Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Acta Neuropathol ; 140(5): 737-764, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32642868

RESUMEN

Impaired neuronal proteostasis is a salient feature of many neurodegenerative diseases, highlighting alterations in the function of the endoplasmic reticulum (ER). We previously reported that targeting the transcription factor XBP1, a key mediator of the ER stress response, delays disease progression and reduces protein aggregation in various models of neurodegeneration. To identify disease modifier genes that may explain the neuroprotective effects of XBP1 deficiency, we performed gene expression profiling of brain cortex and striatum of these animals and uncovered insulin-like growth factor 2 (Igf2) as the major upregulated gene. Here, we studied the impact of IGF2 signaling on protein aggregation in models of Huntington's disease (HD) as proof of concept. Cell culture studies revealed that IGF2 treatment decreases the load of intracellular aggregates of mutant huntingtin and a polyglutamine peptide. These results were validated using induced pluripotent stem cells (iPSC)-derived medium spiny neurons from HD patients and spinocerebellar ataxia cases. The reduction in the levels of mutant huntingtin was associated with a decrease in the half-life of the intracellular protein. The decrease in the levels of abnormal protein aggregation triggered by IGF2 was independent of the activity of autophagy and the proteasome pathways, the two main routes for mutant huntingtin clearance. Conversely, IGF2 signaling enhanced the secretion of soluble mutant huntingtin species through exosomes and microvesicles involving changes in actin dynamics. Administration of IGF2 into the brain of HD mice using gene therapy led to a significant decrease in the levels of mutant huntingtin in three different animal models. Moreover, analysis of human postmortem brain tissue and blood samples from HD patients showed a reduction in IGF2 level. This study identifies IGF2 as a relevant factor deregulated in HD, operating as a disease modifier that buffers the accumulation of abnormal protein species.


Asunto(s)
Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Agregación Patológica de Proteínas/metabolismo , Animales , Humanos , Factor II del Crecimiento Similar a la Insulina/farmacología , Ratones , Ratones Transgénicos , Agregado de Proteínas/efectos de los fármacos
2.
J Pharmacol Exp Ther ; 339(1): 115-24, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21775475

RESUMEN

Ghrelin influences a variety of metabolic functions through a direct action at its receptor, the GhrR (GhrR-1a). Ghrelin knockout (KO) and GhrR KO mice are resistant to the negative effects of high-fat diet (HFD) feeding. We have generated several classes of small-molecule GhrR antagonists and evaluated whether pharmacologic blockade of ghrelin signaling can recapitulate the phenotype of ghrelin/GhrR KO mice. Antagonist treatment blocked ghrelin-induced and spontaneous food intake; however, the effects on spontaneous feeding were absent in GhrR KO mice, suggesting target-specific effects of the antagonists. Oral administration of antagonists to HFD-fed mice improved insulin sensitivity in both glucose tolerance and glycemic clamp tests. The insulin sensitivity observed was characterized by improved glucose disposal with dramatically decreased insulin secretion. It is noteworthy that these results mimic those obtained in similar tests of HFD-fed GhrR KO mice. HFD-fed mice treated for 56 days with antagonist experienced a transient decrease in food intake but a sustained body weight decrease resulting from decreased white adipose, but not lean tissue. They also had improved glucose disposal and a striking reduction in the amount of insulin needed to achieve this. These mice had reduced hepatic steatosis, improved liver function, and no evidence of systemic toxicity relative to controls. Furthermore, GhrR KO mice placed on low- or high-fat diets had lifespans similar to the wild type, emphasizing the long-term safety of ghrelin receptor blockade. We have therefore demonstrated that chronic pharmacologic blockade of the GhrR is an effective and safe strategy for treating metabolic syndrome.


Asunto(s)
Resistencia a la Insulina/fisiología , Insulina/metabolismo , Receptores de Ghrelina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Fármacos Antiobesidad/farmacología , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Grasas de la Dieta/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ghrelina/antagonistas & inhibidores , Ghrelina/farmacología , Técnica de Clampeo de la Glucosa , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/tratamiento farmacológico , Receptores de Ghrelina/fisiología , Estrés Fisiológico/fisiología
3.
BMC Physiol ; 11: 1, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21211044

RESUMEN

BACKGROUND: We and others have demonstrated previously that ghrelin receptor (GhrR) knock out (KO) mice fed a high fat diet (HFD) have increased insulin sensitivity and metabolic flexibility relative to WT littermates. A striking feature of the HFD-fed GhrR KO mouse is the dramatic decrease in hepatic steatosis. To characterize further the underlying mechanisms of glucose homeostasis in GhrR KO mice, we conducted both hyperglycemic (HG) and hyperinsulinemic-euglycemic (HI-E) clamps. Additionally, we investigated tissue glucose uptake and specifically examined liver insulin sensitivity. RESULTS: Consistent with glucose tolerance-test data, in HG clamp experiments, GhrR KO mice showed a reduction in glucose-stimulated insulin release relative to WT littermates. Nevertheless, a robust 1st phase insulin secretion was still achieved, indicating that a healthy ß-cell response is maintained. Additionally, GhrR KO mice demonstrated both a significantly increased glucose infusion rate and significantly reduced insulin requirement for maintenance of the HG clamp, consistent with their relative insulin sensitivity. In HI-E clamps, both LFD-fed and HFD-fed GhrR KO mice showed higher peripheral insulin sensitivity relative to WT littermates as indicated by a significant increase in insulin-stimulated glucose disposal (Rd), and decreased hepatic glucose production (HGP). HFD-fed GhrR KO mice showed a marked increase in peripheral tissue glucose uptake in a variety of tissues, including skeletal muscle, brown adipose tissue and white adipose tissue. GhrR KO mice fed a HFD also showed a modest, but significant decrease in conversion of pyruvate to glucose, as would be anticipated if these mice displayed increased liver insulin sensitivity. Additionally, the levels of UCP2 and UCP1 were reduced in the liver and BAT, respectively, in GhrR KO mice relative to WT mice. CONCLUSIONS: These results indicate that improved glucose homeostasis of GhrR KO mice is characterized by robust improvements of glucose disposal in both normal and metabolically challenged states, relative to WT controls. GhrR KO mice have an intact 1st phase insulin response but require significantly less insulin for glucose disposal. Our experiments reveal that the insulin sensitivity of GhrR KO mice is due to both BW independent and dependent factors. We also provide several lines of evidence that a key feature of the GhrR KO mouse is maintenance of hepatic insulin sensitivity during metabolic challenge.


Asunto(s)
Técnica de Clampeo de la Glucosa/métodos , Resistencia a la Insulina/genética , Insulina/sangre , Receptores de Ghrelina/deficiencia , Animales , Grasas de la Dieta/administración & dosificación , Prueba de Tolerancia a la Glucosa/métodos , Índice Glucémico/genética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R747-54, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20018821

RESUMEN

To define the relationship between the respiratory quotient (RQ) and energy intake (EI) and to determine the impact of spontaneous locomotor activity (LMA) in the development of diet-induced obesity (DIO), we fed C57BL/6 mice a high-fat diet (HFD) for either 4 days or 17 wk and analyzed them using indirect calorimetry. Importantly, changes in body mass during calorimetry (DeltaM(b)) significantly covaried with RQ and EI; adjusting the data for DeltaM(b) permitted an analysis of the energy-balanced state. The 24-h RQ strongly predicted 24-h EI, and the slope of this relationship was diet dependent (HFD or chow) but independent of the HFD feeding period. Early-stage DIO was characterized by dark-period hyperphagia and fat storage, offset by greater light-period lipid oxidation; later stage DIO mice had a milder hyperphagia and lower substrate flexibility. Consequently, whereas 24-h RQ equaled the food quotient of the HFD in both early- and late-stage DIO, the range of RQ values was negatively correlated with, and mostly explained by, 24-h EI only in late-stage DIO. Lean and early-stage DIO mice had similar LMA values that were reduced in late-stage DIO. However, LMA significantly explained variance in total energy expenditure (EE) in only early-stage DIO mice. This indicated that the link between LMA and EE was a transient adaptive response to early DIO, whereas the later loss of LMA did not explain body weight gain in C57BL/6 DIO mice.


Asunto(s)
Peso Corporal/fisiología , Metabolismo Energético/fisiología , Hiperfagia/metabolismo , Obesidad/metabolismo , Consumo de Oxígeno/fisiología , Animales , Calorimetría Indirecta , Grasas de la Dieta/farmacología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Valor Predictivo de las Pruebas
5.
Mol Ther Methods Clin Dev ; 19: 162-173, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33209959

RESUMEN

Novel treatments for Huntington's disease (HD), a progressive neurodegenerative disorder, include selective targeting of the mutant allele of the huntingtin gene (mHTT) carrying the abnormally expanded disease-causing cytosine-adenine-guanine (CAG) repeat. WVE-120101 and WVE-120102 are investigational stereopure antisense oligonucleotides that enable selective suppression of mHTT by targeting single-nucleotide polymorphisms (SNPs) that are in haplotype phase with the CAG repeat expansion. Recently developed long-read sequencing technologies can capture CAG expansions and distant SNPs of interest and potentially facilitate haplotype-based identification of patients for clinical trials of oligonucleotide therapies. However, improved methods are needed to phase SNPs with CAG repeat expansions directly and reliably without need for familial genotype/haplotype data. Our haplotype phasing method uses single-molecule real-time sequencing and a custom algorithm to determine with confidence bases at SNPs on mutant alleles, even without familial data. Herein, we summarize this methodology and validate the approach using patient-derived samples with known phasing results. Comparison of experimentally measured CAG repeat lengths, heterozygosity, and phasing with previously determined results showed improved performance. Our methodology enables the haplotype phasing of SNPs of interest and the disease-causing, expanded CAG repeat of the huntingtin gene, enabling accurate identification of patients with HD eligible for allele-selective clinical studies.

6.
Neurol Genet ; 6(3): e430, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32548276

RESUMEN

BACKGROUND: The huntingtin gene (HTT) pathogenic cytosine-adenine-guanine (CAG) repeat expansion responsible for Huntington disease (HD) is phased with single nucleotide polymorphisms (SNPs), providing targets for allele-selective treatments. OBJECTIVE: This prospective observational study defined the frequency at which rs362307 (SNP1) or rs362331 (SNP2) was found on the same allele with pathogenic CAG expansions. METHODS: Across 7 US sites, 202 individuals with HD provided blood samples that were processed centrally to determine the number and size of CAG repeats, presence and heterozygosity of SNPs, and whether SNPs were present on the mutant HTT allele using long-read sequencing and phasing. RESULTS: Heterozygosity of SNP1 and/or SNP2 was identified in 146 (72%) individuals. The 2 polymorphisms were associated only with the mHTT allele in 61% (95% high density interval: 55%, 67%) of individuals. CONCLUSIONS: These results are consistent with previous reports and demonstrate the feasibility of genotyping, phasing, and targeting of HTT SNPs for personalized treatment of HD.

7.
J Pharmacol Exp Ther ; 329(3): 1178-86, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19252061

RESUMEN

The orexigenic peptide ghrelin has been shown to have prokinetic activity in the gastrointestinal (GI) system of several species, including humans. In this series of experiments, we have evaluated the prokinetic activity of novel, small-molecule ghrelin receptor (GhrR) agonists after parenteral and peroral dosing in mice and rats. Gastric emptying, small intestinal transport, and fecal output were determined after intraperitoneal and intracerebroventricular dosing of GhrR agonists, using ghrelin as a positive control. These same parameters were evaluated after oral gavage dosing of the synthetic agonists. Regardless of dose route, GhrR agonist treatment increased gastric emptying, small intestinal transit, and fecal output. However, fecal output was only increased by GhrR agonist treatment if mice were able to feed during the stimulatory period. Thus, GhrR agonists can stimulate upper GI motility, and the orexigenic action of the compounds can indirectly contribute to prokinetic activity along the entire GI tract. The orexigenic and prokinetic effects of either ghrelin or small-molecule GhrR agonists were selective for the GhrR because they were absent when evaluated in GhrR knockout mice. We next evaluated the efficacy of the synthetic GhrR agonists dosed in a model of opiate-induced bowel dysfunction induced by a single injection of morphine. Oral dosing of a GhrR agonist normalized GI motility in opiate-induced dysmotility. These data demonstrate the potential utility of GhrR agonists for treating gastrointestinal hypomotility disorders.


Asunto(s)
Motilidad Gastrointestinal/efectos de los fármacos , Ghrelina/administración & dosificación , Ghrelina/farmacología , Hormonas Peptídicas/administración & dosificación , Hormonas Peptídicas/farmacología , Receptores de Ghrelina/agonistas , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Enfermedad de Bowen/inducido químicamente , Enfermedad de Bowen/tratamiento farmacológico , Enfermedad de Bowen/fisiopatología , Sistema Nervioso Central/efectos de los fármacos , Defecación/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Vaciamiento Gástrico/efectos de los fármacos , Tránsito Gastrointestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Hormonas Peptídicas/sangre , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo
8.
Diabetes ; 56(2): 295-303, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17259372

RESUMEN

The Wnt family of secreted signaling molecules has profound effects on diverse developmental processes, including the fate of mesenchymal progenitors. While activation of Wnt signaling blocks adipogenesis, inhibition of endogenous Wnt/beta-catenin signaling by Wnt10b promotes spontaneous preadipocyte differentiation. Transgenic mice with expression of Wnt10b from the FABP4 promoter (FABP4-Wnt10b) have less adipose tissue when maintained on a normal chow diet and are resistant to diet-induced obesity. Here we demonstrate that FABP4-Wnt10b mice largely avert weight gain and metabolic abnormalities associated with genetic obesity. FABP4-Wnt10b mice do not gain significant body weight on the ob/ob background, and at 8 weeks of age, they have an approximately 70% reduction in visceral and subcutaneous adipose tissues compared with ob/ob mice. Similarly, on the lethal yellow agouti (A(y)) background, FABP4-Wnt10b mice have 50-70% less adipose tissue weight and circulating leptin at 5 months of age. Wnt10b-Ay mice are more glucose tolerant and insulin sensitive than A(y) controls, perhaps due to reduced expression and circulation of resistin. Reduced expression of inflammatory cytokines may also contribute to improved glucose homeostasis.


Asunto(s)
Tejido Adiposo/fisiología , Proteínas de Unión a Ácidos Grasos/fisiología , Resistencia a la Insulina/fisiología , Obesidad/fisiopatología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Wnt/fisiología , Proteína de Señalización Agouti , Animales , Glucemia/fisiología , Modelos Animales de Enfermedad , Ingestión de Energía/fisiología , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Leptina/deficiencia , Leptina/genética , Masculino , Ratones , Ratones Transgénicos , Obesidad/genética , Consumo de Oxígeno/fisiología , Paniculitis/fisiopatología
9.
Regul Pept ; 150(1-3): 55-61, 2008 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-18453014

RESUMEN

Stimulation of the ghrelin receptor (GhrR) by ghrelin results in a variety of metabolic changes including increased food intake, fat storage and insulin resistance. Loss of ghrelin signaling is protective against diet-induced obesity, suggesting that ghrelin plays a significant homeostatic role in conditions of metabolic stress. We examined glycemic control in GhrR -/- mice fed a high-fat diet, and used indirect calorimetry to assess fuel substrate usage and energy expenditure. GhrR -/- mice fed a high-fat diet had several measures of greater insulin sensitivity, including: lower fasted blood glucose and plasma insulin, lower %Hb(A1c), lower insulin levels during glucose tolerance tests, and improved performance in hyperinsulinemic-euglycemic and hyperglycemic clamp studies. GhrR -/- mice fed a high-fat diet did not develop hepatic steatosis and had lower total cholesterol, relative to controls. Furthermore, GhrR -/- mice demonstrated a lower intestinal triglyceride secretion rate of dietary lipid. GhrR -/- mice have higher respiratory quotients (RQ), indicating a preference for carbohydrate as fuel. The range of RQ values was wider in GhrR -/- mice, indicating greater metabolic flexibility and insulin sensitivity in these animals. We therefore propose that loss of ghrelin signaling promotes insulin sensitivity and metabolic flexibility, and protects against several fatty diet-induced features of metabolic syndrome due to convergent changes in the intake, absorption and utilization of energy.


Asunto(s)
Grasas de la Dieta/metabolismo , Metabolismo Energético/fisiología , Resistencia a la Insulina/fisiología , Receptores de Ghrelina/genética , Animales , Glucemia/análisis , Calorimetría Indirecta/métodos , Colesterol/metabolismo , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Ayuno , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Hemoglobina Glucada/metabolismo , Insulina/sangre , Ratones , Ratones Noqueados , Triglicéridos/metabolismo
10.
Mol Cell Biol ; 25(4): 1272-82, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15684380

RESUMEN

Activation of canonical Wnt signaling inhibits brown adipogenesis of cultured cells by impeding induction of PPARgamma and C/EBPalpha. Although enforced expression of these adipogenic transcription factors restores lipid accumulation and expression of FABP4 in Wnt-expressing cells, additional expression of PGC-1alpha is required for activation of uncoupling protein 1 (UCP1). Wnt10b blocks brown adipose tissue development and expression of UCP1 when expressed from the fatty acid binding protein 4 promoter, even when mice are administered a beta3-agonist. In differentiated brown adipocytes, activation of Wnt signaling suppresses expression of UCP1 through repression of PGC-1alpha. Consistent with these in vitro observations, UCP1-Wnt10b transgenic mice, which express Wnt10b in interscapular tissue, lack functional brown adipose tissue. While interscapular tissue of UCP1-Wnt10b mice lacks expression of PGC-1alpha and UCP1, the presence of unilocular lipid droplets and expression of white adipocyte genes suggest conversion of brown adipose tissue to white. Reciprocal expression of Wnt10b with UCP1 and PGC-1alpha in interscapular tissue from cold-challenged or genetically obese mice provides further evidence for regulation of brown adipocyte metabolism by Wnt signaling. Taken together, these data suggest that activation of canonical Wnt signaling early in differentiation blocks brown adipogenesis, whereas activating Wnt signaling in mature brown adipocytes stimulates their conversion to white adipocytes.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Diferenciación Celular/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Adipocitos/citología , Tejido Adiposo Pardo/citología , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Portadoras/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Canales Iónicos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción , Proteína Desacopladora 1 , Proteínas Wnt
11.
Mol Biol Cell ; 16(4): 2039-48, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15673614

RESUMEN

Adult myoblasts retain plasticity in developmental potential and can be induced to undergo myogenic, adipogenic, or osteoblastogenic differentiation in vitro. In this report, we show that the balance between myogenic and adipogenic potential in myoblasts is controlled by Wnt signaling. Furthermore, this balance is altered during aging such that aspects of both differentiation programs are coexpressed in myoblasts due to decreased Wnt10b abundance. Mimicking Wnt signaling in aged myoblasts through inhibition of glycogen synthase kinase or through overexpression of Wnt10b resulted in inhibition of adipogenic gene expression and sustained or enhanced myogenic differentiation. On the other hand, myoblasts isolated from Wnt10b null mice showed increased adipogenic potential, likely contributing to excessive lipid accumulation in actively regenerating myofibers in vivo in Wnt10b-/- mice. Whereas Wnt10b deficiency contributed to increased adipogenic potential in myoblasts, the augmented myogenic differentiation potential observed is likely the result of a compensatory increase in Wnt7b during differentiation of Wnt10b-/- myoblasts. No such compensation was apparent in aged myoblasts and in fact, both Wnt5b and Wnt10b were down-regulated. Thus, alteration in Wnt signaling in myoblasts with age may contribute to impaired muscle regenerative capacity and to increased muscle adiposity, both characteristic of aged muscle.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Diferenciación Celular , Mioblastos/citología , Mioblastos/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Animales , Células Cultivadas , Regulación de la Expresión Génica , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Desarrollo de Músculos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Wnt
12.
Mol Cell Biol ; 22(16): 5989-99, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12138207

RESUMEN

Wnt signaling maintains preadipocytes in an undifferentiated state. When Wnt signaling is enforced, 3T3-L1 preadipocytes no longer undergo adipocyte conversion in response to adipogenic medium. Here we used microarray analyses to identify subsets of genes whose expression is aberrant when differentiation is blocked through enforced Wnt signaling. Furthermore, we used the microarray data to identify potentially important adipocyte genes and chose one of these, the liver X receptor alpha (LXR alpha), for further analyses. Our studies indicate that enforced Wnt signaling blunts the changes in gene expression that correspond to mitotic clonal expansion, suggesting that Wnt signaling inhibits adipogenesis in part through dysregulation of the cell cycle. Experiments designed to uncover the potential role of LXR alpha in adipogenesis revealed that this transcription factor, unlike CCAAT/enhancer binding protein alpha and peroxisome proliferator-activated receptor gamma, is not adipogenic but rather inhibits adipogenesis if inappropriately expressed and activated. However, LXR alpha has several important roles in adipocyte function. Our studies show that this nuclear receptor increases basal glucose uptake and glycogen synthesis in 3T3-L1 adipocytes. In addition, LXR alpha increases cholesterol synthesis and release of nonesterified fatty acids. Finally, treatment of mice with an LXR alpha agonist results in increased serum levels of glycerol and nonesterified fatty acids, consistent with increased lipolysis within adipose tissue. These findings demonstrate new metabolic roles for LXR alpha and increase our understanding of adipogenesis.


Asunto(s)
Adipocitos/fisiología , Diferenciación Celular/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas/metabolismo , Receptores Citoplasmáticos y Nucleares , Receptores de Ácido Retinoico/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Proteínas de Pez Cebra , Adipocitos/efectos de los fármacos , Tejido Adiposo/citología , Animales , Anticolesterolemiantes/farmacología , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ácidos Grasos no Esterificados/sangre , Femenino , Perfilación de la Expresión Génica , Glicerol/sangre , Humanos , Hidrocarburos Fluorados , Ligandos , Metabolismo de los Lípidos , Receptores X del Hígado , Ratones , Ratones Endogámicos C57BL , Receptores Nucleares Huérfanos , Fenotipo , Proteínas Proto-Oncogénicas/genética , Receptores de Ácido Retinoico/genética , Receptores de Hormona Tiroidea/genética , Transducción de Señal/fisiología , Sulfonamidas , Proteínas Wnt
13.
BMC Med Genomics ; 9: 5, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26793951

RESUMEN

BACKGROUND: Parkinson disease (PD) is a neurodegenerative disease characterized by the accumulation of alpha-synuclein (SNCA) and other proteins in aggregates termed "Lewy Bodies" within neurons. PD has both genetic and environmental risk factors, and while processes leading to aberrant protein aggregation are unknown, past work points to abnormal levels of SNCA and other proteins. Although several genome-wide studies have been performed for PD, these have focused on DNA sequence variants by genome-wide association studies (GWAS) and on RNA levels (microarray transcriptomics), while genome-wide proteomics analysis has been lacking. METHODS: This study employed two state-of-the-art technologies, three-stage Mass Spectrometry Tandem Mass Tag Proteomics (12 PD, 12 controls) and RNA-sequencing transcriptomics (29 PD, 44 controls), evaluated in the context of PD GWAS implicated loci and microarray transcriptomics (19 PD, 24 controls). The technologies applied for this study were performed in a set of overlapping prefrontal cortex (Brodmann area 9) samples obtained from PD patients and sex and age similar neurologically healthy controls. RESULTS: After appropriate filters, proteomics robustly identified 3558 unique proteins, with 283 of these (7.9 %) significantly different between PD and controls (q-value < 0.05). RNA-sequencing identified 17,580 protein-coding genes, with 1095 of these (6.2 %) significantly different (FDR p-value < 0.05); only 166 of the FDR significant protein-coding genes (0.94 %) were present among the 3558 proteins characterized. Of these 166, eight genes (4.8 %) were significant in both studies, with the same direction of effect. Functional enrichment analysis of the proteomics results strongly supports mitochondrial-related pathways, while comparable analysis of the RNA-sequencing results implicates protein folding pathways and metallothioneins. Ten of the implicated genes or proteins co-localized to GWAS loci. Evidence implicating SNCA was stronger in proteomics than in RNA-sequencing analyses. CONCLUSIONS: We report the largest analysis of proteomics in PD to date, and the first to combine this technology with RNA-sequencing to investigate GWAS implicated loci. Notably, differentially expressed protein-coding genes were more likely to not be characterized in the proteomics analysis, which lessens the ability to compare across platforms. Combining multiple genome-wide platforms offers novel insights into the pathological processes responsible for this disease by identifying pathways implicated across methodologies.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Estudio de Asociación del Genoma Completo , Mitocondrias/metabolismo , Enfermedad de Parkinson/genética , Pliegue de Proteína , Proteómica/métodos , Anciano , Anciano de 80 o más Años , Ontología de Genes , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Sistemas de Lectura Abierta/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN
14.
Mol Cancer ; 2: 40, 2003 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-14641918

RESUMEN

BACKGROUND: The chromosomal location of CUL-5 (11q 22-23) is associated with LOH in breast cancer, suggesting that CUL-5 may be a tumor suppressor. The purpose of this research was to determine if there is differential expression of CUL-5 in breast epithelial cells versus breast cancer cell lines, and normal human tissues versus human tumors. The expression of CUL-5 in breast epithelial cells (HMEC, MCF-10A), and breast cancer cells (MCF-7, MDA-MB-231) was examined using RT-PCR, Northern blot analysis, and Western blot analysis. The expression of mRNA for other CUL family members (CUL-1, -2, -3, -4A, and -4B) in these cells was evaluated by RT-PCR. A normal human tissue expression array and a cancer profiling array were used to examine CUL-5 expression in normal human tissues and matched normal tissues versus tumor tissues, respectively. RESULTS: CUL-5 is expressed at the mRNA and protein levels by breast epithelial cells (HMEC, MCF-10A) and breast cancer cells (MCF-7, MDA-MB-231). These cells also express mRNA for other CUL family members. The normal human tissue expression array revealed that CUL-5 is widely expressed. The cancer profiling array revealed that 82% (41/50) of the breast cancers demonstrated a decrease in CUL-5 expression versus the matched normal tissue. For the 50 cases of matched breast tissue there was a statistically significant approximately 2.2 fold decreased expression of CUL-5 in tumor tissue versus normal tissue (P < 0.0001). CONCLUSIONS: The data demonstrate no apparent decrease in CUL-5 expression in the breast cancer cell lines (MCF-7, MDA-MB-231) versus the breast epithelial cells (HMEC, MCF-10A). The decrease in CUL-5 expression in breast tumor tissue versus matched normal tissue supports the hypothesis that decreased expression of CUL-5 may play a role in breast tumorigenesis.


Asunto(s)
Neoplasias de la Mama/genética , Mama/citología , Mama/patología , Proteínas Cullin/biosíntesis , Células Epiteliales/química , Células Epiteliales/metabolismo , Receptores de Vasopresinas/biosíntesis , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Epiteliales/patología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Especificidad de Órganos/genética , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis
15.
J Gerontol A Biol Sci Med Sci ; 69(3): 274-81, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23788654

RESUMEN

The goal of this study was to test whether the "loss of the complexity" hypothesis can be applied to compare the metabolic patterns of mouse models with known differences in metabolic and endocrine function as well as life span. Here, we compare the complexity of locomotor activity and metabolic patterns (energy expenditure, VO2, and respiratory quotient) of the long-lived growth hormone receptor gene deleted mice (GHR(-/-)) and their wild-type littermates. Using approximate entropy as a measure of complexity, we observed greater metabolic complexity, as indicated by greater irregularity in the physiological fluctuations of the GHR(-/-) mice. Further analysis of the data also revealed lower energy costs of locomotor activity and a stronger relationship between locomotor activity and respiratory quotient in the GHR(-/-) mice relative to controls. These findings suggest underlying differences in metabolic modulation in the GHR(-/-) mice revealed especially through measures of complexity of their time-dependent fluctuations.


Asunto(s)
Metabolismo Energético/fisiología , Longevidad/genética , Receptores de Somatotropina/genética , Algoritmos , Animales , Índice de Masa Corporal , Entropía , Femenino , Eliminación de Gen , Locomoción/fisiología , Ratones , Ratones Noqueados , Modelos Animales , Consumo de Oxígeno/fisiología , Respiración
16.
J Clin Invest ; 122(7): 2405-16, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22728933

RESUMEN

Preadipocytes secrete several WNT family proteins that act through autocrine/paracrine mechanisms to inhibit adipogenesis. The activity of WNT ligands is often decreased by secreted frizzled-related proteins (SFRPs). Sfrp5 is strongly induced during adipocyte differentiation and increases in adipocytes during obesity, presumably to counteract WNT signaling. We tested the hypothesis that obesity-induced Sfrp5 expression promotes the development of new adipocytes by inhibiting endogenous suppressors of adipogenesis. As predicted, mice that lack functional SFRP5 were resistant to diet-induced obesity. However, counter to our hypothesis, we found that adipose tissue of SFRP5-deficient mice had similar numbers of adipocytes, but a reduction in large adipocytes. Transplantation of adipose tissue from SFRP5-deficient mice into leptin receptor-deficient mice indicated that the effects of SFRP5 deficiency are tissue-autonomous. Mitochondrial gene expression was increased in adipose tissue and cultured adipocytes from SFRP5-deficient mice. In adipocytes, lack of SFRP5 stimulated oxidative capacity through increased mitochondrial activity, which was mediated in part by PGC1α and mitochondrial transcription factor A. WNT3a also increased oxygen consumption and the expression of mitochondrial genes. Thus, our findings support a model of adipogenesis in which SFRP5 inhibits WNT signaling to suppress oxidative metabolism and stimulate adipocyte growth during obesity.


Asunto(s)
Adipocitos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Mitocondrias/metabolismo , Obesidad/metabolismo , Vía de Señalización Wnt , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales , Adipogénesis , Tejido Adiposo Blanco/patología , Animales , Aumento de la Célula , Células Cultivadas , Oído Externo/patología , Metabolismo Energético , Matriz Extracelular/metabolismo , Femenino , Glucosa/metabolismo , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leptina/sangre , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/sangre , Obesidad/patología , Consumo de Oxígeno , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética , Activación Transcripcional , Proteína Wnt3A/metabolismo , Proteína Wnt3A/fisiología
17.
Growth Horm IGF Res ; 20(1): 73-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19747867

RESUMEN

The goal of this study was to examine factors that contribute to energy balance in female GHR -/- mice. We measured energy intake, energy expenditure (EE), fuel utilization, body mass (M(b)) changes and physical activity in 17month-old female GHR -/- mice and their age-matched wild type littermates. The GHR -/- mice were smaller, consumed more food per unit M(b), had greater EE per unit M(b) and had an increase in 24-h EE/M(b) that was similar to the increase in their surface-area-to-volume ratio. Locomotor activity (LMA) was reduced in the GHR -/- mice, but the energetic cost associated with their LMA was greater than in wild type controls. Furthermore, M(b) and LMA were independent explanatory covariates of most of the variance in EE, and when adjusted for M(b) and LMA, the GHR -/- mice had higher EE during both the light and dark phases of the daily cycle. Respiratory quotient was lower in GHR -/- mice during the light phase, which indicated a greater utilization of lipid relative to carbohydrate in these mice. Additionally, GHR -/- mice had higher ratios of caloric intake to EE at several intervals during the dark phase, and this effect was greater and more sustained in the final 3h of the dark phase. Therefore, we conclude that GHR -/- mice are able to overcome the substantial energetic challenges of dwarfism through several mechanisms that promote stable M(b). Relative to wild type mice, the GHR -/- mice consumed more calories per unit M(b), which offset the disproportionate increase in their daily energy expenditure. While GHR -/- mice oxidized a greater proportion of lipid during the light phase in order to meet their energy requirements, they achieved greater energy efficiency and storage during the dark phase through a combination of higher energy consumption and lower LMA.


Asunto(s)
Metabolismo Energético/fisiología , Receptores de Somatotropina/metabolismo , Ciclos de Actividad/fisiología , Animales , Enanismo/metabolismo , Ingestión de Energía/fisiología , Femenino , Metabolismo de los Lípidos/fisiología , Ratones , Ratones Mutantes , Actividad Motora/fisiología , Receptores de Somatotropina/genética , Receptores de Somatotropina/fisiología
18.
Proc Natl Acad Sci U S A ; 102(9): 3324-9, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15728361

RESUMEN

Wnts comprise a family of secreted signaling proteins that regulate diverse developmental processes. Activation of Wnt signaling by Wnt10b inhibits differentiation of preadipocytes and blocks adipose tissue development; however, the effect of Wnt10b on other mesenchymal lineages has not been defined. To explore the physiological role of Wnt signaling in bone development, we analyzed FABP4-Wnt10b mice, which express the Wnt10b transgene in marrow. Femurs from FABP4-Wnt10b mice have almost four times as much bone in the distal metaphyses and are mechanically stronger. These mice maintain elevated bone mass at least through 23 months of age. In addition, FABP4-Wnt10b mice are protected from the bone loss characteristic of estrogen deficiency. We used pharmacological and genetic approaches to demonstrate that canonical Wnt signaling stimulates osteoblastogenesis and inhibits adipogenesis of bipotential mesenchymal precursors. Wnt10b shifts cell fate toward the osteoblast lineage by induction of the osteoblastogenic transcription factors Runx2, Dlx5, and osterix and suppression of the adipogenic transcription factors C/EBPalpha and PPARgamma. One mechanism whereby Wnt10b promotes osteoblastogenesis is suppression of PPARgamma expression. Finally, Wnt10b-/- mice have decreased trabecular bone and serum osteocalcin, confirming that Wnt10b is an endogenous regulator of bone formation.


Asunto(s)
Densidad Ósea/fisiología , División Celular/fisiología , Osteoblastos/citología , Proteínas Proto-Oncogénicas/fisiología , Envejecimiento/fisiología , Animales , Femenino , Ratones , Ratones Noqueados , Ovariectomía , Proteínas Proto-Oncogénicas/genética , Proteínas Wnt
19.
J Biol Chem ; 277(41): 38239-44, 2002 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-12154096

RESUMEN

Ectopic expression of Wnt-1 in 3T3-L1 preadipocytes stabilizes beta-catenin, activates TCF-dependent gene transcription, and blocks adipogenesis. Here we report that upon serum withdrawal, Wnt-1 causes 3T3-L1 cells to resist apoptosis through a mechanism that is partially dependent on phosphatidylinositol 3-kinase. Although activation of Wnt signaling by inhibition of GSK-3 activity or ectopic expression of dominant stable beta-catenin blocks apoptosis, inhibition of Wnt signaling through expression of dominant negative TCF-4 increases apoptosis. Wnt-1 stimulates 3T3-L1 preadipocytes to secrete factors that increase PKB/Akt phosphorylation at levels comparable with treatment with 10% serum. With DNA microarrays, we identified several secreted antiapoptotic genes that are induced by Wnt-1, notably insulin-like growth factor I (IGF-I) and IGF-II. Consistent with IGFs mediating the antiapoptotic effects of Wnt-1 in preadipocytes, conditioned medium from Wnt-1 expressing 3T3-L1 cells was unable to promote protein kinase B phosphorylation after the addition of recombinant IGFBP-4. Thus, we demonstrated that Wnt-1 induces expression of antiapoptotic genes in 3T3-L1 preadipocytes such as IGF-I and IGF-II, which allows these cells to resist apoptosis in response to serum deprivation.


Asunto(s)
Adipocitos/metabolismo , Apoptosis/fisiología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra , Células 3T3 , Adipocitos/efectos de los fármacos , Androstadienos/farmacología , Animales , Cromonas/farmacología , Medios de Cultivo Condicionados , Medio de Cultivo Libre de Suero , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/genética , Litio/metabolismo , Ratones , Morfolinas/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Wnt , Proteína Wnt1 , Wortmanina , beta Catenina
20.
J Biol Chem ; 277(34): 30998-1004, 2002 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-12055200

RESUMEN

We have identified Wnt10b as a potent inhibitor of adipogenesis that must be suppressed for preadipocytes to differentiate in vitro. Here, we demonstrate that a specific inhibitor of glycogen synthase kinase 3, CHIR 99021, mimics Wnt signaling in preadipocytes. CHIR 99021 stabilizes free cytosolic beta-catenin and inhibits adipogenesis by blocking induction of CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma. Preadipocyte differentiation is inhibited when 3T3-L1 cells are exposed to CHIR 99021 for any 24 h period during the first 3 days of adipogenesis. Consistent with this time frame of inhibition, expression of Wnt10b mRNA is suppressed upon induction of differentiation, with a 50% decline by 6 h and complete inhibition by 36 h. Of the agents used to induce differentiation, exposure of 3T3-L1 cells to methyl-isobutylxanthine or cAMP is sufficient to suppress expression of Wnt10b mRNA. Inhibition of adipogenesis by Wnt10b is likely mediated by Wnt receptors, Frizzled 1, 2, and/or 5, and co-receptors low density lipoprotein receptor-related proteins 5 and 6. These receptors, like Wnt10b, are highly expressed in preadipocytes and stromal vascular cells. Finally, we demonstrate that disruption of extracellular Wnt signaling by expression of secreted Frizzled related proteins causes spontaneous adipocyte conversion.


Asunto(s)
Adipocitos/fisiología , Proteínas Proto-Oncogénicas/fisiología , Células 3T3 , Animales , Proteína alfa Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Diferenciación Celular , AMP Cíclico/fisiología , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3 , Glucógeno Sintasa Quinasas , Ratones , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Células Madre/fisiología , Factores de Transcripción/antagonistas & inhibidores , Proteínas Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA