Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 40(11): 2332-2342, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32005763

RESUMEN

Emotional disorders are common comorbid conditions that further exacerbate the severity and chronicity of chronic pain. However, individuals show considerable vulnerability to the development of chronic pain under similar pain conditions. In this study on male rat and mouse models of chronic neuropathic pain, we identify the histone deacetylase Sirtuin 1 (SIRT1) in central amygdala as a key epigenetic regulator that controls the development of comorbid emotional disorders underlying the individual vulnerability to chronic pain. We found that animals that were vulnerable to developing behaviors of anxiety and depression under the pain condition displayed reduced SIRT1 protein levels in central amygdala, but not those animals resistant to the emotional disorders. Viral overexpression of local SIRT1 reversed this vulnerability, but viral knockdown of local SIRT1 mimicked the pain effect, eliciting the pain vulnerability in pain-free animals. The SIRT1 action was associated with CaMKIIα downregulation and deacetylation of histone H3 lysine 9 at the CaMKIIα promoter. These results suggest that, by transcriptional repression of CaMKIIα in central amygdala, SIRT1 functions to guard against the emotional pain vulnerability under chronic pain conditions. This study indicates that SIRT1 may serve as a potential therapeutic molecule for individualized treatment of chronic pain with vulnerable emotional disorders.SIGNIFICANCE STATEMENT Chronic pain is a prevalent neurological disease with no effective treatment at present. Pain patients display considerably variable vulnerability to developing chronic pain, indicating individual-based molecular mechanisms underlying the pain vulnerability, which is hardly addressed in current preclinical research. In this study, we have identified the histone deacetylase Sirtuin 1 (SIRT1) as a key regulator that controls this pain vulnerability. This study reveals that the SIRT1-CaMKIIaα pathway in central amygdala acts as an epigenetic mechanism that guards against the development of comorbid emotional disorders under chronic pain, and that its dysfunction causes increased vulnerability to the development of chronic pain. These findings suggest that SIRT1 activators may be used in a novel therapeutic approach for individual-based treatment of chronic pain.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Núcleo Amigdalino Central/fisiopatología , Dolor Crónico/fisiopatología , Distrés Psicológico , Sirtuina 1/fisiología , Neuralgia del Trigémino/fisiopatología , Acetilación , Animales , Ansiedad/etiología , Ansiedad/fisiopatología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Núcleo Amigdalino Central/enzimología , Dolor Crónico/psicología , Depresión/etiología , Depresión/fisiopatología , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Conducta Exploratoria , Neuronas GABAérgicas/enzimología , Vectores Genéticos , Histonas/metabolismo , Hiperalgesia/fisiopatología , Masculino , Ratones , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Natación , Transcripción Genética , Neuralgia del Trigémino/psicología
2.
J Psychopharmacol ; 33(5): 568-576, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30835157

RESUMEN

BACKGROUND: Phosphodiesterase-2 (PDE2) is a cyclic nucleotide phosphodiesterase and is highly expressed in the amygdala, which suggests its important role in anxiety-like behavior. AIMS: The present study examined whether reduced PDE2A expression in the central nucleus of the amygdala (CeA) produces anxiolytic-like effects in mice. METHODS: PDE2A knockdown in amygdaloid (AR5) cells or the CeA was established using a lentiviral vector-based siRNA system. The anxiety-like behaviors were detected by the elevated plus maze (EPM) and hole-board tests in mice. The related proteins involved in cAMP/cGMP-dependent signaling, such as specific marker VASPser239, CREBser133 and BDNF were detected by immunoblot analysis. RESULTS: PDE2A inhibition in AR-5 cells resulted in increases in cAMP/cGMP-related pVASPser239 and pCREBser133. Behavioral tests showed that PDE2A knockdown in the CeA induced anxiolytic-like effects as evidenced by the increases in percentages of open-arm entries and time spent in the open arms in the EPM test, and the increases in head dips and time spent in head dipping in the hole-board test. However, these anxiolytic-like effects were antagonized by pre-treatment of soluble guanylyl cyclase inhibitor ODQ or adenylate cyclase inhibitor SQ. Furthermore, PDE2A knockdown significantly increased pVASPSer239, pCREBSer133 and decreased BDNF expression in the amygdala. Pre-intra-CeA of ODQ or SQ reversed or partially prevented the effects of PDE2A knockdown on these proteins. CONCLUSIONS: The results suggest that PDE2A plays a crucial role in the regulation of anxiety by the cGMP/cAMP-dependent pVASP-pCREB-BDNF signaling pathway.


Asunto(s)
Ansiedad/metabolismo , Conducta Animal/fisiología , Núcleo Amigdalino Central/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Transducción de Señal/fisiología , Animales , Ansiedad/enzimología , Línea Celular , Núcleo Amigdalino Central/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/deficiencia , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
3.
Biol Psychiatry ; 84(3): 193-201, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29198469

RESUMEN

BACKGROUND: Despite the high cost and widespread prevalence of alcohol use disorders, treatment options are limited, underscoring the need for new, effective medications. Previous results using protein kinase C epsilon (PKCε) knockout mice, RNA interference against PKCε, and peptide inhibitors of PKCε predict that small-molecule inhibitors of PKCε should reduce alcohol consumption in humans. METHODS: We designed a new class of PKCε inhibitors based on the Rho-associated protein kinase (ROCK) inhibitor Y-27632. In vitro kinase and binding assays were used to identify the most potent compounds. Their effects on ethanol-stimulated synaptic transmission; ethanol, sucrose, and quinine consumption; ethanol-induced loss of righting; and ethanol clearance were studied in mice. RESULTS: We identified two compounds that inhibited PKCε with Ki <20 nM, showed selectivity for PKCε over other kinases, crossed the blood-brain barrier, achieved effective concentrations in mouse brain, prevented ethanol-stimulated gamma-aminobutyric acid release in the central amygdala, and reduced ethanol consumption when administered intraperitoneally at 40 mg/kg in wild-type but not in Prkce-/- mice. One compound also reduced sucrose and saccharin consumption, while the other was selective for ethanol. Both transiently impaired locomotion through an off-target effect that did not interfere with their ability to reduce ethanol intake. One compound prolonged recovery from ethanol-induced loss of righting but this was also due to an off-target effect since it was present in Prkce-/- mice. Neither altered ethanol clearance. CONCLUSIONS: These results identify lead compounds for development of PKCε inhibitors that reduce alcohol consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Núcleo Amigdalino Central/enzimología , Proteína Quinasa C-epsilon/genética , Inhibidores de Proteínas Quinasas/farmacología , Transmisión Sináptica/efectos de los fármacos , Alcoholismo/enzimología , Alcoholismo/fisiopatología , Amidas/farmacocinética , Amidas/farmacología , Animales , Núcleo Amigdalino Central/efectos de los fármacos , Núcleo Amigdalino Central/fisiopatología , Depresores del Sistema Nervioso Central/farmacología , Condicionamiento Psicológico , Modelos Animales de Enfermedad , Etanol , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de Proteínas Quinasas/farmacocinética , Piridinas/farmacocinética , Piridinas/farmacología
4.
Addict Biol ; 23(6): 1223-1232, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29071769

RESUMEN

Fatty acid amide hydrolase (FAAH) is an enzyme that prominently degrades the major endocannabinoid N-arachidonoylethanolamine (anandamide). Inhibition of this enzyme leads to increased anandamide levels in brain regions that modulate stress and anxiety. Recently, we found that genetically selected Marchigian Sardinian alcohol-preferring (msP) rats display hyperactive FAAH in amygdalar regions that was associated with increased stress sensitivity and a hyper-anxious phenotype. Our previous work has also demonstrated that msPs display an innate preference for and excessive consumption of alcohol, potentially reflecting a form of self-medication to gain relief from hyper-anxious states. Here, we expand on our previous work by microinjecting the selective FAAH inhibitor URB597 (vehicle, 0.03, 0.1 and 1.0 µg per rat) into the central amygdala (CeA) and basolateral amygdala in msP versus non-selected Wistar rats to evaluate the effects of localized FAAH inhibition on operant alcohol self-administration and restraint-induced anxiety using the elevated plus maze. Intra-CeA URB597 significantly reduced alcohol self-administration in msP but not in Wistar rats. Intra-basolateral amygdala URB597 also attenuated alcohol drinking in msPs, although the effect was less pronounced relative to CeA treatment. In contrast, control experiments administering URB597 into the ventral tegmental area produced no genotypic differences in drinking. We also found that URB597 treatment in the CeA significantly reduced the anxiogenic effects of restraint stress in msPs, although no effects were detected in Wistars. Dysregulation of FAAH regulated systems in the major output region of the amygdala may drive the propensity for co-morbid expression of anxiety and excessive alcohol use.


Asunto(s)
Alcoholismo/enzimología , Amidohidrolasas/antagonistas & inhibidores , Trastornos de Ansiedad/enzimología , Núcleo Amigdalino Central/enzimología , Análisis de Varianza , Animales , Benzamidas/administración & dosificación , Benzamidas/farmacología , Carbamatos/administración & dosificación , Carbamatos/farmacología , Depresores del Sistema Nervioso Central/administración & dosificación , Condicionamiento Operante , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Etanol/administración & dosificación , Masculino , Aprendizaje por Laberinto , Microinyecciones , Ratas Endogámicas , Ratas Wistar , Restricción Física , Autoadministración , Estrés Psicológico/etiología
5.
Neurobiol Aging ; 56: 100-107, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28526294

RESUMEN

Aging populations are more sensitive to noxious stimuli as a result of altered somatosensory systems. In these experiments, we examined pain-like behaviors in young, middle-aged, and old mice during peripheral inflammation to determine if the same sensitivity exists in preclinical animal models. Immediately following injury, middle-aged and old mice exhibited more spontaneous pain-like behaviors than young mice, matching pain prevalence in clinical populations. Middle-aged and old mice also developed persistent mechanical hypersensitivity in the injured paw. Furthermore, old mice developed mechanical hypersensitivity in the noninjured paw suggesting age-dependent changes in central nociceptive systems. To address this end, pain-related protein expression was examined in the central nucleus of the amygdala, a limbic brain region that modulates somatic pain. Following injury, increased phosphorylation of extracellular signal-regulated kinase 1, a protein with known nociceptive functions, was observed in the right central nucleus of the amygdala of old mice and not middle-aged or young animals. These findings suggest that age-dependent changes in supraspinal nociceptive systems may account for increased pain-like behaviors in aging populations.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/fisiología , Núcleo Amigdalino Central/enzimología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nocicepción/fisiología , Dolor , Animales , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Fosforilación , Estimulación Física , Receptor del Glutamato Metabotropico 5/metabolismo , Corteza Somatosensorial/fisiopatología
6.
J Pharmacol Exp Ther ; 359(1): 82-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27451410

RESUMEN

Opioid-induced hyperalgesia (OIH) is a less-studied phenomenon that has been reported in both preclinical and clinical studies. Although the underlying cause is not entirely understood, OIH is a real-life problem that affects millions of patients on a daily basis. Research has implicated the important contribution of Ca(2+)/calmodulin-dependent protein kinase IIα (CaMKIIα) to OIH at the level of spinal nociceptors. To expand our understanding of the entire brain circuitry driving OIH, in this study we investigated the role of CaMKIIα in the laterocapcular division of the central amygdala (CeLC), the conjunctive point between the spinal cord and rostro-ventral medulla. OIH was produced by repeated fentanyl administration in the rat. Correlating with the development of mechanical allodynia and thermal hyperalgesia, CaMKIIα activity was significantly elevated in the CeLC in OIH. In addition, the frequency and amplitude of spontaneous miniature excitatory postsynaptic currents (mEPSCs) in CeLC neurons were significantly increased in OIH. 2-[N-(2-hidroxyethyl)-N-(4-methoxy-benzenesulfonyl)]-amino-N-(4-chlorocinnamyl)-N-methylbenzylamine, a CaMKIIα inhibitor, dose dependently reversed sensory hypersensitivity, activation of CeLC CaMKIIα, and mEPSCs in OIH. Taken together, our data for the first time implicate a critical role of CeLC CaMKIIα in OIH.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Núcleo Amigdalino Central/efectos de los fármacos , Núcleo Amigdalino Central/enzimología , Fentanilo/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Bencilaminas/farmacología , Bencilaminas/uso terapéutico , Relación Dosis-Respuesta a Droga , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Hiperalgesia/enzimología , Hiperalgesia/patología , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
7.
Neuropharmacology ; 107: 1-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26946429

RESUMEN

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that is expressed in the brain and implicated in alcohol abuse in humans and behavioral responses to ethanol in mice. Previous studies have shown an association of human ALK with acute responses to alcohol and alcohol dependence. In addition, Alk knockout (Alk -/-) mice consume more ethanol in a binge-drinking test and show increased sensitivity to ethanol sedation. However, the function of ALK in excessive drinking following the establishment of ethanol dependence has not been examined. In this study, we tested Alk -/- mice for dependence-induced drinking using the chronic intermittent ethanol-two bottle choice drinking (CIE-2BC) protocol. We found that Alk -/- mice initially consume more ethanol prior to CIE exposure, but do not escalate ethanol consumption after exposure, suggesting that ALK may promote the escalation of drinking after ethanol dependence. To determine the mechanism(s) responsible for this behavioral phenotype we used an electrophysiological approach to examine GABA neurotransmission in the central nucleus of the amygdala (CeA), a brain region that regulates alcohol consumption and shows increased GABA signaling after chronic ethanol exposure. GABA transmission in ethanol-naïve Alk -/- mice was enhanced at baseline and potentiated in response to acute ethanol application when compared to wild-type (Alk +/+) mice. Moreover, basal GABA transmission was not elevated by CIE exposure in Alk -/- mice as it was in Alk +/+ mice. These data suggest that ALK plays a role in dependence-induced drinking and the regulation of presynaptic GABA release in the CeA.


Asunto(s)
Alcoholismo/enzimología , Proteínas Tirosina Quinasas Receptoras/deficiencia , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Quinasa de Linfoma Anaplásico , Animales , Núcleo Amigdalino Central/efectos de los fármacos , Núcleo Amigdalino Central/enzimología , Depresores del Sistema Nervioso Central/administración & dosificación , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Estudios de Cohortes , Etanol/administración & dosificación , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Proteínas Tirosina Quinasas Receptoras/genética , Técnicas de Cultivo de Tejidos
8.
Epilepsy Behav ; 51: 96-103, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26262937

RESUMEN

The prevalence of depression and suicide is increased in patients with mesial temporal lobe epilepsy (MTLE); however, the underlying mechanism remains unknown. Anhedonia, a core symptom of depression that is predictive of suicide, is common in patients with MTLE. Glutamine synthetase, an astrocytic enzyme that metabolizes glutamate and ammonia to glutamine, is reduced in the amygdala in patients with epilepsy and depression and in suicide victims. Here, we sought to develop a novel model of anhedonia in MTLE by testing the hypothesis that deficiency in glutamine synthetase in the central nucleus of the amygdala (CeA) leads to epilepsy and comorbid anhedonia. Nineteen male Sprague-Dawley rats were implanted with an osmotic pump infusing either the glutamine synthetase inhibitor methionine sulfoximine [MSO (n=12)] or phosphate buffered saline [PBS (n=7)] into the right CeA. Seizure activity was monitored by video-intracranial electroencephalogram (EEG) recordings for 21days after the onset of MSO infusion. Sucrose preference, a measure of anhedonia, was assessed after 21days. Methionine sulfoximine-infused rats exhibited recurrent seizures during the monitoring period and showed decreased sucrose preference over days when compared with PBS-infused rats (p<0.01). Water consumption did not differ between the PBS-treated group and the MSO-treated group. Neurons were lost in the CeA, but not the medial amygdala, lateral amygdala, basolateral amygdala, or the hilus of the dentate gyrus, in the MSO-treated rats. The results suggest that decreased glutamine synthetase activity in the CeA is a possible common cause of anhedonia and seizures in TLE. We propose that the MSO CeA model can be used for mechanistic studies that will lead to the development and testing of novel drugs to prevent seizures, depression, and suicide in patients with TLE.


Asunto(s)
Amígdala del Cerebelo/enzimología , Anhedonia/fisiología , Encéfalo/enzimología , Núcleo Amigdalino Central/enzimología , Epilepsia del Lóbulo Temporal/enzimología , Glutamato-Amoníaco Ligasa/deficiencia , Análisis de Varianza , Anhedonia/efectos de los fármacos , Animales , Encéfalo/fisiopatología , Comorbilidad , Trastorno Depresivo/enzimología , Modelos Animales de Enfermedad , Electroencefalografía , Inhibidores Enzimáticos/farmacología , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/fisiopatología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Hipocampo/fisiología , Masculino , Metionina Sulfoximina/farmacología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Convulsiones/enzimología
9.
J Neurosci Res ; 93(5): 714-21, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25502299

RESUMEN

Roman high (RHA)- and low (RLA)-avoidance rats are selectively bred for rapid vs. poor acquisition of active avoidance, respectively, and differ markedly in emotional reactivity, coping style, and behavioral and neurochemical responses to morphine and psychostimulants. Accordingly, acute cocaine induces more robust increments in locomotion and dopamine output in the nucleus accumbens shell (AcbSh) of RHA than of RLA rats. Cocaine induces short- and long-term neuronal plasticity via activation of the extracellular signal-regulated kinase (ERK) pathway. This study compares the effects of acute cocaine on ERK phosphorylation (pERK) in limbic brain areas of Roman rats. In RHA but not RLA rats, cocaine (5 mg/kg) increased pERK in the infralimbic prefrontal cortex and AcbSh, two areas involved in its acute effects, but did not modify pERK in the prelimbic prefrontal cortex and Acb core, which mediate the chronic effects of cocaine. Moreover, cocaine failed to affect pERK immunolabeling in the bed nucleus of stria terminalis pars lateralis and central amygdala of either line but increased it in the basolateral amygdala of RLA rats. These results extend to pERK expression previous findings on the greater sensitivity to acute cocaine of RHA vs. RLA rats and confirm the notion that genetic factors influence the differential responses of the Roman lines to addictive drugs. Moreover, they support the view that the Roman lines are a useful tool to investigate the molecular underpinnings of individual vulnerability to drug addiction.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Núcleo Amigdalino Central/efectos de los fármacos , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Corteza Prefrontal/efectos de los fármacos , Análisis de Varianza , Animales , Reacción de Prevención/fisiología , Núcleo Amigdalino Central/enzimología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Masculino , Fosforilación/efectos de los fármacos , Corteza Prefrontal/enzimología , Ratas , Ratas Endogámicas/fisiología
10.
Brain Res ; 1498: 85-94, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23295188

RESUMEN

The increase in PKCθ expression in the amygdala of rats fed high fat diet (HFD) has been related to the loss of the anorectic response to insulin injections into the central nucleus of the amygdala (CeA) in these animals. PKCθ overexpression in the CeA increases food intake, body weight and body fat and inhibits insulin stimulation of Akt signaling. To study the effects of bilateral overexpression of PKCθ in the CeA of rats on peripheral metabolism, rats were injected into the CeA or 3rd ventricle with a lentiviral (LV)-PKCθ construct or LV-Green fluorescent protein (GFP) construct as a control and fed either LFD or HFD. Insulin and glucose tolerance tests were undertaken and hepatic AMPK activation, Pepck, Srebp1c gene expression and lipid levels assayed. CeA LV-PKCθ injected rats increased food intake, body weight and body fat and increased hepatic, but not serum, triglyceride levels compared to control rats that received a CeA-LV-GFP construct. Hepatic AMP-kinase activity was reduced but expression of Pepck increased while serum insulin decreased, glucose tolerance improved and the hypoglycemic response to insulin was enhanced in CeA LV-PKCθ injected rats. In contrast, rats that received LV-PKCθ injections into the 3rd Ventricle did not show any changes in food intake or body weight although serum, but not hepatic, triglyceride levels were increased and glucose tolerance was impaired. The data suggest that activation of PKCθ in the CeA and hypothalamus have different effects on energy balance and peripheral metabolism and that insulin signaling in the amygdala regulates peripheral metabolism.


Asunto(s)
Núcleo Amigdalino Central/enzimología , Glucosa/metabolismo , Homeostasis/fisiología , Hipotálamo/enzimología , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Adenilato Quinasa/metabolismo , Tejido Adiposo/fisiología , Animales , Peso Corporal/fisiología , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Ingestión de Alimentos/fisiología , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoenzimas/genética , Lentivirus/genética , Hígado/metabolismo , Masculino , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C-theta , Ratas Sprague-Dawley , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA