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










Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 15(701): eabq7839, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37343080

RESUMEN

Ca2+/calmodulin-dependent protein kinase II (CaMKII) hyperactivity causes cardiac arrhythmias, a major source of morbidity and mortality worldwide. Despite proven benefits of CaMKII inhibition in numerous preclinical models of heart disease, translation of CaMKII antagonists into humans has been stymied by low potency, toxicity, and an enduring concern for adverse effects on cognition due to an established role of CaMKII in learning and memory. To address these challenges, we asked whether any clinically approved drugs, developed for other purposes, were potent CaMKII inhibitors. For this, we engineered an improved fluorescent reporter, CaMKAR (CaMKII activity reporter), which features superior sensitivity, kinetics, and tractability for high-throughput screening. Using this tool, we carried out a drug repurposing screen (4475 compounds in clinical use) in human cells expressing constitutively active CaMKII. This yielded five previously unrecognized CaMKII inhibitors with clinically relevant potency: ruxolitinib, baricitinib, silmitasertib, crenolanib, and abemaciclib. We found that ruxolitinib, an orally bioavailable and U.S. Food and Drug Administration-approved medication, inhibited CaMKII in cultured cardiomyocytes and in mice. Ruxolitinib abolished arrhythmogenesis in mouse and patient-derived models of CaMKII-driven arrhythmias. A 10-min pretreatment in vivo was sufficient to prevent catecholaminergic polymorphic ventricular tachycardia, a congenital source of pediatric cardiac arrest, and rescue atrial fibrillation, the most common clinical arrhythmia. At cardioprotective doses, ruxolitinib-treated mice did not show any adverse effects in established cognitive assays. Our results support further clinical investigation of ruxolitinib as a potential treatment for cardiac indications.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Cardiopatías , Animales , Niño , Humanos , Ratones , Arritmias Cardíacas , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cardiopatías/metabolismo , Miocitos Cardíacos/metabolismo , Pirazoles/farmacología
2.
FASEB J ; 36(6): e22347, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35579659

RESUMEN

C1q/TNF-related proteins (CTRP1-15) constitute a conserved group of secreted proteins of the C1q family with diverse functions. In vitro studies have shown that CTRP11/C1QL4 can inhibit adipogenesis, antagonize myoblast fusion, and promote testosterone synthesis and secretion. Whether CTRP11 is required for these processes in vivo remains unknown. Here, we show that knockout (KO) mice lacking CTRP11 have normal skeletal muscle mass and function, and testosterone level, suggesting that CTRP11 is dispensable for skeletal muscle development and testosterone production. We focused our analysis on whether this nutrient-responsive secreted protein plays a role in controlling sugar and fat metabolism. At baseline when mice are fed a standard chow, CTRP11 deficiency affects metabolic parameters in a sexually dimorphic manner. Only Ctrp11-KO female mice have significantly higher fasting serum ketones and reduced physical activity. In the refeeding phase following food withdrawal, Ctrp11-KO female mice have reduced food intake and increased metabolic rate and energy expenditure, highlighting CTRP11's role in fasting-refeeding response. When challenged with a high-fat diet to induce obesity and metabolic dysfunction, CTRP11 deficiency modestly exacerbates obesity-induced glucose intolerance, with more pronounced effects seen in Ctrp11-KO male mice. Switching to a low-fat diet after obesity induction results in greater fat loss in wild type relative to KO male mice, suggesting impaired response to obesity reversal and reduced metabolic flexibility in the absence of CTRP11. Collectively, our data provide genetic evidence for novel sex-dependent metabolic regulation by CTRP11, but note the overall modest contribution of CTRP11 to systemic energy homeostasis.


Asunto(s)
Complemento C1/metabolismo , Complemento C1q , Dieta Alta en Grasa , Animales , Complemento C1q/metabolismo , Metabolismo Energético/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Testosterona
3.
FASEB J ; 35(11): e21910, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34610176

RESUMEN

C1q/TNF-related protein (CTRP) family comprises fifteen highly conserved secretory proteins with diverse central and peripheral functions. In zebrafish, mouse, and human, CTRP4 is most highly expressed in the brain. We previously showed that CTRP4 is a metabolically responsive regulator of food intake and energy balance, and mice lacking CTRP4 exhibit sexually dimorphic changes in ingestive behaviors and systemic metabolism. Recent single-cell RNA sequencing also revealed Ctrp4/C1qtnf4 expression in diverse neuronal cell types across distinct anatomical brain regions, hinting at additional roles in the central nervous system not previously characterized. To uncover additional central functions of CTRP4, we subjected Ctrp4 knockout (KO) mice to a battery of behavioral tests. Relative to wild-type (WT) littermates, loss of CTRP4 does not alter exploratory, anxiety-, or depressive-like behaviors, motor function and balance, sensorimotor gating, novel object recognition, and spatial memory. While pain-sensing mechanisms in response to thermal stress and mild shock are intact, both male and female Ctrp4 KO mice have increased sensitivity to pain induced by higher-level shock, suggesting altered nociceptive function. Importantly, CTRP4 deficiency impairs hippocampal-dependent associative learning and memory as assessed by trace fear conditioning paradigm. This deficit is sex-dependent, affects only female mice, and is associated with altered expression of learning and memory genes (Arc, c-fos, and Pde4d) in the hippocampus and cortex. Altogether, our behavioral and gene expression analyses have uncovered novel aspects of the CTRP4 function and provided a physiological context to further investigate its mechanism of action in the central and peripheral nervous system.


Asunto(s)
Adipoquinas/genética , Expresión Génica , Técnicas de Inactivación de Genes/métodos , Aprendizaje por Laberinto , Memoria Espacial , Adipoquinas/metabolismo , Animales , Ansiedad/genética , Conducta Animal , Corteza Cerebelosa/metabolismo , Femenino , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Prueba de Desempeño de Rotación con Aceleración Constante
4.
Elife ; 92020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33084572

RESUMEN

In the hippocampus, a widely accepted model posits that the dentate gyrus improves learning and memory by enhancing discrimination between inputs. To test this model, we studied conditional knockout mice in which the vast majority of dentate granule cells (DGCs) fail to develop - including nearly all DGCs in the dorsal hippocampus - secondary to eliminating Wntless (Wls) in a subset of cortical progenitors with Gfap-Cre. Other cells in the Wlsfl/-;Gfap-Cre hippocampus were minimally affected, as determined by single nucleus RNA sequencing. CA3 pyramidal cells, the targets of DGC-derived mossy fibers, exhibited normal morphologies with a small reduction in the numbers of synaptic spines. Wlsfl/-;Gfap-Cre mice have a modest performance decrement in several complex spatial tasks, including active place avoidance. They were also modestly impaired in one simpler spatial task, finding a visible platform in the Morris water maze. These experiments support a role for DGCs in enhancing spatial learning and memory.


Asunto(s)
Reacción de Prevención , Giro Dentado/anomalías , Memoria , Receptores Acoplados a Proteínas G/genética , Aprendizaje Espacial , Animales , Giro Dentado/crecimiento & desarrollo , Giro Dentado/fisiopatología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Prueba del Laberinto Acuático de Morris , Receptores Acoplados a Proteínas G/metabolismo , Análisis de Secuencia de ARN
5.
Hum Mol Genet ; 29(17): 2936-2950, 2020 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-32803234

RESUMEN

Our understanding of the contribution of genetic risk factors to neuropsychiatric diseases is limited to abnormal neurodevelopment and neuronal dysfunction. Much less is known about the mechanisms whereby risk variants could affect the physiology of glial cells. Our prior studies have shown that a mutant (dominant-negative) form of a rare but highly penetrant psychiatric risk factor, Disrupted-In-Schizophrenia-1 (DISC1), impairs metabolic functions of astrocytes and leads to cognitive dysfunction. In order to overcome the limitations of the mutant DISC1 model and understand the putative regional properties of astrocyte DISC1, we assessed whether knockdown of Disc1 (Disc1-KD) in mature mouse astrocytes of the prefrontal cortex (PFC) or the hippocampus would produce behavioral abnormalities that could be attributed to astrocyte bioenergetics. We found that Disc1-KD in the hippocampus but not PFC impaired trace fear conditioning in adult mice. Using the innovative deep learning approach and convolutional deep neural networks (cDNNs), ResNet50 or ResNet18, and single cell-based analysis, we found that Disc1-KD decreased the spatial density of astrocytes associated with abnormal levels and distribution of the mitochondrial markers and the glutamate transporter, GLAST. Disc1-KD in astrocytes also led to decreased expression of the glutamatergic and increased expression of the GABA-ergic synaptic markers, possibly via non-apoptotic activation of caspase 3 in neurons located within the individual territories of Disc1-KD astrocytes. Our results indicate that altered expression of DISC1 in astrocytes could impair astrocyte bioenergetics, leading to abnormalities in synaptic neurotransmission and cognitive function in a region-dependent fashion.


Asunto(s)
Encéfalo/metabolismo , Cognición/fisiología , Degeneración Nerviosa/genética , Proteínas del Tejido Nervioso/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/patología , Mapeo Encefálico , Aprendizaje Profundo , Técnicas de Silenciamiento del Gen , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Ratones , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Red Nerviosa/patología , Neuroglía/metabolismo , Neuroglía/patología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología
6.
FASEB J ; 33(12): 14734-14747, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31689372

RESUMEN

Cytokines and chemokines play diverse roles in different organ systems. Family with sequence similarity 19, member A1-5 (FAM19A1-A5; also known as TAFA1-5) is a group of conserved chemokine-like proteins enriched in the CNS of mice and humans. Their functions are only beginning to emerge. Here, we show that the expression of Fam19a1-a5 in different mouse brain regions are induced or suppressed by unfed and refed states. The striking nutritional regulation of Fam19a family members in the brain suggests a potential central role in regulating metabolism. Using a knockout (KO) mouse model, we show that loss of FAM19A1 results in sexually dimorphic phenotypes. In male mice, FAM19A1 deficiency alters food intake patterns during the light and dark cycle. Fam19a1 KO mice are hyperactive, and locomotor hyperactivity is more pronounced in female KO mice. Behavior tests indicate that Fam19a1 KO female mice have reduced anxiety and sensitivity to pain. Spatial learning and exploration, however, is preserved in Fam19a1 KO mice. Altered behaviors are associated with elevated norepinephrine and dopamine turnover in the striatum. Our results establish an in vivo function of FAM19A1 and highlight central roles for this family of neurokines in modulating animal physiology and behavior.-Lei, X., Liu, L., Terrillion, C. E., Karuppagounder, S. S., Cisternas, P., Lay, M., Martinelli, D. C., Aja, S., Dong, X., Pletnikov, M. V., Wong, G. W. FAM19A1, a brain-enriched and metabolically responsive neurokine, regulates food intake patterns and mouse behaviors.


Asunto(s)
Quimiocinas/fisiología , Cuerpo Estriado/metabolismo , Ingestión de Alimentos , Locomoción , Aprendizaje Espacial , Animales , Células Cultivadas , Quimiocinas/genética , Dopamina/metabolismo , Conducta Exploratoria , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Ratas , Factores Sexuales
8.
Neuropsychopharmacology ; 42(11): 2242-2251, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28631721

RESUMEN

The functional role of genetic variants in glia in the pathogenesis of psychiatric disorders remains poorly studied. Disrupted-In-Schizophrenia 1 (DISC1), a genetic risk factor implicated in major mental disorders, has been implicated in regulation of astrocyte functions. As both astrocytes and DISC1 influence adult neurogenesis in the dentate gyrus (DG) of the hippocampus, we hypothesized that selective expression of dominant-negative C-terminus-truncated human DISC1 (mutant DISC1) in astrocytes would affect adult hippocampal neurogenesis and hippocampus-dependent behaviors. A series of behavioral tests were performed in mice with or without expression of mutant DISC1 in astrocytes during late postnatal development. In conjunction with behavioral tests, we evaluated adult neurogenesis, including neural progenitor proliferation and dendrite development of newborn neurons in the DG. The ameliorative effects of D-serine on mutant DISC1-associated behaviors and abnormal adult neurogenesis were also examined. Expression of mutant DISC1 in astrocytes decreased neural progenitor proliferation and dendrite growth of newborn neurons, and produced elevated anxiety, attenuated social behaviors, and impaired hippocampus-dependent learning and memory. Chronic treatment with D-serine ameliorated the behavioral alterations and rescued abnormal adult neurogenesis in mutant DISC1 mice. Our findings suggest that psychiatric genetic risk factors expressed in astrocytes could affect adult hippocampal neurogenesis and contribute to aspects of psychiatric disease through abnormal production of D-serine.


Asunto(s)
Astrocitos/metabolismo , Conducta Animal/fisiología , Hipocampo/citología , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Animales , Animales Recién Nacidos , Ansiedad/tratamiento farmacológico , Ansiedad/genética , Ansiedad/patología , Astrocitos/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Serina/farmacología
9.
Neurobiol Dis ; 103: 144-153, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28392471

RESUMEN

In addition to motor function, the cerebellum has been implicated in cognitive and social behaviors. Various structural and functional abnormalities of Purkinje cells (PCs) have been observed in schizophrenia and autism. As PCs express the gene Disrupted-In-Schizophrenia-1 (DISC1), and DISC1 variants have been associated with neurodevelopmental disorders, we evaluated the role of DISC1 in cerebellar physiology and associated behaviors using a mouse model of inducible and selective expression of a dominant-negative, C-terminus truncated human DISC1 (mutant DISC1) in PCs. Mutant DISC1 male mice demonstrated impaired social and novel placement recognition. No group differences were found in novelty-induced hyperactivity, elevated plus maze test, spontaneous alternation, spatial recognition in Y maze, sociability or accelerated rotarod. Expression of mutant DISC1 was associated with a decreased number of large somata PCs (volume: 3000-5000µm3) and an increased number of smaller somata PCs (volume: 750-1000µm3) without affecting the total number of PCs or the volume of the cerebellum. Compared to control mice, attached loose patch recordings of PCs in mutant DISC1 mice revealed increased spontaneous firing of PCs; and whole cell recordings showed increased amplitude and frequency of mEPSCs without significant changes in either Rinput or parallel fiber EPSC paired-pulse ratio. Our findings indicate that mutant DISC1 alters the physiology of PCs, possibly leading to abnormal recognition memory in mice.


Asunto(s)
Disfunción Cognitiva/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Locomoción/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Células de Purkinje/metabolismo , Conducta Social , Animales , Disfunción Cognitiva/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética
10.
Int J Neuropsychopharmacol ; 20(5): 428-433, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28165117

RESUMEN

Background: Polymorphisms in the CACNA1C gene are associated with human mood disorders. The rodent social defeat model of stress/mood-disorder susceptibility results in maladaptive consequences mediated by altered function of mesolimbic circuits. Methods: mRNA levels of Cacna1c in the nucleus accumbens of mice exposed to social defeat were assessed. Cacna1c was selectively deleted in the nucleus accumbens of floxed Cacna1c mice using viral Cre-recombinase to examine Cacna1c in social defeat susceptibility. Results: Reduced expression of Cacan1c in the nucleus accumbens is associated with increased susceptibility to social defeat stress, and a knockdown of Cacna1c in the nucleus accumbens significantly increases susceptibility measured by social interaction and female urine preference. Conclusions: Cacna1c reduction causally predisposes to the maladaptive outcomes of social stress. Normal Cacna1c function in the nucleus accumbens is crucial for resiliency to social stressors. Variations in expression of CACNA1C in the nucleus accumbens may mediate human risk for developing mood disorders and be a target for therapeutic intervention.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Predisposición Genética a la Enfermedad/genética , Núcleo Accumbens/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/patología , Animales , Ansiedad/genética , Canales de Calcio Tipo L/genética , Modelos Animales de Enfermedad , Dominación-Subordinación , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Olfato/genética , Transducción Genética
11.
Eur J Neurosci ; 42(8): 2499-507, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25989111

RESUMEN

Increased calcium influx through L-type voltage-gated calcium channels has been implicated in the neuronal dysfunction underlying age-related memory declines. The present study aimed to test the specific role of Cacna1c (which encodes Cav 1.2) in modulating age-related memory dysfunction. Short-term, spatial and contextual/emotional memory was evaluated in young and aged, wild-type as well as mice with one functional copy of Cacna1c (haploinsufficient), using the novel object recognition, Y-maze and passive avoidance tasks, respectively. Hippocampal expression of Cacna1c mRNA was measured by quantitative polymerase chain reaction. Ageing was associated with object recognition and contextual/emotional memory deficits, and a significant increase in hippocampal Cacna1c mRNA expression. Cacna1c haploinsufficiency was associated with decreased Cacna1c mRNA expression in both young and old animals. However, haploinsufficient mice did not manifest an age-related increase in expression of this gene. Behaviourally, Cacna1c haploinsufficiency prevented object recognition deficits during ageing in both male and female mice. A significant correlation between higher Cacna1c levels and decreased object recognition performance was observed in both sexes. Also, a sex-dependent protective role of decreased Cacna1c levels in contextual/emotional memory loss has been observed, specifically in male mice. These data provide evidence for an association between increased hippocampal Cacna1c expression and age-related cognitive decline. Additionally, they indicate an interaction between the Cacna1c gene and sex in the modulation of age-related contextual memory declines.


Asunto(s)
Envejecimiento/fisiología , Envejecimiento/psicología , Canales de Calcio Tipo L/metabolismo , Trastornos del Conocimiento/fisiopatología , Trastornos de la Memoria/fisiopatología , Caracteres Sexuales , Animales , Reacción de Prevención/fisiología , Canales de Calcio Tipo L/genética , Cognición/fisiología , Emociones/fisiología , Femenino , Haploinsuficiencia , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/metabolismo , Reconocimiento en Psicología/fisiología , Memoria Espacial/fisiología
12.
PLoS One ; 9(1): e87513, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24475300

RESUMEN

Huntington's Disease (HD) is a neurodegenerative disorder that is caused by abnormal expansion of a polyglutamine tract in huntingtin (htt) protein. The expansion leads to increased htt aggregation and toxicity. Factors that aid in the clearance of mutant huntingtin proteins should relieve the toxicity. We previously demonstrated that overexpression of ubiqulin-1, which facilitates protein clearance through the proteasome and autophagy pathways, reduces huntingtin aggregates and toxicity in mammalian cell and invertebrate models of HD. Here we tested whether overexpression of ubiquilin-1 delays or prevents neurodegeneration in R6/2 mice, a well-established model of HD. We generated transgenic mice overexpressing human ubiquilin-1 driven by the neuron-specific Thy1.2 promoter. Immunoblotting and immunohistochemistry revealed robust and widespread overexpression of ubiquilin-1 in the brains of the transgenic mice. Similar analysis of R6/2 animals revealed that ubiquilin is localized in huntingtin aggregates and that ubiquilin levels decrease progressively to 30% during the end-stage of disease. We crossed our ubiquilin-1 transgenic line with R6/2 mice to assess whether restoration of ubiquilin levels would delay HD symptoms and pathology. In the double transgenic progeny, ubiquilin levels were fully restored, and this correlated with a 20% increase in lifespan and a reduction in htt inclusions in the hippocampus and cortex. Furthermore, immunoblots indicated that endoplasmic reticulum stress response that is elevated in the hippocampus of R6/2 animals was attenuated by ubiquilin-1 overexpression. However, ubiquilin-1 overexpression neither altered the load of htt aggregates in the striatum nor improved motor impairments in the mice.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Análisis de Varianza , Animales , Proteínas Relacionadas con la Autofagia , Encéfalo/patología , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Cruzamientos Genéticos , Cartilla de ADN/genética , Humanos , Proteína Huntingtina , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Transgénicos , Prueba de Desempeño de Rotación con Aceleración Constante
13.
Diabetes ; 61(11): 2833-41, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22751689

RESUMEN

Maternal high-fat (HF) diet throughout gestation and suckling has long-term consequences on the offspring's metabolic phenotype. Here we determine the relative contribution of pre- or postnatal maternal HF diet on offspring's metabolic phenotype. Pregnant Sprague-Dawley rats were maintained on normal chow or HF diet throughout gestation and suckling. All litters were cross-fostered to chow or HF dams on postnatal day (PND)1, resulting in four groups. Body weight, body composition, and glucose tolerance were measured at weaning and in adulthood. Leptin sensitivity was assessed by signal transducer and activator of transcription (STAT)3 activation on PND10 and PND21. Pups cross-fostered to HF dams gained more body weight than chow pups by PND7 and persisted until weaning. Postnatal HF pups had greater adiposity, higher plasma leptin concentration, impaired glucose tolerance, and reduced phosphorylated STAT3 in response to leptin in the arcuate nucleus at weaning. After weaning, male offspring cross-fostered to HF dams were hyperphagic and maintained greater body weight than postnatal chow pups. Postnatal HF diet during suckling continued to impair glucose tolerance in male and female offspring in adulthood. Maternal HF diet during suckling has a greater influence in determining offspring's metabolic phenotype than prenatal HF diet exposure and could provide insight regarding optimal perinatal nutrition for mothers and children.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/etiología , Lactancia , Leptina/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Obesidad/etiología , Obesidad/metabolismo , Adiposidad , Animales , Animales Lactantes , Núcleo Arqueado del Hipotálamo/metabolismo , Susceptibilidad a Enfermedades , Femenino , Leptina/sangre , Metabolismo de los Lípidos , Masculino , Leche/metabolismo , Obesidad/sangre , Obesidad/fisiopatología , Fosforilación , Embarazo , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Caracteres Sexuales
14.
Prog Neurobiol ; 99(1): 1-14, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705413

RESUMEN

One of the most consistent genetic findings to have emerged from bipolar disorder genome wide association studies (GWAS) is with CACNA1C, a gene that codes for the α(1C) subunit of the Ca(v)1.2 voltage-dependent L-type calcium channel (LTCC). Genetic variation in CACNA1C have also been associated with depression, schizophrenia, autism spectrum disorders, as well as changes in brain function and structure in control subjects who have no diagnosable psychiatric illness. These data are consistent with a continuum of shared neurobiological vulnerability between diverse-Diagnostic and Statistical Manual (DSM) defined-neuropsychiatric diseases. While involved in numerous cellular functions, Ca(v)1.2 is most frequently implicated in coupling of cell membrane depolarization to transient increase of the membrane permeability for calcium, leading to activation and, potentially, changes in intracellular signaling pathway activity, gene transcription, and synaptic plasticity. Ca(v)1.2 is involved in the proper function of numerous neurological circuits including those involving the hippocampus, amygdala, and mesolimbic reward system, which are strongly implicated in psychiatric disease pathophysiology. A number of behavioral effects of LTCC inhibitors have been described including antidepressant-like behavioral actions in rodent models. Clinical studies suggest possible treatment effects in a subset of patients with mood disorders. We review the genetic structure and variation of CACNA1C, discussing relevant human genetic and clinical findings, as well as the biological actions of Ca(v)1.2 that are most relevant to psychiatric illness.


Asunto(s)
Canales de Calcio Tipo L/genética , Variación Genética/genética , Trastornos Mentales/genética , Trastornos Mentales/fisiopatología , Humanos
15.
J Vis Exp ; (59): e3638, 2012 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-22314943

RESUMEN

The forced swim test is a rodent behavioral test used for evaluation of antidepressant drugs, antidepressant efficacy of new compounds, and experimental manipulations that are aimed at rendering or preventing depressive-like states. Mice are placed in an inescapable transparent tank that is filled with water and their escape related mobility behavior is measured. The forced swim test is straightforward to conduct reliably and it requires minimal specialized equipment. Successful implementation of the forced swim test requires adherence to certain procedural details and minimization of unwarranted stress to the mice. In the protocol description and the accompanying video, we explain how to conduct the mouse version of this test with emphasis on potential pitfalls that may be detrimental to interpretation of results and how to avoid them. Additionally, we explain how the behaviors manifested in the test are assessed.


Asunto(s)
Conducta Animal , Evaluación Preclínica de Medicamentos/métodos , Natación , Animales , Ratones , Estrés Psicológico
16.
J Vis Exp ; (59): e3769, 2012 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-22315011

RESUMEN

The tail-suspension test is a mouse behavioral test useful in the screening of potential antidepressant drugs, and assessing of other manipulations that are expected to affect depression related behaviors. Mice are suspended by their tails with tape, in such a position that it cannot escape or hold on to nearby surfaces. During this test, typically six minutes in duration, the resulting escape oriented behaviors are quantified. The tail-suspension test is a valuable tool in drug discovery for high-throughput screening of prospective antidepressant compounds. Here, we describe the details required for implementation of this test with additional emphasis on potential problems that may occur and how to avoid them. We also offer a solution to the tail climbing behavior, a common problem that renders this test useless in some mouse strains, such as the widely used C57BL/6. Specifically, we prevent tail climbing behaviors by passing mouse tails through a small plastic cylinder prior to suspension. Finally, we detail how to manually score the behaviors that are manifested in this test.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Suspensión Trasera/métodos , Animales , Conducta Animal , Ratones , Ratones Endogámicos C57BL , Estrés Psicológico
17.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1459-67, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21368270

RESUMEN

We have previously demonstrated that running-wheel access normalizes the food intake and body weight of Otsuka Long-Evens Tokushima Fatty (OLETF) rats. Following 6 wk of running-wheel access beginning at 8 wk of age, the body weight of OLETF rats remains reduced, demonstrating a lasting effect on their phenotype. In contrast, access to a high-fat diet exacerbates the hyperphagia and obesity of OLETF rats. To determine whether diet modulates the long-term effects of exercise, we examined the effects of high-fat diet on food intake and body weight in OLETF rats that had prior access to running wheels for 4 wk. We found that 4 wk of running exercise significantly decreased food intake and body weight of OLETF rats. Consistent with prior results, 4 wk of exercise also produced long-lasting effects on food intake and body weight in OLETF rats fed a regular chow. When running wheels were relocked, OLETF rats stabilized at lower levels of body weight than sedentary OLETF rats. However, access to a high-fat diet offset these effects. When OLETF rats were switched to a high-fat diet following wheel relocking, they significantly increased food intake and body weight, so that they reached levels similar to those of sedentary OLETF rats fed a high-fat diet. Gene expression determination of hypothalamic neuropeptides revealed changes that appeared to be appropriate responses to the effects of diet and running exercise. Together, these results demonstrate that high-fat diet modulates the long-lasting effects of exercise on food intake and body weight in OLETF rats.


Asunto(s)
Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Grasas de la Dieta/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Condicionamiento Físico Animal/fisiología , Tejido Adiposo/fisiopatología , Proteína Relacionada con Agouti/metabolismo , Animales , Hormona Liberadora de Corticotropina/metabolismo , Diabetes Mellitus Tipo 2/sangre , Modelos Animales de Enfermedad , Intolerancia a la Glucosa/fisiopatología , Hipotálamo/metabolismo , Leptina/sangre , Masculino , Proopiomelanocortina/metabolismo , Ratas , Ratas Endogámicas OLETF , Factores de Tiempo
18.
Obesity (Silver Spring) ; 18(5): 904-10, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20134410

RESUMEN

Intestinal nutrient infusions result in variable decreases in food intake and body weight based on the nutrient type and the specific intestinal infusion site. Only intrajejunal infusions of fatty acids decrease food intake beyond the calories infused. To test whether this extra-compensatory decrease in food intake is specific to fatty acids, small volume intrajejunal infusions of glucose (Glu) and casein hydrolysate (Cas), as well as linoleic acid (LA) were administered to male Sprague-Dawley rats. Equal kilocalorie (kcal) loads of these nutrients (11.4) or vehicle were infused into the jejunum over 7 h/day for five consecutive days. Food intake was continuously monitored and body weight was measured daily. After the infusion on the final day, rats were killed and plasma collected. Intrajejunal infusions of LA and Glu, but not Cas, suppressed food intake beyond the caloric load of the infusate with no compensatory increase in food intake after the infusion period. Rats receiving LA and Glu infusions also lost significant body weight across the infusion days. Plasma glucagon-like peptide-1 (GLP-1) was increased in both the LA and Glu rats compared with control animals, with no significant change in the Cas-infused animals. Peptide YY (PYY) levels increased in response to LA and Cas infusions. These results suggest that intrajejunal infusions of LA and Glu may decrease food intake and body weight via alterations in GLP-1 signaling. Thus, particular nutrients are more effective at producing decreases in food intake, body weight, and inducing changes in peptide levels and could lead to a novel therapy for obesity.


Asunto(s)
Peso Corporal/efectos de los fármacos , Caseínas/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Glucosa/administración & dosificación , Yeyuno/efectos de los fármacos , Ácido Linoleico/administración & dosificación , Análisis de Varianza , Animales , Ingestión de Energía/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Péptido 1 Similar al Glucagón/sangre , Leptina/sangre , Masculino , Péptido YY/sangre , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley
19.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R622-31, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19535681

RESUMEN

Repetitive cycles of palatable food access and chronic calorie restriction alter feeding behaviors and forebrain neural systems. The purpose of this study was to determine the behavioral, endocrine, and meal-related hindbrain neural activation in adult male Sprague-Dawley rats exposed to a binge-access feeding schedule. The binge-access schedule consisted of repeated twice-per-week episodes of acute calorie restriction (to one-third of the previous day's intake) followed by 2 h of concurrent access to high-calorie palatable food (sweetened fat: 90% vegetable shortening-10% sucrose) and chow. The binge-access rats consumed more calories during the "binge" period than rats with continuous access to sweetened fat (continuous-access group) or subjected to repeated acute calorie restriction only (chow-restricted group). The binge-access group also exhibited a approximately 25% increase in sweetened fat intake from week 1 to week 6. Persistence of the binge phenotype in the binge-access animals was demonstrated 2 wk, but not 4 wk, after ad libitum chow. The binge-access and chow-restricted groups maintained a similar normal body composition and hormonal profiles, whereas the continuous-access animals developed an obese phenotype. Terminal ghrelin levels were significantly higher in the binge-access group than in the continuous-access group. Consumption of a standardized meal resulted in more c-Fos-positive cells along the anterior-posterior nucleus of the solitary tract regions in the binge-access group than in naive controls. These results suggest that repeated cycles of acute calorie restriction followed by palatable food produce physiological alterations that may facilitate overconsumption of a highly palatable food during limited-access periods.


Asunto(s)
Bulimia/metabolismo , Conducta Alimentaria , Preferencias Alimentarias , Hormonas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Rombencéfalo/metabolismo , Tejido Adiposo/metabolismo , Animales , Peso Corporal , Bulimia/psicología , Restricción Calórica , Grasas de la Dieta/administración & dosificación , Sacarosa en la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Ingestión de Energía , Ghrelina/metabolismo , Masculino , Obesidad/metabolismo , Obesidad/psicología , Fenotipo , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/metabolismo , Factores de Tiempo
20.
Diabetes ; 58(5): 1116-25, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19188431

RESUMEN

OBJECTIVE: Perturbations to the prenatal environment have been associated with the development of adult chronic disease, findings that gave rise to the "Barker Hypothesis" or the "developmental origins of adult disease" concept. In this study, we used an animal model to determine the metabolic consequences of maternal prenatal stress and high-fat feeding on the developing offspring. RESEARCH DESIGN AND METHODS: Pregnant female Sprague-Dawley rats were maintained on standard chow or 60% high-fat diet throughout gestation and lactation. Half of each group were exposed to a novel variable stress paradigm during the 3rd week of gestation, whereas control dams were left undisturbed. Body weight, body composition, glucose tolerance, and endocrine parameters were measured in offspring through early adulthood. RESULTS: Male and female pups from dams that experienced prenatal stress and/or were on a high-fat diet weighed more beginning on postnatal day 7 compared with standard chow-control pups. Access to high-fat diet at weaning increased the body weight effect through early adulthood and was attributable to greater adiposity. Pups weaned onto standard chow diet showed no significant difference in glucose clearance or insulin secretion. However, pups weaned onto high-fat diet had impaired glucose tolerance if their dams were on a high-fat diet, experienced prenatal stress, or both. CONCLUSIONS: Our data demonstrate that prenatal stress and/or high-fat diet during the intrauterine or postnatal environment affects offspring in a manner that increases their susceptibility to diet-induced obesity and leads to secondary adverse metabolic consequences.


Asunto(s)
Grasas de la Dieta/efectos adversos , Obesidad/genética , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Niño , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Ingestión de Energía , Femenino , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Obesidad/epidemiología , Embarazo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...