fast synaptic potentials were blocked, indicating that it represents a postsynaptic effect. Single-cell reverse transcription-PCR analysis revealed expression of H3 receptors in a population of GABAergic neurons, while H1 receptors were expressed in non-GABAergic cells. Histamine applied in the median preoptic nucleus induced a robust, long-lasting hyperthermia effect that was mimicked by either H1 or H3 histamine receptor subtype-specific agonists. Our data indicate that histamine modulates the core body temperature by acting at two distinct populations of preoptic neurons that express H1 and H3 receptor subtypes, respectively.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Histamina/farmacología , Neuronas/efectos de los fármacos , Área Preóptica/citología , Receptores Histamínicos H1/metabolismo , Receptores Histamínicos H3/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Calcio/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/genética , Agonistas de los Receptores Histamínicos/farmacología , Antagonistas de los Receptores Histamínicos/farmacología , Técnicas In Vitro , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Técnicas de Placa-Clamp , Receptores Histamínicos H1/efectos de los fármacos , Receptores Histamínicos H3/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Telemetría/métodos , Tetrodotoxina/farmacología , Fosfolipasas de Tipo C/metabolismo , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda