Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Genom ; 3(7): 100346, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37492099

RESUMEN

A primary obstacle in translating genetic associations with disease into therapeutic strategies is elucidating the cellular programs affected by genetic risk variants and effector genes. Here, we introduce LipocyteProfiler, a cardiometabolic-disease-oriented high-content image-based profiling tool that enables evaluation of thousands of morphological and cellular profiles that can be systematically linked to genes and genetic variants relevant to cardiometabolic disease. We show that LipocyteProfiler allows surveillance of diverse cellular programs by generating rich context- and process-specific cellular profiles across hepatocyte and adipocyte cell-state transitions. We use LipocyteProfiler to identify known and novel cellular mechanisms altered by polygenic risk of metabolic disease, including insulin resistance, fat distribution, and the polygenic contribution to lipodystrophy. LipocyteProfiler paves the way for large-scale forward and reverse deep phenotypic profiling in lipocytes and provides a framework for the unbiased identification of causal relationships between genetic variants and cellular programs relevant to human disease.

2.
J Endocr Soc ; 4(11): bvaa121, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33150273

RESUMEN

Glucocorticoids have multiple therapeutic benefits and are used both for immunosuppression and treatment purposes. Notwithstanding their benefits, glucocorticoid use often leads to hyperglycemia. Owing to the pathophysiologic overlap in glucocorticoid-induced hyperglycemia (GIH) and type 2 diabetes (T2D), we hypothesized that genetic variation in glucocorticoid pathways contributes to T2D risk. To determine the genetic contribution of glucocorticoid action on T2D risk, we conducted multiple genetic studies. First, we performed gene-set enrichment analyses on 3 collated glucocorticoid-related gene sets using publicly available genome-wide association and whole-exome data and demonstrated that genetic variants in glucocorticoid-related genes are associated with T2D and related glycemic traits. To identify which genes are driving this association, we performed gene burden tests using whole-exome sequence data. We identified 20 genes within the glucocorticoid-related gene sets that are nominally enriched for T2D-associated protein-coding variants. The most significant association was found in coding variants in coiled-coil α-helical rod protein 1 (CCHCR1) in the HLA region (P = .001). Further analyses revealed that noncoding variants near CCHCR1 are also associated with T2D at genome-wide significance (P = 7.70 × 10-14), independent of type 1 diabetes HLA risk. Finally, gene expression and colocalization analyses demonstrate that variants associated with increased T2D risk are also associated with decreased expression of CCHCR1 in multiple tissues, implicating this gene as a potential effector transcript at this locus. Our discovery of a genetic link between glucocorticoids and T2D findings support the hypothesis that T2D and GIH may have shared underlying mechanisms.

3.
Cell Death Differ ; 27(1): 297-309, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31164717

RESUMEN

The Siva protein, named after the Hindu God of Destruction, plays important roles in apoptosis in various contexts, including downstream of death receptor activation or p53 tumor suppressor engagement. The function of Siva in organismal development and homeostasis, however, has remained uncharacterized. Here, we generate Siva knockout mice to characterize the physiological function of Siva in vivo. Interestingly, we find that Siva deficiency causes early embryonic lethality accompanied by multiple phenotypes, including developmental delay, abnormal neural tube closure, and defective placenta and yolk sac formation. Examination of Siva expression during embryogenesis shows that Siva is expressed in both embryonic and extra-embryonic tissues, including within the mesoderm, which may explain the vascular defects observed in the placenta and yolk sac. The embryonic phenotypes caused by Siva loss are not rescued by p53 deficiency, nor do they resemble those of p53 null embryos, suggesting that the embryonic function of Siva is not related to the p53 pathway. Moreover, loss of the Ripk3 necroptosis protein does not rescue the observed lethality or developmental defects, suggesting that Siva may play a non-apoptotic role in development. Collectively, these studies reveal a key role for Siva in proper embryonic development.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Desarrollo Embrionario , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Femenino , Genes Letales , Corazón/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Tubo Neural/anomalías , Fenotipo , Placenta/irrigación sanguínea , Embarazo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología , Proteína p53 Supresora de Tumor/fisiología , Saco Vitelino/irrigación sanguínea
4.
Diabetes ; 69(1): 112-120, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31636172

RESUMEN

Coronary artery disease (CAD) is more frequent among individuals with dysglycemia. Preventive interventions for diabetes can improve cardiometabolic risk factors (CRFs), but it is unclear whether the benefits on CRFs are similar for individuals at different genetic risk for CAD. We built a 201-variant polygenic risk score (PRS) for CAD and tested for interaction with diabetes prevention strategies on 1-year changes in CRFs in 2,658 Diabetes Prevention Program (DPP) participants. We also examined whether separate lifestyle behaviors interact with PRS and affect changes in CRFs in each intervention group. Participants in both the lifestyle and metformin interventions had greater improvement in the majority of recognized CRFs compared with placebo (P < 0.001) irrespective of CAD genetic risk (P interaction > 0.05). We detected nominal significant interactions between PRS and dietary quality and physical activity on 1-year change in BMI, fasting glucose, triglycerides, and HDL cholesterol in individuals randomized to metformin or placebo, but none of them achieved the multiple-testing correction for significance. This study confirms that diabetes preventive interventions improve CRFs regardless of CAD genetic risk and delivers hypothesis-generating data on the varying benefit of increasing physical activity and improving diet on intermediate cardiovascular risk factors depending on individual CAD genetic risk profile.


Asunto(s)
Enfermedades Cardiovasculares/genética , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus Tipo 2/prevención & control , Interacción Gen-Ambiente , Síndrome Metabólico/genética , Estado Prediabético , Servicios Preventivos de Salud , Adulto , Enfermedades Cardiovasculares/terapia , Enfermedad de la Arteria Coronaria/terapia , Diabetes Mellitus Tipo 2/genética , Ejercicio Físico , Terapia por Ejercicio , Femenino , Predisposición Genética a la Enfermedad , Humanos , Estilo de Vida , Masculino , Síndrome Metabólico/terapia , Metformina/uso terapéutico , Persona de Mediana Edad , Estado Prediabético/genética , Estado Prediabético/terapia , Factores de Riesgo , Estados Unidos/epidemiología
5.
Cell Rep ; 29(3): 778-780, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31618643

RESUMEN

Human genetic variants in SLC16A11 are associated with increased risk of type 2 diabetes (T2D). We previously identified two distinct mechanisms through which co-inherited T2D-risk coding and non-coding variants disrupt SLC16A11 expression and activity, thus implicating reduced SLC16A11 function as the disease-relevant direction of effect. In a recent publication, Zhao et al. (2019a) argue that human SLC16A11 coding variants confer gain of function, basing their conclusions on phenotypic changes observed following overexpression of mutant murine Slc16a11. However, data necessary to demonstrate gain-of-function activity are not reported. Furthermore, several fundamental flaws in their experimental system-including inaccurate modeling of the human variant haplotype and expression conditions that are not physiologically relevant-prevent conclusions about T2D-risk variant effects on human physiology. This Matters Arising paper is in response to Zhao et al. (2019a), published in Cell Reports. See also the response by Zhao et al. (2019b) in this issue of Cell Reports.


Asunto(s)
Diabetes Mellitus Tipo 2 , Animales , Mutación con Ganancia de Función , Haplotipos , Humanos , Ratones , Transportadores de Ácidos Monocarboxílicos/genética
6.
Cell Metab ; 29(4): 856-870.e7, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30686744

RESUMEN

The reactions catalyzed by the delta-5 and delta-6 desaturases (D5D/D6D), key enzymes responsible for highly unsaturated fatty acid (HUFA) synthesis, regenerate NAD+ from NADH. Here, we show that D5D/D6D provide a mechanism for glycolytic NAD+ recycling that permits ongoing glycolysis and cell viability when the cytosolic NAD+/NADH ratio is reduced, analogous to lactate fermentation. Although lesser in magnitude than lactate production, this desaturase-mediated NAD+ recycling is acutely adaptive when aerobic respiration is impaired in vivo. Notably, inhibition of either HUFA synthesis or lactate fermentation increases the other, underscoring their interdependence. Consistent with this, a type 2 diabetes risk haplotype in SLC16A11 that reduces pyruvate transport (thus limiting lactate production) increases D5D/D6D activity in vitro and in humans, demonstrating a chronic effect of desaturase-mediated NAD+ recycling. These findings highlight key biologic roles for D5D/D6D activity independent of their HUFA end products and expand the current paradigm of glycolytic NAD+ regeneration.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Glucólisis , NAD/metabolismo , Animales , Células Cultivadas , delta-5 Desaturasa de Ácido Graso , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
7.
Eur J Endocrinol ; 180(2): 99-107, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30475225

RESUMEN

Objective A haplotype at chromosome 17p13 that reduces expression and function of the solute carrier transporter SLC16A11 is associated with increased risk for type 2 diabetes in Mexicans. We aim to investigate the detailed metabolic profile of SLC16A11 risk haplotype carriers to identify potential physiological mechanisms explaining the increased type 2 diabetes risk. Design Cross-sectional study. Methods We evaluated carriers (n = 72) and non-carriers (n = 75) of the SLC16A11 risk haplotype, with or without type 2 diabetes. An independent sample of 1069 subjects was used to replicate biochemical findings. The evaluation included euglycemic-hyperinsulinemic clamp, frequently sampled intravenous glucose tolerance test (FSIVGTT), dual-energy X-ray absorptiometry (DXA), MRI and spectroscopy and subcutaneous abdominal adipose tissue biopsies. Results Fat-free mass (FFM)-adjusted M value was lower in carriers of the SLC16A11 risk haplotype after adjusting for age and type 2 diabetes status (ß = -0.164, P = 0.04). Subjects with type 2 diabetes and the risk haplotype demonstrated an increase of 8.76 U/L in alanine aminotransferase (ALT) (P = 0.02) and of 7.34 U/L in gamma-glutamyltransferase (GGT) (P = 0.05) compared with non-carriers and after adjusting for gender, age and ancestry. Among women with the risk haplotype and normal BMI, the adipocyte size was higher (P < 0.001). Conclusions Individuals carrying the SLC16A11 risk haplotype exhibited decreased insulin action. Higher serum ALT and GGT levels were found in carriers with type 2 diabetes, and larger adipocytes in subcutaneous fat in the size distribution in carrier women with normal weight.


Asunto(s)
Adipocitos/citología , Diabetes Mellitus Tipo 2/genética , Haplotipos , Resistencia a la Insulina/genética , Transportadores de Ácidos Monocarboxílicos/genética , Alanina Transaminasa/sangre , Composición Corporal/fisiología , Índice de Masa Corporal , Tamaño de la Célula , Estudios Transversales , Diabetes Mellitus Tipo 2/sangre , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Persona de Mediana Edad , Grasa Subcutánea/metabolismo , gamma-Glutamiltransferasa/sangre
8.
Cell ; 170(1): 199-212.e20, 2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28666119

RESUMEN

Type 2 diabetes (T2D) affects Latinos at twice the rate seen in populations of European descent. We recently identified a risk haplotype spanning SLC16A11 that explains ∼20% of the increased T2D prevalence in Mexico. Here, through genetic fine-mapping, we define a set of tightly linked variants likely to contain the causal allele(s). We show that variants on the T2D-associated haplotype have two distinct effects: (1) decreasing SLC16A11 expression in liver and (2) disrupting a key interaction with basigin, thereby reducing cell-surface localization. Both independent mechanisms reduce SLC16A11 function and suggest SLC16A11 is the causal gene at this locus. To gain insight into how SLC16A11 disruption impacts T2D risk, we demonstrate that SLC16A11 is a proton-coupled monocarboxylate transporter and that genetic perturbation of SLC16A11 induces changes in fatty acid and lipid metabolism that are associated with increased T2D risk. Our findings suggest that increasing SLC16A11 function could be therapeutically beneficial for T2D. VIDEO ABSTRACT.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Basigina/metabolismo , Membrana Celular/metabolismo , Cromosomas Humanos Par 17/metabolismo , Técnicas de Silenciamiento del Gen , Haplotipos , Hepatocitos/metabolismo , Heterocigoto , Código de Histonas , Humanos , Hígado/metabolismo , Modelos Moleculares , Transportadores de Ácidos Monocarboxílicos/química
9.
Diabetes ; 66(7): 2019-2032, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28341696

RESUMEN

To identify novel coding association signals and facilitate characterization of mechanisms influencing glycemic traits and type 2 diabetes risk, we analyzed 109,215 variants derived from exome array genotyping together with an additional 390,225 variants from exome sequence in up to 39,339 normoglycemic individuals from five ancestry groups. We identified a novel association between the coding variant (p.Pro50Thr) in AKT2 and fasting plasma insulin (FI), a gene in which rare fully penetrant mutations are causal for monogenic glycemic disorders. The low-frequency allele is associated with a 12% increase in FI levels. This variant is present at 1.1% frequency in Finns but virtually absent in individuals from other ancestries. Carriers of the FI-increasing allele had increased 2-h insulin values, decreased insulin sensitivity, and increased risk of type 2 diabetes (odds ratio 1.05). In cellular studies, the AKT2-Thr50 protein exhibited a partial loss of function. We extend the allelic spectrum for coding variants in AKT2 associated with disorders of glucose homeostasis and demonstrate bidirectional effects of variants within the pleckstrin homology domain of AKT2.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Ayuno/metabolismo , Resistencia a la Insulina/genética , Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Población Blanca/genética , Negro o Afroamericano/genética , Alelos , Pueblo Asiatico/genética , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/metabolismo , Finlandia , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Hispánicos o Latinos/genética , Humanos , Oportunidad Relativa
10.
Cancer Discov ; 5(6): 622-35, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25813352

RESUMEN

UNLABELLED: Although p53 transcriptional activation potential is critical for its ability to suppress cancer, the specific target genes involved in tumor suppression remain unclear. SIVA is a p53 target gene essential for p53-dependent apoptosis, although it can also promote proliferation through inhibition of p53 in some settings. Thus, the role of SIVA in tumorigenesis remains unclear. Here, we seek to define the contribution of SIVA to tumorigenesis by generating Siva conditional knockout mice. Surprisingly, we find that SIVA loss inhibits non-small cell lung cancer (NSCLC) development, suggesting that SIVA facilitates tumorigenesis. Similarly, SIVA knockdown in mouse and human NSCLC cell lines decreases proliferation and transformation. Consistent with this protumorigenic role for SIVA, high-level SIVA expression correlates with reduced NSCLC patient survival. SIVA acts independently of p53 and, instead, stimulates mTOR signaling and metabolism in NSCLC cells. Thus, SIVA enables tumorigenesis in a p53-independent manner, revealing a potential new cancer therapy target. SIGNIFICANCE: These findings collectively reveal a novel role for the p53 target gene SIVA both in regulating metabolism and in enabling tumorigenesis, independently of p53. Importantly, these studies further identify SIVA as a new prognostic marker and as a potential target for NSCLC cancer therapy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Autofagia/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Femenino , Orden Génico , Marcación de Gen , Sitios Genéticos , Humanos , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas p21(ras)/genética , Serina-Treonina Quinasas TOR/metabolismo
11.
JAMA ; 311(22): 2305-14, 2014 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-24915262

RESUMEN

IMPORTANCE: Latino populations have one of the highest prevalences of type 2 diabetes worldwide. OBJECTIVES: To investigate the association between rare protein-coding genetic variants and prevalence of type 2 diabetes in a large Latino population and to explore potential molecular and physiological mechanisms for the observed relationships. DESIGN, SETTING, AND PARTICIPANTS: Whole-exome sequencing was performed on DNA samples from 3756 Mexican and US Latino individuals (1794 with type 2 diabetes and 1962 without diabetes) recruited from 1993 to 2013. One variant was further tested for allele frequency and association with type 2 diabetes in large multiethnic data sets of 14,276 participants and characterized in experimental assays. MAIN OUTCOME AND MEASURES: Prevalence of type 2 diabetes. Secondary outcomes included age of onset, body mass index, and effect on protein function. RESULTS: A single rare missense variant (c.1522G>A [p.E508K]) was associated with type 2 diabetes prevalence (odds ratio [OR], 5.48; 95% CI, 2.83-10.61; P = 4.4 × 10(-7)) in hepatocyte nuclear factor 1-α (HNF1A), the gene responsible for maturity onset diabetes of the young type 3 (MODY3). This variant was observed in 0.36% of participants without type 2 diabetes and 2.1% of participants with it. In multiethnic replication data sets, the p.E508K variant was seen only in Latino patients (n = 1443 with type 2 diabetes and 1673 without it) and was associated with type 2 diabetes (OR, 4.16; 95% CI, 1.75-9.92; P = .0013). In experimental assays, HNF-1A protein encoding the p.E508K mutant demonstrated reduced transactivation activity of its target promoter compared with a wild-type protein. In our data, carriers and noncarriers of the p.E508K mutation with type 2 diabetes had no significant differences in compared clinical characteristics, including age at onset. The mean (SD) age for carriers was 45.3 years (11.2) vs 47.5 years (11.5) for noncarriers (P = .49) and the mean (SD) BMI for carriers was 28.2 (5.5) vs 29.3 (5.3) for noncarriers (P = .19). CONCLUSIONS AND RELEVANCE: Using whole-exome sequencing, we identified a single low-frequency variant in the MODY3-causing gene HNF1A that is associated with type 2 diabetes in Latino populations and may affect protein function. This finding may have implications for screening and therapeutic modification in this population, but additional studies are required.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Adulto , Edad de Inicio , Anciano , Femenino , Genotipo , Hispánicos o Latinos/genética , Humanos , Masculino , México , Persona de Mediana Edad , Mutación Missense , Análisis de Secuencia de ADN , Estados Unidos
12.
Nat Genet ; 46(4): 357-63, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24584071

RESUMEN

Loss-of-function mutations protective against human disease provide in vivo validation of therapeutic targets, but none have yet been described for type 2 diabetes (T2D). Through sequencing or genotyping of ~150,000 individuals across 5 ancestry groups, we identified 12 rare protein-truncating variants in SLC30A8, which encodes an islet zinc transporter (ZnT8) and harbors a common variant (p.Trp325Arg) associated with T2D risk and glucose and proinsulin levels. Collectively, carriers of protein-truncating variants had 65% reduced T2D risk (P = 1.7 × 10(-6)), and non-diabetic Icelandic carriers of a frameshift variant (p.Lys34Serfs*50) demonstrated reduced glucose levels (-0.17 s.d., P = 4.6 × 10(-4)). The two most common protein-truncating variants (p.Arg138* and p.Lys34Serfs*50) individually associate with T2D protection and encode unstable ZnT8 proteins. Previous functional study of SLC30A8 suggested that reduced zinc transport increases T2D risk, and phenotypic heterogeneity was observed in mouse Slc30a8 knockouts. In contrast, loss-of-function mutations in humans provide strong evidence that SLC30A8 haploinsufficiency protects against T2D, suggesting ZnT8 inhibition as a therapeutic strategy in T2D prevention.


Asunto(s)
Proteínas de Transporte de Catión/genética , Diabetes Mellitus Tipo 2/genética , Mutación Missense/genética , Animales , Secuencia de Bases , Glucemia/genética , Estudios de Asociación Genética , Genotipo , Humanos , Transporte Iónico/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proinsulina/sangre , Análisis de Secuencia de ADN , Transportador 8 de Zinc
13.
Nature ; 506(7486): 97-101, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24390345

RESUMEN

Performing genetic studies in multiple human populations can identify disease risk alleles that are common in one population but rare in others, with the potential to illuminate pathophysiology, health disparities, and the population genetic origins of disease alleles. Here we analysed 9.2 million single nucleotide polymorphisms (SNPs) in each of 8,214 Mexicans and other Latin Americans: 3,848 with type 2 diabetes and 4,366 non-diabetic controls. In addition to replicating previous findings, we identified a novel locus associated with type 2 diabetes at genome-wide significance spanning the solute carriers SLC16A11 and SLC16A13 (P = 3.9 × 10(-13); odds ratio (OR) = 1.29). The association was stronger in younger, leaner people with type 2 diabetes, and replicated in independent samples (P = 1.1 × 10(-4); OR = 1.20). The risk haplotype carries four amino acid substitutions, all in SLC16A11; it is present at ~50% frequency in Native American samples and ~10% in east Asian, but is rare in European and African samples. Analysis of an archaic genome sequence indicated that the risk haplotype introgressed into modern humans via admixture with Neanderthals. The SLC16A11 messenger RNA is expressed in liver, and V5-tagged SLC16A11 protein localizes to the endoplasmic reticulum. Expression of SLC16A11 in heterologous cells alters lipid metabolism, most notably causing an increase in intracellular triacylglycerol levels. Despite type 2 diabetes having been well studied by genome-wide association studies in other populations, analysis in Mexican and Latin American individuals identified SLC16A11 as a novel candidate gene for type 2 diabetes with a possible role in triacylglycerol metabolism.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad/genética , Transportadores de Ácidos Monocarboxílicos/genética , Polimorfismo de Nucleótido Simple/genética , Alelos , Animales , Pueblo Asiatico/genética , Población Negra/genética , Estudios de Cohortes , Retículo Endoplásmico/genética , Femenino , Estudio de Asociación del Genoma Completo , Haplotipos/genética , Células HeLa , Humanos , Indígenas Norteamericanos/genética , Metabolismo de los Lípidos/genética , Hígado/citología , Hígado/metabolismo , Masculino , México , Hombre de Neandertal/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Triglicéridos/metabolismo , Población Blanca/genética
14.
Dermatol Res Pract ; 2010: 759731, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20628490

RESUMEN

Epithelial wound closure is a complex biological process that relies on the concerted action of activated keratinocytes and dermal fibroblasts to resurface and close the exposed wound. Modulation of cell-cell adhesion junctions is thought to facilitate cellular proliferation and migration of keratinocytes across the wound. In particular, desmosomes, adhesion complexes critical for maintaining epithelial integrity, are downregulated at the wound edge. It is unclear, however, how compromised desmosomal adhesion would affect wound reepithelialization, given the need for a delicate balance between downmodulating adhesive strength to permit changes in cellular morphology and maintaining adhesion to allow coordinated migration of keratinocyte sheets. Here, we explore the contribution of desmosomal adhesion to wound healing using mice deficient for the desmosomal component Perp. We find that Perp conditional knockout mice display delayed wound healing relative to controls. Furthermore, we determine that while loss of Perp compromises cell-cell adhesion, it does not impair keratinocyte proliferation and actually enhances keratinocyte migration in in vitro assays. Thus, Perp's role in promoting cell adhesion is essential for wound closure. Together, these studies suggest a role for desmosomal adhesion in efficient wound healing.

15.
Mol Cell ; 30(3): 303-14, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18471976

RESUMEN

The p53 tumor suppressor restricts tumorigenesis through the transcriptional activation of target genes involved in cell-cycle arrest and apoptosis. Here, we identify Prl-3 (phosphatase of regenerating liver-3) as a p53-inducible gene. Whereas previous studies implicated Prl-3 in metastasis because of its overexpression in metastatic human colorectal cancer and its ability to promote invasiveness and motility, we demonstrate here that Prl-3 is an important cell-cycle regulator. Consistent with a role in DNA damage-induced cell-cycle arrest, Prl-3 overexpression induces G(1) arrest downstream of p53 by triggering a PI3K-Akt-activated negative feedback loop. Surprisingly, attenuation of Prl-3 expression also elicits an arrest response, suggesting that basal level Prl-3 expression is pivotal for normal cell-cycle progression. Our findings highlight key dose-dependent functions of Prl-3 in both positive and negative regulation of cell-cycle progression and provide insight into Prl-3's role in cancer progression.


Asunto(s)
Ciclo Celular/fisiología , Proteínas Inmediatas-Precoces/genética , Metástasis de la Neoplasia/genética , Proteínas Tirosina Fosfatasas/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células Cultivadas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fibroblastos/citología , Fibroblastos/fisiología , Regulación de la Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/genética
16.
J Biol Chem ; 279(1): 152-62, 2004 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-14570911

RESUMEN

Regulation of follicle-stimulating hormone (FSH) synthesis is a central point of convergence for signals controlling reproduction. The FSHbeta subunit is primarily regulated by gonadotropin-releasing hormone (GnRH), gonadal steroids, and activin. Here, we identify elements in the mouse FSHbeta promoter responsible for GnRH-mediated induction utilizing the LbetaT2 cell line that endogenously expresses FSH. The proximal 398 bp of the mouse FSHbeta promoter is sufficient for response to GnRH. This response localizes primarily to an AP-1 half-site (-72/-69) juxtaposed to a CCAAT box, which binds nuclear factor-Y. Both elements are required for AP-1 binding, creating a novel AP-1 site. Multimers of this site confer GnRH induction, and mutation or internal deletion of this site reduces GnRH induction by 35%. The same reduction was achieved using a dominant negative Fos protein. This is the only functional AP-1 site identified in the proximal 398 bp, since its mutation eliminates FSHbeta induction by c-Fos and c-Jun. GnRH regulation of the FSHbeta gene occurs through induction of multiple Fos and Jun isoforms, forming at least four different AP-1 molecules, all of which bind to this site. Mitogen-activated protein kinase activity is required for induction of FSHbeta and JunB protein. Finally, AP-1 interacts with nuclear factor-Y, which occupies its overlapping site in vivo.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/genética , Regulación de la Expresión Génica/fisiología , Hormona Liberadora de Gonadotropina/farmacología , Factor de Transcripción AP-1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Bovinos , Regulación de la Expresión Génica/efectos de los fármacos , Genes fos , Genes jun , Humanos , Células L , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Oligodesoxirribonucleótidos , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Ovinos , Factor de Transcripción AP-1/química , Transcripción Genética , Transfección
17.
Mol Endocrinol ; 17(8): 1470-83, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12730328

RESUMEN

FSH is a heterodimeric glycoprotein hormone secreted from the gonadotrope cell population of the anterior pituitary. Despite its crucial role in mammalian reproduction, very little is known about regulation of the FSH beta-subunit gene at the molecular level. In this report, we examine the basis for cell-specific expression of FSH beta using the mouse L beta T2 and alpha T3-1 gonadotrope-derived cell lines. Characterization of the hormonal content of L beta T2 and alpha T3-1 cells at the protein level classifies these cells as relatively mature and immature gonadotropes, respectively. We studied L beta T2 cell-specific expression of FSH beta using 398 bp of the mouse FSH beta regulatory region linked to a luciferase reporter gene in transient transfection assays. This mouse FSH beta promoter can direct reporter gene expression specifically to L beta T2 cells when compared with other pituitary- and non-pituitary-derived cell lines, including alpha T3-1 cells. Furthermore, it is induced by activin, and interruption of the autocrine activin loop in L beta T2 cells by the addition of follistatin reduces its expression. Truncation analysis indicates that several regions of the promoter are involved in this specificity and that these can be dissociated from activin regulation. We identify binding sites for the orphan nuclear receptor steroidogenic factor-1 and the heterotrimeric transcription factor nuclear factor Y and show that these elements functionally interact to regulate FSH beta gene expression in an L beta T2 cell-specific manner. Moreover, steroidogenic factor-1 and nuclear factor Y are shown to physically interact with each other. This study is the first to demonstrate the presence of basal FSH beta protein in L beta T2 cells and to identify specific elements within the FSH beta promoter that contribute to basal and cell-specific expression of the gene.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Hormona Folículo Estimulante de Subunidad beta/genética , Adenohipófisis/metabolismo , Factores de Transcripción/metabolismo , Células 3T3/metabolismo , Activinas/farmacología , Animales , Sitios de Unión , Factor de Unión a CCAAT/efectos de los fármacos , Factor de Unión a CCAAT/genética , Células Cultivadas , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Hormona Folículo Estimulante de Subunidad beta/efectos de los fármacos , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Folistatina/farmacología , Factores de Transcripción Fushi Tarazu , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Proteínas de Homeodominio , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Especificidad de Órganos , Adenohipófisis/citología , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Factor Esteroidogénico 1 , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...