Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Cell Neurosci ; 112: 103614, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33845123

RESUMEN

Homozygous Dab1 yotari mutant mice, Dab1yot (yot/yot) mice, have an autosomal recessive mutation of Dab1 and show reeler-like phenotype including histological abnormality of the cerebellum, hippocampus, and cerebral cortex. We here show abnormal hippocampal development of yot/yot mice where granule cells and pyramidal cells fail to form orderly rows but are dispersed diffusely in vague multiplicative layers. Possibly due to the positioning failure of granule cells and pyramidal cells and insufficient synaptogenesis, axons of the granule cells did not extend purposefully to connect with neighboring regions in yot/yot mice. We found that both hippocampal granule cells and pyramidal cells of yot/yot mice expressed proteins reactive with the anti-Dab1 antibody. We found that Y198- phosphorylated Dab1 of yot/yot mice was greatly decreased. Accordingly the downstream molecule, Akt was hardly phosphorylated. Especially, synapse formation was defective and the distribution of neurons was scattered in hippocampus of yot/yot mice. Some of neural cell adhesion molecules and hippocampus associated transcription factors of the neurons were expressed aberrantly, suggesting that the Reelin-Dab1 signaling pathway seemed to be importantly involved in not only neural migration as having been shown previously but also neural maturation and/or synaptogenesis of the mice. It is interesting to clarify whether the defective neural maturation is a direct consequence of the dysfunctional Dab1, or alternatively secondarily due to the Reelin-Dab1 intracellular signaling pathways.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Proteínas de la Matriz Extracelular/fisiología , Hipocampo/anomalías , Ratones Mutantes/anomalías , Proteínas del Tejido Nervioso/fisiología , Serina Endopeptidasas/fisiología , Transducción de Señal/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/deficiencia , Movimiento Celular , Activación Enzimática , Proteínas de la Matriz Extracelular/deficiencia , Genes Recesivos , Hipocampo/embriología , Hipocampo/metabolismo , Hipocampo/patología , Homocigoto , Ratones , Ratones Mutantes/genética , Ratones Mutantes/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Moléculas de Adhesión de Célula Nerviosa/genética , Fenotipo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Reelina , Serina Endopeptidasas/deficiencia , Sinapsis/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
2.
Methods Mol Biol ; 2128: 11-24, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180183

RESUMEN

Many animal models that are currently used in appetite and obesity research share at least some main features of human obesity and its comorbidities. Hence, even though no animal model replicates all aspects of "common" human obesity, animal models are imperative in studying the control of energy balance and reasons for its imbalance that may eventually lead to overt obesity. The most frequently used animal models are small rodents that may be based on mutations or manipulations of individual or several genes and on the exposure to obesogenic diets or other manipulations that predispose the animals to gaining or maintaining excessive weight. Characteristics include hyperphagia or changes in energy metabolism and at least in some models the frequent comorbidities of obesity, like hyperglycemia, insulin resistance, or diabetes-like syndromes. Some of the most frequently used animal models of obesity research involve animals with monogenic mutations of the leptin pathway which in fact are useful to study specific mechanistic aspects of eating controls, but typically do not recapitulate "common" obesity in the human population. Hence, this review will mention advantages and disadvantages of respective animal models in order to build a basis for the most appropriate use in biomedical research.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Obesidad/genética , Obesidad/metabolismo , Animales , Animales Modificados Genéticamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Dieta/efectos adversos , Modelos Animales de Enfermedad , Leptina/genética , Leptina/metabolismo , Ratones , Ratones Mutantes/metabolismo , Ratas , Ratas Mutantes/metabolismo
3.
Mol Neurobiol ; 57(5): 2301-2313, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32020500

RESUMEN

Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition with unknown etiology. Recent experimental evidences suggest the contribution of non-coding RNAs (ncRNAs) in the pathophysiology of ASD. In this work, we aimed to investigate the expression profile of the ncRNA class of circular RNAs (circRNAs) in the hippocampus of the BTBR T + tf/J (BTBR) mouse model and age-matched C57BL/6J (B6) mice. Alongside, we analyzed BTBR hippocampal gene expression profile to evaluate possible correlations between the differential abundance of circular and linear gene products. From RNA sequencing data, we identified circRNAs highly modulated in BTBR mice. Thirteen circRNAs and their corresponding linear isoforms were validated by RT-qPCR analysis. The BTBR-regulated circCdh9 was better characterized in terms of molecular structure and expression, highlighting altered levels not only in the hippocampus, but also in the cerebellum, prefrontal cortex, and amygdala. Finally, gene expression analysis of the BTBR hippocampus pinpointed altered biological and molecular pathways relevant for the ASD phenotype. By comparison of circRNA and gene expression profiles, we identified 6 genes significantly regulated at either circRNA or mRNA gene products, suggesting low overall correlation between circRNA and host gene expression. In conclusion, our results indicate a consistent deregulation of circRNA expression in the hippocampus of BTBR mice. ASD-related circRNAs should be considered in functional studies to identify their contribution to the etiology of the disorder. In addition, as abundant and highly stable molecules, circRNAs represent interesting potential biomarkers for autism.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones Endogámicos/metabolismo , Ratones Mutantes/metabolismo , ARN Circular/biosíntesis , ARN Mensajero/biosíntesis , Animales , Trastorno del Espectro Autista/genética , Química Encefálica , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos/genética , Ratones Mutantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie
4.
J Proteome Res ; 17(7): 2293-2306, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29873499

RESUMEN

In the present study, we performed a metabolomics analysis to evaluate a MODY5/RCAD mouse mutant line as a potential model for HNF1B-associated diseases. Gas chromatography time-of-flight mass spectrometry (GC-TOF-MS) of gut, kidney, liver, muscle, pancreas, and plasma samples uncovered the tissue specific metabolite distribution. Orthogonal projections to latent structures discriminant analysis (OPLS-DA) was used to identify the differences between MODY5/RCAD and wild-type mice in each of the tissues. The differences included, for example, increased levels of amino acids in the kidneys and reduced levels of fatty acids in the muscles of the MODY5/RCAD mice. Interestingly, campesterol was found in higher concentrations in the MODY5/RCAD mice, with a four-fold and three-fold increase in kidneys and pancreas, respectively. As expected, the MODY5/RCAD mice displayed signs of impaired renal function in addition to disturbed liver lipid metabolism, with increased lipid and fatty acid accumulation in the liver. From a metabolomics perspective, the MODY5/RCAD model was proven to display a metabolic pattern similar to what would be suspected in HNF1B-associated diseases. These findings were in line with the presumed outcome of the mutation based on the different anatomy and function of the tissues as well as the effect of the mutation on development.


Asunto(s)
Modelos Animales de Enfermedad , Metabolómica/métodos , Ratones Mutantes/metabolismo , Animales , Cadherinas/genética , Cromatografía de Gases y Espectrometría de Masas , Factor Nuclear 1-beta del Hepatocito/genética , Riñón/metabolismo , Hígado/metabolismo , Ratones , Páncreas/metabolismo
5.
Behav Pharmacol ; 28(6): 466-476, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28609327

RESUMEN

The C57BL6/J mouse is the most commonly used strain in genetic investigations and behavioural tests. However, only a few studies have used C57BL6/J mice to assess the effects of antidepressant compounds. We carried out a study to compare the behavioural effects of fluoxetine (FLX) in a model of depression in two mice strains: C57BL6/J and BALB/c. We used an 8-week unpredictable chronic mild stress (UCMS) protocol during which FLX was administered (15 mg/kg, oral) from the third week to the end of the protocol. We found that UCMS induced degradation of the coat state in the two strains. Moreover, as expected, we observed that FLX elicited antidepressant-like effects in the BALB/c mice by reducing the coat state deterioration and the latency of grooming in splash test. However, in the C57BL6/J mice, it did not induce this action, but instead triggered an opposite effect: an increased sniffing latency in the novelty suppression of feeding test. We conclude that FLX exerts a paradoxical effect in the C57Bl6/J strain. This observation is consistent with some clinical features of hyper-reactivity to FLX observed in humans. Therefore, the UCMS protocol used in C57Bl6/J mice could be a good model to study the mechanisms of the paradoxical effects caused by selective serotonin reuptake inhibitors.


Asunto(s)
Antidepresivos/farmacología , Fluoxetina/farmacología , Ratones Mutantes/metabolismo , Animales , Antidepresivos/metabolismo , Conducta Animal/efectos de los fármacos , Depresión/metabolismo , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/metabolismo , Modelos Animales de Enfermedad , Fluoxetina/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C/metabolismo , Ratones Endogámicos C57BL/metabolismo , Actividad Motora/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estrés Psicológico
6.
Transl Psychiatry ; 3: e263, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23715298

RESUMEN

The db/db mouse is a widely used preclinical model in diabetes research. Recent studies have shown that these mice also display aspects of psychosis and depression-like behaviors as seen in some psychiatric disorders. Here, we have performed multiplex immunoassay and liquid chromatography mass spectrometry profiling of the plasma and brain samples from db/db and control mice to identify altered pathways, which could be related to these behavioral abnormalities. This is the first study to carry out profiling of the brain proteome in this model. Plasma from the db/db mice had increased levels of leptin and insulin, decreased levels of peptide YY, glucagon and prolactin and alterations in inflammation-related proteins, compared with control mice. Frontal cortex tissue from the db/db mice showed changes in proteins involved in energy metabolism, cellular structure and neural functioning, and the hippocampus had changes in proteins involved in the same pathways, with additional effects on cellular signalling proteins. The overlap of these findings with effects seen in type 2 diabetes, schizophrenia, major depressive disorder and Alzheimer's disease might contribute to a common endophenotype seen in metabolic and neurological disorders.


Asunto(s)
Sistema Nervioso Central/metabolismo , Diabetes Mellitus Experimental/metabolismo , Ratones Mutantes/metabolismo , Animales , Modelos Animales de Enfermedad , Lóbulo Frontal/química , Lóbulo Frontal/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Hipocampo/química , Hipocampo/metabolismo , Inmunoensayo , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Proteómica
7.
J Biomed Biotechnol ; 2011: 130947, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22219655

RESUMEN

Modifier screening is a powerful genetic tool. While not widely used in the vertebrate system, we applied these tools to transgenic mouse strains that recapitulate key aspects of Alzheimer's disease (AD), such as tau-expressing mice. These are characterized by a robust pathology including both motor and memory impairment. The phenotype can be modulated by ENU mutagenesis, which results in novel mutant mouse strains and allows identifying the underlying gene/mutation. Here we discuss this strategy in detail. We firstly obtained pedigrees that modify the tau-related motor phenotype, with mapping ongoing. We further obtained transgene-independent motor pedigrees: (i) hyperactive, circling ENU 37 mice with a causal mutation in the Tbx1 gene-the complete knock-out of Tbx1 models DiGeorge Syndrome; (ii) ENU12/301 mice that show sudden jerky movements and tremor constantly; they have a causal mutation in the Kcnq1 gene, modelling aspects of the Romano-Ward and Jervell and Lange-Nielsen syndromes; and (iii) ENU16/069 mice with tremor and hypermetric gait that have a causal mutation in the Mpz (Myelin Protein Zero) gene, modelling Charcot-Marie-Tooth disease type 1 (CMT1B). Together, we provide evidence for a real potential of an ENU mutagenesis to dissect motor functions in wild-type and tau mutant mice.


Asunto(s)
Genes Modificadores/genética , Ratones Mutantes/genética , Trastornos del Movimiento/genética , Mutagénesis/genética , Mutación/genética , Proteínas tau/genética , Enfermedad de Alzheimer/genética , Animales , Marcha/genética , Hipercinesia/genética , Canal de Potasio KCNQ1/genética , Ratones , Ratones Mutantes/metabolismo , Proteína P0 de la Mielina/genética , Linaje , Fenotipo , Temblor/genética
8.
J Cell Mol Med ; 14(12): 2816-26, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19818092

RESUMEN

Morphine is a potent analgesic, but the molecular mechanism for tolerance formation after repeated use is not fully understood. Binding immunoglobulin protein (BiP) is an endoplasmic reticulum (ER) chaperone that is central to ER function. We examined knock-in mice expressing a mutant BiP with the retrieval sequence deleted in order to elucidate physiological processes that are sensitive to BiP functions. We tested the thermal antinociceptive effect of morphine in heterozygous mutant BiP mice in a hot plate test. Paw withdrawal latencies before and after a single administration of morphine were not significantly different between the wild-type and mutant BiP mice. Repeated morphine administration caused the development of morphine tolerance in the wild-type mice. The activation of glycogen synthase kinase 3b (GSK-3b) was associated with morphine tolerance, because an inhibitor of GSK-3ß prevented it. On the other hand, the mutant BiP mice showed less morphine tolerance, and the activation of GSK-3b was suppressed in their brain. These results suggest that BiP may play an important role in the development of morphine tolerance. Furthermore, we found that a chemical chaperone which improves ER protein folding capacity also attenuated the development of morphine tolerance in wild-type mice, suggesting a possible clinical application of chemical chaperones in preventing morphine tolerance.


Asunto(s)
Analgésicos/farmacología , Tolerancia a Medicamentos , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Analgésicos/administración & dosificación , Animales , Western Blotting , Chaperón BiP del Retículo Endoplásmico , Técnica del Anticuerpo Fluorescente , Técnicas de Sustitución del Gen , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas de Choque Térmico/genética , Ratones , Ratones Mutantes/metabolismo , Morfina/administración & dosificación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores Opioides mu/antagonistas & inhibidores , Eliminación de Secuencia , Transducción de Señal
9.
Lab Invest ; 90(1): 83-97, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19918242

RESUMEN

Reduced nephron numbers may predispose to renal failure. We hypothesized that glucose transporters (GLUTs) may contribute to progression of the renal disease, as GLUTs have been implicated in diabetic glomerulosclerosis and hypertensive renal disease with mesangial cell (MC) stretch. The Os (oligosyndactyly) allele that typically reduces nephron number by approximately 50%, was repeatedly backcrossed from ROP (Ra/+ (ragged), Os/+ (oligosyndactyly), and Pt/+ (pintail)) Os/+ mice more than six times into the Fvb mouse background to obtain Os/+ and +/+ mice with the Fvb background for study. Glomerular function, GLUT1, signaling, albumin excretion, and structural and ultrastructural changes were assessed. The FvbROP Os/+ mice (Fvb background) exhibited increased glomerular GLUT1, glucose uptake, VEGF, glomerular hypertrophy, hyperfiltration, extensive podocyte foot process effacement, marked albuminuria, severe extracellular matrix (ECM) protein deposition, and rapidly progressive renal failure leading to their early demise. Glomerular GLUT1 was increased 2.7-fold in the FvbROP Os/+ mice vs controls at 4 weeks of age, and glucose uptake was increased 2.7-fold. These changes were associated with the activation of glomerular PKCbeta1 and NF-kappaB p50 which contribute to ECM accumulation. The cyclic mechanical stretch of MCs in vitro, used as a model for increased MC stretch in vivo, reproduced increased GLUT1 at 48 h, a stimulus for increased VEGF expression which followed at 72 h. VEGF was also shown to act in a positive feedback manner on MC GLUT1, increasing GLUT1 expression, glucose uptake and fibronectin (FN) accumulation in vitro, whereas antisense suppression of GLUT1 largely blocked FN upregulation by VEGF. The FvbROP Os/+ mice exhibited an early increase in glomerular GLUT1 leading to increased glomerular glucose uptake PKCbeta1, and NF-kappaB activation, with excess ECM accumulation. A GLUT1-VEGF-GLUT1 positive feedback loop may play a key role in contributing to renal disease in this model of nondiabetic glomerulosclerosis.


Asunto(s)
Albuminuria/etiología , Transportador de Glucosa de Tipo 1/metabolismo , Ratones Mutantes/metabolismo , Nefronas/anomalías , Insuficiencia Renal/etiología , Insuficiencia Renal/fisiopatología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Alelos , Animales , Células Cultivadas , Creatinina/metabolismo , Progresión de la Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Mesangio Glomerular/metabolismo , Mesangio Glomerular/patología , Inmunohistoquímica , Isoenzimas/metabolismo , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Riñón/patología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes/genética , Microscopía Electrónica , FN-kappa B/metabolismo , Proteína Quinasa C/metabolismo , Estrés Mecánico , Sindactilia/genética , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba
10.
Invest Ophthalmol Vis Sci ; 49(7): 2863-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18344445

RESUMEN

PURPOSE: In previous studies of light-induced (LRD) and age-related (ageRD) retinal degeneration (RD) between the BALB/cByJ (BALB) and B6(Cg)-Tyr(c-2J)/J (B6a) albino mouse strains, RD-modifying quantitative trait loci (QTLs) were identified. After breeding BALB- and B6a-rd3/rd3 congenic strains and finding significant differences in RD, an F1 intercross to determine rd3 QTLs that influence this inherited RD was performed. METHODS: N10, F2 BALB- and B6a-rd3/rd3 strains were measured for retinal outer nuclear layer (ONL) thickness from 5 to 12 weeks of age. Since 10 weeks showed significant differences in the ONL, F2 progeny from an F1 intercross were measured for ONL thickness. F2 DNAs were genotyped for SNPs by the Center for Inherited Disease Research. Correlation of genotype with phenotype was made with Map Manager QTX. RESULTS: One hundred forty-eight SNPs approximately 10 cM apart were typed in the F2 progeny and analyzed. Significant QTLs were identified on chromosomes (Chrs) 17, 8, 14, and 6 (B6a alleles protective) and two on Chr 5 (BALB alleles protective). Suggestive QTLs were found as well. For the strongest QTLs, follow-up SNPs were analyzed to narrow the critical intervals. Additional studies demonstrated that rd3 disease is exacerbated by light but not protected by the absence of rhodopsin regeneration. CONCLUSIONS: QTLs were identified that modulate rd3-RD. These overlapped some QTLs from previous ageRD and LRD studies. The presence of some of the same QTLs in several studies suggests partial commonality in RD pathways. Identifying natural gene/alleles that modify RDs opens avenues of study that may lead to therapies for RD diseases.


Asunto(s)
Ratones Mutantes/genética , Ratones Mutantes/metabolismo , Sitios de Carácter Cuantitativo , Degeneración Retiniana/genética , Animales , Cromosomas de los Mamíferos , Ojo/metabolismo , Genotipo , Luz , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple , Retina/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Pigmentos Retinianos/metabolismo , Rodopsina/metabolismo , Especificidad de la Especie
11.
J Dermatol Sci ; 45(1): 55-62, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17123789

RESUMEN

BACKGROUND: The murine recessive yellow (Mc1r(e)) is a loss-of-function mutation in the receptor for alpha-melanocyte-stimulating hormone (MSH), melanocortin receptor 1 (MC1R), and produces yellow coats by inducing pheomelanin synthesis in hair follicular melanocytes. OBJECTIVE: It is not known whether the Mc1r(e) mutation affects pheomelanin synthesis in other skin sites. In this study, the eumelanin and pheomelanin contents in the epidermis and dermis as well as hairs of wild-type and mutant mice were measured. MATERIALS AND METHODS: The content of melanin was measured by high performance liquid chromatography. RESULTS: The eumelanin contents in the epidermis and dermis of newborn wild-type (Mc1r(+)/Mc1r(+)) mice (0.5, 3.5, 5.5, and 7.5 days) were much greater than those of mutant (Mc1r(e)/Mc1r(e)) mice, whereas the pheomelanin contents in the epidermis and dermis of mutant mice were much greater than those of wild-type mice. No sex differences in the contents of eumelanin and pheomalanin in the epidermis and dermis both in mutant and wild-type mice were observed. The eumelanin contents in mutant hairs (5-week-old) was much smaller than in wild-type hairs, whereas the pheomelanin contents in mutant hairs was much greater than in wild-type hairs. However, the eumelanin and pheomelanin contents in mutant female hairs were greater than in male. These sex differences were not observed in wild-type mice. CONCLUSION: The Mc1r(e) gene stimulates pheomelanin synthesis in the epidermis, dermis and hair follicles. In addition, eumelanin and pheomelanin contents in Mc1r(e)/Mc1r(e) hairs may be influenced by the sex difference.


Asunto(s)
Genes Recesivos , Color del Cabello/genética , Cabello/metabolismo , Melaninas/metabolismo , Ratones Mutantes/genética , Receptor de Melanocortina Tipo 1/genética , Animales , Animales Recién Nacidos , Cromatografía Líquida de Alta Presión , Dermis/metabolismo , Epidermis/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes/metabolismo , Mutación , Receptor de Melanocortina Tipo 1/metabolismo , Caracteres Sexuales
12.
Physiol Genomics ; 27(2): 131-40, 2006 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-16849632

RESUMEN

Tub is a member of a small gene family, the tubby-like proteins (TULPs), with predominant expression in neurons. Mice carrying a mutation in Tub develop retinal and cochlear degeneration as well as late-onset obesity with insulin resistance. During behavioral and metabolic testing, we found that homozygous C57BL/6J-Tub(tub) mice have a lower respiratory quotient than C57BL/6J controls before the onset of obesity, indicating that tubby homozygotes fail to activate carbohydrate metabolism and instead rely on fat metabolism for energy needs. In concordance with this, tubby mice show higher excretion of ketone bodies and accumulation of glycogen in the liver. Quantitation of liver mRNA levels shows that, during the transition from light to dark period, tubby mice fail to induce glucose-6-phosphate dehydrogenase (G6pdh), the rate-limiting enzyme in the pentose phosphate pathway that normally supplies NADPH for de novo fatty acid synthesis and glutathione reduction. Reduced G6PDH protein levels and enzymatic activity in tubby mice lead accordingly to lower levels of NADPH and reduced glutathione (GSH), respectively. mRNA levels for the lipolytic enzymes acetyl-CoA synthetase and carnitine palmitoyltransferase are increased during the dark cycle and decreased during the light period, and several citric acid cycle genes are dysregulated in tubby mice. Examination of hypothalamic gene expression showed high levels of preproorexin mRNA leading to accumulation of orexin peptide in the lateral hypothalamus. We hypothesize that abnormal hypothalamic orexin expression leads to changes in liver carbohydrate metabolism and may contribute to the moderate obesity observed in tubby mice.


Asunto(s)
Metabolismo de los Hidratos de Carbono/genética , Metabolismo Energético/genética , Ratones Mutantes/metabolismo , Proteínas/genética , Acetato CoA Ligasa/biosíntesis , Acetato CoA Ligasa/genética , Proteínas Adaptadoras Transductoras de Señales , Proteína Relacionada con Agouti , Animales , Química Encefálica , Dióxido de Carbono/metabolismo , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Ritmo Circadiano , Ciclo del Ácido Cítrico/genética , Enfermedades Cocleares/genética , Ingestión de Alimentos , Inducción Enzimática/genética , Genes Recesivos , Glucosafosfato Deshidrogenasa/biosíntesis , Glucosafosfato Deshidrogenasa/genética , Glutatión/deficiencia , Homocigoto , Hipotálamo/metabolismo , Resistencia a la Insulina/genética , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Metabolismo de los Lípidos , Lipólisis/genética , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes/genética , Actividad Motora , NADP/deficiencia , Neuropéptido Y/biosíntesis , Neuropéptido Y/genética , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Obesidad/genética , Orexinas , Oxígeno/metabolismo , Consumo de Oxígeno/genética , Vía de Pentosa Fosfato/genética , Proteínas/fisiología , Degeneración Retiniana/genética
13.
Comp Med ; 56(2): 96-104, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16639975

RESUMEN

The ability to move water across lipid membranes is crucial for nutrient intake, energy generation, waste excretion, and a myriad of other functions associated with life. Aquaporins, a family of integral membrane proteins, are now recognized as the channels responsible for transporting hydrophilic molecules, including water, across relatively impervious, hydrophobic cell membranes. A tremendous amount of work has been published on characterizing these proteins, which have been found in all bacteria, yeast, plants, and animals examined to date. In addition, an increasing number of mouse models with genetically altered aquaporin expression are being reported. This article will briefly review the basic biochemistry of aquaporins and then evaluate the use (and misuse) of mice in the quest for understanding the comparative pathophysiology of aquaporins in humans.


Asunto(s)
Acuaporinas/genética , Ratones Mutantes/genética , Animales , Acuaporinas/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes/metabolismo , Ratones Transgénicos , Mutación , Fenotipo , Agua/metabolismo
14.
Mol Endocrinol ; 20(7): 1610-22, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16513793

RESUMEN

Mutations in the human ALMS1 gene are responsible for Alström syndrome, a disorder in which key metabolic and endocrinological features include childhood-onset obesity, metabolic syndrome, and diabetes, as well as infertility. ALMS1 localizes to the basal bodies of cilia and plays a role in intracellular trafficking, but the biological functions of ALMS1 and how these relate to the pathogenesis of obesity, diabetes, and infertility remain unclear. Here we describe a new mouse model of Alström syndrome, fat aussie, caused by a spontaneous mutation in the Alms1 gene. Fat aussie (Alms1 foz/foz) mice are of normal weight when young but, by 120 d of age, they become obese and hyperinsulinemic. Diabetes develops in Alms1 foz/foz mice accompanied by pancreatic islet hyperplasia and islet cysts. Female mice are fertile before the onset of obesity and metabolic syndrome; however, male fat aussie mice are sterile due to a progressive germ cell loss followed by an almost complete block of development at the round-to-elongating spermatid stage of spermatogenesis. In conclusion, Alms1 foz/foz mouse is a new animal model in which to study the pathogenesis of the metabolic and fertility defects of Alström syndrome, including the role of ALMS1 in appetite regulation, pathogenesis of the metabolic syndrome, pancreatic islet physiology, and spermatogenesis.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Diabetes Mellitus Experimental/genética , Ratones Mutantes/genética , Modelos Animales , Obesidad/genética , Espermatogénesis/genética , Animales , Secuencia de Bases , Composición Corporal , Proteínas de Ciclo Celular , Proteínas de Unión al ADN/genética , Ingestión de Alimentos , Femenino , Mutación del Sistema de Lectura , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Mutantes/metabolismo , Ratones Mutantes/fisiología , Datos de Secuencia Molecular , Espermatogénesis/fisiología , Síndrome
15.
Zhong Xi Yi Jie He Xue Bao ; 4(1): 48-51, 2006 Jan.
Artículo en Chino | MEDLINE | ID: mdl-16409970

RESUMEN

OBJECTIVE: To investigate the effects of Naohuandan Recipe on learning and memory abilities of SAM-P/8 mice and its role in anti-oxidation and anti-apoptosis. METHODS: Forty SAM-P/8 mice were randomly divided into four groups, which were untreated (normal saline-treated) group, Yinkeluo Tablets (extracts of gingko leaf)-treated group, low-dose Naohuandan Recipe-treated group and high-dose Naohuandan Recipe-treated group. Mice in these groups were given corresponding drugs orally for 105 days. Then the performances of learning and memory of mice were tested by a step-down passive avoidance task and a Y-maze test. The serum levels of superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) were detected. The expression level of bcl-xl mRNA in cerebral cortex and hippocampus of mice was detected by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: The performances of learning and memory in the Yinkeluo Tablets-treated group, low- and high-dose Naohuandan Recipe-treated groups were significantly improved as compared with those in the untreated group (P<0.05 or P<0.01), and such performance was the best in the high-dose Naohuandan Recipe-treated group among these four groups (P<0.01). The serum levels of SOD and GSH-Px and the expression of bcl-xl mRNA in cerebral cortex and hippocampus of mice in the Yinkeluo Tablets-treated group, low- and high-dose Naohuandan Recipe-treated groups were also significantly higher than those in the untreated group (P<0.05 or P<0.01), while the serum level of MDA in the untreated group was higher than that in the other three groups (P<0.01). CONCLUSION: Naohuandan Recipe can improve learning and memory abilities of SAM-P/8 mice, and this effect may be related to its anti-oxidation efficacy and enhancement of expression level of bcl-xl mRNA.


Asunto(s)
Envejecimiento/metabolismo , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Aprendizaje/efectos de los fármacos , Envejecimiento/genética , Animales , Glutatión Peroxidasa/sangre , Memoria/efectos de los fármacos , Ratones , Ratones Mutantes/genética , Ratones Mutantes/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Superóxido Dismutasa/sangre , Proteína bcl-X/biosíntesis , Proteína bcl-X/genética
16.
Indian J Exp Biol ; 43(11): 1048-57, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16315394

RESUMEN

As a prerequisite for studies using mutant mice, we established a mouse model for induction of male germ cell apoptosis after deprivation of gonadotropins and intratesticular testosterone (T). We employed a potent long acting gonadotropin-releasing hormone antagonist (GnRH-A), acyline, alone or in combination with an antiandrogen, flutamide for effective induction of germ cell apoptosis in mice. Combined treatment with continuous release of acyline (3 mg/kg BW/day) with flutamide (in the form of sc pellets of 25 mg) resulted in almost the same level of suppression of spermatogenesis, as judged by testis weight and by germ cell apoptotic index, in 2 weeks as that reported for rats after treatment with 1.25 mg/kg BW Nal-Glu GnRH-A for the same time period. Within the study paradigm, the maximum suppression of spermatogenesis occurred after a single sc injection of high (20 mg/kg BW) dose of acyline with flutamide. The combined treatment resulted in complete absence of elongated spermatids. Germ cell counts at stages VII-VIII showed a significant (P < 0.05) reduction in the number of preleptotene (27.1%) and pachytene spermatocytes (81.9%), and round spermatids (96.6%) in acyline + flutamide group in comparison with controls. In fact, treatment with a single high (20 mg/kg BW) dose of acyline combined with flutamide in mice achieved same or greater level of suppression, measured by germ cell counts at stages VII-VIII, in two weeks when compared with those reported after daily treatment with Nal-Glu GnRH-A for 4 weeks in rats. Both plasma and testicular T levels were markedly suppressed after administration of acyline alone either by miniosmotic pump or by a single sc injection. Addition of flutamide to acyline had no discernible effect on plasma or intratesticular T levels when compared with acyline alone. These results demonstrate that optimum suppression of spermatogenesis through increased germ cell death is only possible in mice if total abolition of androgen action is achieved and further emphasize the usefulness of acyline + flutamide treated mice as a suitable model system to study hormonal regulation of testicular germ cell apoptosis.


Asunto(s)
Apoptosis , Células Germinativas/citología , Células Germinativas/patología , Hormonas/metabolismo , Animales , Daño del ADN , Flutamida/metabolismo , Flutamida/farmacología , Células Germinativas/efectos de los fármacos , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes/metabolismo , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Oligopéptidos/farmacología , Ratas , Células de Sertoli/patología , Espermatogénesis , Testículo/patología , Testosterona/metabolismo , Factores de Tiempo
17.
J Cell Biol ; 171(6): 1013-22, 2005 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-16365167

RESUMEN

Ablation of the Raf-1 protein causes fetal liver apoptosis, embryonic lethality, and selective hypersensitivity to Fas-induced cell death. Furthermore, Raf-1-deficient cells show defective migration as a result of the deregulation of the Rho effector kinase Rok-alpha. In this study, we show that the kinase-independent modulation of Rok-alpha signaling is also the basis of the antiapoptotic function of Raf-1. Fas activation stimulates the formation of Raf-1-Rok-alpha complexes, and Rok-alpha signaling is up-regulated in Raf-1-deficient cells. This leads to increased clustering and membrane expression of Fas, which is rescued both by kinase-dead Raf-1 and by interfering with Rok-alpha or its substrate ezrin. Increased Fas clustering and membrane expression are also evident in the livers of Raf-1-deficient embryos, and genetically reducing Fas expression counteracts fetal liver apoptosis, embryonic lethality, and the apoptotic defects of embryonic fibroblasts. Thus, Raf-1 has an essential function in regulating Fas expression and setting the threshold of Fas sensitivity during embryonic life.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Animales , Apoptosis/genética , Apoptosis/fisiología , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Relación Dosis-Respuesta a Droga , Desarrollo Embrionario/fisiología , Fibroblastos/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Genes Letales , Péptidos y Proteínas de Señalización Intracelular , Hígado/embriología , Hígado/metabolismo , Membranas/metabolismo , Ratones , Ratones Mutantes/embriología , Ratones Mutantes/metabolismo , Modelos Biológicos , Proteínas Proto-Oncogénicas c-raf/genética , Sensibilidad y Especificidad , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Quinasas Asociadas a rho
18.
Neuroscience ; 134(3): 877-87, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15994019

RESUMEN

A reduction of transforming growth factor-alpha (TGFalpha) expression in the spontaneous Waved-1 (Wa-1) mutant mouse causes specific behavioral and anatomical changes, including reduced fear learning and stress response and enlarged lateral ventricles. These alterations are observed predominantly in male Wa-1 mice after puberty. We hypothesized that regional differences in the expression of TGFalpha and its receptor, epidermal growth factor receptor (EGFR), may regulate the sexual dimorphism of the brain structures and functions during postnatal development. In general, fear learning-associated structures, including hippocampus and amygdala, showed maximum expression before puberty, regardless of genotype. In contrast, an overall temporal delay in the rise of both transcript levels, which peaked around or after puberty onset, was observed for the major stress regulatory hypothalamo-pituitary-adrenal axis. This pattern of expression was reversed for amygdala EGFR and hypothalamus TGFalpha and EGFR transcripts in males. When regional TGFalpha expression was compared between control and Wa-1 mice, far more complex patterns than expected were observed that revealed sex- and structure-dependent differences. In fact, the amygdala, hypothalamus, and pituitary TGFalpha expression pattern in Wa-1 exhibited a clear sex dependency across various age groups. Surprisingly, there was no compensatory up-regulation of the EGFR transcript in Wa-1 mice. The observed expression patterns of the TGFalpha signaling system during normal development and in the Wa-1 mutant mouse suggest complex sex- and age-dependent transcription regulatory mechanisms.


Asunto(s)
Encéfalo/metabolismo , Receptores ErbB/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Diferenciación Sexual , Factor de Crecimiento Transformador alfa/genética , Factores de Edad , Animales , Animales Recién Nacidos , Conducta Animal , Encéfalo/crecimiento & desarrollo , Receptores ErbB/metabolismo , Femenino , Genotipo , Hibridación in Situ/métodos , Masculino , Ratones , Ratones Endogámicos C57BL/metabolismo , Ratones Mutantes/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factor de Crecimiento Transformador alfa/metabolismo
19.
Dev Biol ; 280(2): 295-306, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15882574

RESUMEN

Mutant alleles of the gene encoding cadherin 23 are associated with Usher syndrome type 1 (USH1D), isolated deafness (DFNB12) in humans, and deafness and circling behavior in waltzer (v) mice. Stereocilia of waltzer mice are disorganized and the kinocilia misplaced, indicating the importance of cadherin 23 for hair bundle development. Cadherin 23 was localized to developing stereocilia and proposed as a component of the tip link. We show that, during development of the inner ear, cadherin 23 is initially detected in centrosomes at E14.5, then along the length of emerging stereocilia, and later becomes concentrated at and subsequently disappears from the tops of stereocilia. In mature vestibular hair bundles, cadherin 23 is present along the kinocilium and in the region of stereocilia-kinocilium bonds, a pattern conserved in mammals, chicks, and frogs. Cadherin 23 is also present in Reissner's membrane (RM) throughout development. In homozygous v(6J) mice, a reported null allele, cadherin 23 was absent from stereocilia, but present in kinocilia, RM, and centrosomes. We reconciled these results by identifying two novel isoforms of Cdh23 unaffected in sequence and expression by the v(6J) allele. Our results suggest that Cdh23 participation in stereocilia links may be restricted to developing hair bundles.


Asunto(s)
Cadherinas/biosíntesis , Cadherinas/química , Oído Interno/embriología , Células Ciliadas Auditivas/embriología , Alelos , Animales , Northern Blotting , Western Blotting , Proteínas Relacionadas con las Cadherinas , Cadherinas/metabolismo , Adhesión Celular , Centrosoma/metabolismo , Embrión de Pollo , Cilios/metabolismo , ADN Complementario/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Homocigoto , Humanos , Membranas Intracelulares/metabolismo , Ratones , Ratones Mutantes/metabolismo , Microscopía Fluorescente , Modelos Genéticos , Mutación , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas , Estructura Terciaria de Proteína , Factores de Tiempo , Transfección , Xenopus
20.
Nat Neurosci ; 8(6): 709-15, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15917835

RESUMEN

The Notch pathway, although originally identified in fruit flies, is now among the most heavily studied in mammalian biology. In mice, loss-of-function and gain-of-function work has demonstrated that Notch signaling is essential both during development and in the adult in a multitude of tissues. Prominent among these is the CNS, where Notch has been implicated in processes ranging from neural stem cell regulation to learning and memory. Here we review the role of Notch in the mammalian CNS by focusing specifically on mutations generated in mice. These mutations have provided critical insight into Notch function in the CNS and have led to the identification of promising new directions that are likely to generate important discoveries in the future.


Asunto(s)
Diferenciación Celular/genética , Sistema Nervioso Central/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Malformaciones del Sistema Nervioso/genética , Receptores de Superficie Celular/metabolismo , Factores de Transcripción/metabolismo , Animales , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Humanos , Ratones , Ratones Mutantes/anomalías , Ratones Mutantes/genética , Ratones Mutantes/metabolismo , Mutación/genética , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/fisiopatología , Receptor Notch1 , Receptores de Superficie Celular/genética , Transducción de Señal/genética , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...