Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 165(1): 125-138, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27015310

RESUMEN

Activation of Agouti-related peptide (AgRP) neurons potently promotes feeding, and chronically altering their activity also affects peripheral glucose homeostasis. We demonstrate that acute activation of AgRP neurons causes insulin resistance through impairment of insulin-stimulated glucose uptake into brown adipose tissue (BAT). AgRP neuron activation acutely reprograms gene expression in BAT toward a myogenic signature, including increased expression of myostatin. Interference with myostatin activity improves insulin sensitivity that was impaired by AgRP neurons activation. Optogenetic circuitry mapping reveals that feeding and insulin sensitivity are controlled by both distinct and overlapping projections. Stimulation of AgRP → LHA projections impairs insulin sensitivity and promotes feeding while activation of AgRP → anterior bed nucleus of the stria terminalis (aBNST)vl projections, distinct from AgRP → aBNSTdm projections controlling feeding, mediate the effect of AgRP neuron activation on BAT-myostatin expression and insulin sensitivity. Collectively, our results suggest that AgRP neurons in mice induce not only eating, but also insulin resistance by stimulating expression of muscle-related genes in BAT, revealing a mechanism by which these neurons rapidly coordinate hunger states with glucose homeostasis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Regulación del Apetito , Glucosa/metabolismo , Resistencia a la Insulina , Neuronas/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Conducta Alimentaria , Ratones , Miostatina/genética , Optogenética , Transcriptoma
2.
Hum Mol Genet ; 32(7): 1083-1089, 2023 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-36300302

RESUMEN

Auditory synaptopathy/neuropathy (AS/AN) is a distinct type of sensorineural hearing loss in which the cochlear sensitivity to sound (i.e. active cochlear amplification by outer hair cells) is preserved whereas sound encoding by inner hair cells and/or auditory nerve fibers is disrupted owing to genetic or environmental factors. Autosomal-dominant auditory neuropathy type 2 (AUNA2) was linked either to chromosomal bands 12q24 or 13q34 in a large German family in 2017. By whole-genome sequencing, we now detected a 5500 bp deletion in ATP11A on chromosome 13q34 segregating with the phenotype in this family. ATP11A encodes a P-type ATPase that translocates phospholipids from the exoplasmic to the cytoplasmic leaflet of the plasma membrane. The deletion affects both isoforms of ATP11A and activates a cryptic splice site leading to the formation of an alternative last exon. ATP11A carrying the altered C-terminus loses its flippase activity for phosphatidylserine. Atp11a is expressed in fibers and synaptic contacts of the auditory nerve and in the cochlear nucleus in mice, and conditional Atp11a knockout mice show a progressive reduction of the spiral ganglion neuron compound action potential, recapitulating the human phenotype of AN. By combining whole-genome sequencing, immunohistochemistry, in vitro functional assays and generation of a mouse model, we could thus identify a partial deletion of ATP11A as the genetic cause of AUNA2.


Asunto(s)
Pérdida Auditiva Central , Pérdida Auditiva Sensorineural , Humanos , Ratones , Animales , Pérdida Auditiva Central/genética , Pérdida Auditiva Sensorineural/genética , Mutación , Células Ciliadas Auditivas Internas , Cromosomas , Transportadoras de Casetes de Unión a ATP/genética
3.
PLoS Pathog ; 17(9): e1009943, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34555129

RESUMEN

Regulation of cellular metabolism is now recognized as a crucial mechanism for the activation of innate and adaptive immune cells upon diverse extracellular stimuli. Macrophages, for instance, increase glycolysis upon stimulation with pathogen-associated molecular patterns (PAMPs). Conceivably, pathogens also counteract these metabolic changes for their own survival in the host. Despite this dynamic interplay in host-pathogen interactions, the role of immunometabolism in the context of intracellular bacterial infections is still unclear. Here, employing unbiased metabolomic and transcriptomic approaches, we investigated the role of metabolic adaptations of macrophages upon Salmonella enterica serovar Typhimurium (S. Typhimurium) infections. Importantly, our results suggest that S. Typhimurium abrogates glycolysis and its modulators such as insulin-signaling to impair macrophage defense. Mechanistically, glycolysis facilitates glycolytic enzyme aldolase A mediated v-ATPase assembly and the acidification of phagosomes which is critical for lysosomal degradation. Thus, impairment in the glycolytic machinery eventually leads to decreased bacterial clearance and antigen presentation in murine macrophages (BMDM). Collectively, our results highlight a vital molecular link between metabolic adaptation and phagosome maturation in macrophages, which is targeted by S. Typhimurium to evade cell-autonomous defense.


Asunto(s)
Glucólisis/fisiología , Interacciones Huésped-Patógeno/fisiología , Macrófagos/metabolismo , Fagosomas/metabolismo , Salmonelosis Animal/metabolismo , Animales , Perfilación de la Expresión Génica , Metabolómica , Ratones , Salmonella typhimurium/metabolismo
4.
Mol Cell ; 48(4): 641-6, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23063529

RESUMEN

Together with GTP and initiator methionyl-tRNA, translation initiation factor eIF2 forms a ternary complex that binds the 40S ribosome and then scans an mRNA to select the AUG start codon for protein synthesis. Here, we show that a human X-chromosomal neurological disorder characterized by intellectual disability and microcephaly is caused by a missense mutation in eIF2γ (encoded by EIF2S3), the core subunit of the heterotrimeric eIF2 complex. Biochemical studies of human cells overexpressing the eIF2γ mutant and of yeast eIF2γ with the analogous mutation revealed a defect in binding the eIF2ß subunit to eIF2γ. Consistent with this loss of eIF2 integrity, the yeast eIF2γ mutation impaired translation start codon selection and eIF2 function in vivo in a manner that was suppressed by overexpressing eIF2ß. These findings directly link intellectual disability to impaired translation initiation, and provide a mechanistic basis for the human disease due to partial loss of eIF2 function.


Asunto(s)
Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Discapacidad Intelectual/genética , Iniciación de la Cadena Peptídica Traduccional/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Secuencia de Bases , Factor 2 Eucariótico de Iniciación/química , Humanos , Modelos Moleculares , Mutación Missense , Proteínas de Saccharomyces cerevisiae/química
5.
BMC Genomics ; 20(1): 85, 2019 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-30678634

RESUMEN

BACKGROUND: Next-Generation Sequencing (NGS) has been widely accepted as an essential tool in molecular biology. Reduced costs and automated analysis pipelines make the use of NGS data feasible even for small labs, yet the methods for interpreting the data are not sophisticated enough to account for the amount of information. RESULTS: We propose s ·nr, a Visual Analytics tool that provides simple yet powerful visual interfaces for displaying and querying NGS data. It allows researchers to explore their own data in the context of experimental data deposited in public repositories, as well as to extract specific data sets with similar gene expression signatures. We tested s ·nr on 1543 RNA-Seq based mouse differential expression profiles derived from the public ArrayExpress platform. We provide the repository of processed data with this paper. CONCLUSION: s ·nr, easily deployable utilizing its containerized implementation, empowers researchers to analyze and relate their own RNA-Seq as well as to provide interactive and contextual crosstalk with data from public repositories. This allows users to deduce novel and unbiased hypotheses about the underlying molecular processes. DEMO: Login demo/demo: snr.sf.mpg.de (Tested with Google Chrome).


Asunto(s)
Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Animales , Gráficos por Computador , Ratones , Interfaz Usuario-Computador
6.
BMC Bioinformatics ; 19(1): 156, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29699486

RESUMEN

BACKGROUND: Recent cancer genome studies on many human cancer types have relied on multiple molecular high-throughput technologies. Given the vast amount of data that has been generated, there are surprisingly few databases which facilitate access to these data and make them available for flexible analysis queries in the broad research community. If used in their entirety and provided at a high structural level, these data can be directed into constantly increasing databases which bear an enormous potential to serve as a basis for machine learning technologies with the goal to support research and healthcare with predictions of clinically relevant traits. RESULTS: We have developed the Cancer Systems Biology Database (CancerSysDB), a resource for highly flexible queries and analysis of cancer-related data across multiple data types and multiple studies. The CancerSysDB can be adopted by any center for the organization of their locally acquired data and its integration with publicly available data from multiple studies. A publicly available main instance of the CancerSysDB can be used to obtain highly flexible queries across multiple data types as shown by highly relevant use cases. In addition, we demonstrate how the CancerSysDB can be used for predictive cancer classification based on whole-exome data from 9091 patients in The Cancer Genome Atlas (TCGA) research network. CONCLUSIONS: Our database bears the potential to be used for large-scale integrative queries and predictive analytics of clinically relevant traits.


Asunto(s)
Bases de Datos Factuales , Genómica/métodos , Aprendizaje Automático , Neoplasias/genética , Programas Informáticos , Biología de Sistemas , Exoma , Humanos
7.
Hum Mutat ; 38(10): 1325-1335, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28598576

RESUMEN

Next-generation sequencing (NGS) has turned from a new and experimental technology into a standard procedure for cancer genome studies and clinical investigation. While a multitude of software packages for cancer genome data analysis have been made available, these need to be combined into efficient analytical workflows that cover multiple aspects relevant to a clinical environment and that deliver handy results within a reasonable time frame. Here, we introduce QuickNGS Cancer as a new suite of bioinformatics pipelines that is focused on cancer genomics and significantly reduces the analytical hurdles that still limit a broader applicability of NGS technology, particularly to clinically driven research. QuickNGS Cancer allows a highly efficient analysis of a broad variety of NGS data types, specifically considering cancer-specific issues, such as biases introduced by tumor impurity and aneuploidy or the assessment of genomic variations regarding their biomedical relevance. It delivers highly reproducible analysis results ready for interpretation within only a few days after sequencing, as shown by a reanalysis of 140 tumor/normal pairs from The Cancer Genome Atlas (TCGA) in which QuickNGS Cancer detected a significant number of mutations in key cancer genes missed by a well-established mutation calling pipeline. Finally, QuickNGS Cancer obtained several unexpected mutations in leukemias that could be confirmed by Sanger sequencing.


Asunto(s)
Genoma Humano/genética , Mutación/genética , Neoplasias/genética , Programas Informáticos , Biología Computacional , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ADN , Flujo de Trabajo
8.
BMC Genomics ; 17(1): 855, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27809788

RESUMEN

BACKGROUND: Although the pattern of lateral organ formation from apical meristems establishes species-specific plant architecture, the positional information that confers cell fate to cells as they transit to the meristem flanks where they differentiate, remains largely unknown. We have combined fluorescence-activated cell sorting and RNA-seq to characterise the cell-type-specific transcriptome at the earliest developmental time-point of lateral organ formation using DORNRÖSCHEN-LIKE::GFP to mark founder-cell populations at the periphery of the inflorescence meristem (IM) in apetala1 cauliflower double mutants, which overproliferate IMs. RESULTS: Within the lateral organ founder-cell population at the inflorescence meristem, floral primordium identity genes are upregulated and stem-cell identity markers are downregulated. Additional differentially expressed transcripts are involved in polarity generation and boundary formation, and in epigenetic and post-translational changes. However, only subtle transcriptional reprogramming within the global auxin network was observed. CONCLUSIONS: The transcriptional network of differentially expressed genes supports the hypothesis that lateral organ founder-cell specification involves the creation of polarity from the centre to the periphery of the IM and the establishment of a boundary from surrounding cells, consistent with bract initiation. However, contrary to the established paradigm that sites of auxin response maxima pre-pattern lateral organ initiation in the IM, auxin response might play a minor role in the earliest stages of lateral floral initiation.


Asunto(s)
Brassica/genética , Inflorescencia/genética , Meristema/genética , Transcriptoma , Análisis por Conglomerados , Biología Computacional/métodos , Epigénesis Genética , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Ontología de Genes , Redes Reguladoras de Genes , Genes Reporteros , Fenotipo , Células Vegetales/metabolismo , Procesamiento Postranscripcional del ARN
9.
BMC Genomics ; 16: 487, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26126663

RESUMEN

BACKGROUND: Next-Generation Sequencing (NGS) has emerged as a widely used tool in molecular biology. While time and cost for the sequencing itself are decreasing, the analysis of the massive amounts of data remains challenging. Since multiple algorithmic approaches for the basic data analysis have been developed, there is now an increasing need to efficiently use these tools to obtain results in reasonable time. RESULTS: We have developed QuickNGS, a new workflow system for laboratories with the need to analyze data from multiple NGS projects at a time. QuickNGS takes advantage of parallel computing resources, a comprehensive back-end database, and a careful selection of previously published algorithmic approaches to build fully automated data analysis workflows. We demonstrate the efficiency of our new software by a comprehensive analysis of 10 RNA-Seq samples which we can finish in only a few minutes of hands-on time. The approach we have taken is suitable to process even much larger numbers of samples and multiple projects at a time. CONCLUSION: Our approach considerably reduces the barriers that still limit the usability of the powerful NGS technology and finally decreases the time to be spent before proceeding to further downstream analysis and interpretation of the data.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ARN/métodos , Animales , Automatización de Laboratorios , Bases de Datos Genéticas , Humanos , Programas Informáticos , Factores de Tiempo , Flujo de Trabajo
10.
Hum Mol Genet ; 22(25): 5199-214, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23918663

RESUMEN

Autosomal recessive primary microcephaly (MCPH) is characterized by reduced head circumference, reduction in the size of the cerebral cortex with otherwise grossly normal brain structure and variable intellectual disability. MCPH is caused by mutations of 11 different genes which code for proteins implicated in cell division and cell cycle regulation. We studied a consanguineous eight-generation family from Pakistan with ten microcephalic children using homozygosity mapping and found a new MCPH locus at HSA 7q21.11-q21.3. Sanger sequencing of the most relevant candidate genes in this region revealed a homozygous single nucleotide substitution c.589G>A in CDK6, which encodes cyclin-dependent kinase 6. The mutation changes a highly conserved alanine at position 197 into threonine (p.Ala197Thr). Post hoc whole-exome sequencing corroborated this mutation's identification as the causal variant. CDK6 is an important protein for the control of the cell cycle and differentiation of various cell types. We show here for the first time that CDK6 associates with the centrosome during mitosis; however, this was not observed in patient fibroblasts. Moreover, the mutant primary fibroblasts exhibited supernumerary centrosomes, disorganized microtubules and mitotic spindles, an increased centrosome nucleus distance, reduced cell proliferation and impaired cell motility and polarity. Upon ectopic expression of the mutant protein and knockdown of CDK6 through shRNA, we noted similar effects. We propose that the identified CDK6 mutation leads to reduced cell proliferation and impairs the correct functioning of the centrosome in microtubule organization and its positioning near the nucleus which are key determinants during neurogenesis.


Asunto(s)
Centrosoma/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Mitosis/genética , Mapeo Cromosómico , Cromosomas Humanos Par 7/genética , Quinasa 6 Dependiente de la Ciclina/química , Quinasa 6 Dependiente de la Ciclina/metabolismo , Femenino , Estudios de Asociación Genética , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Microcefalia/fisiopatología , Microtúbulos/genética , Microtúbulos/metabolismo , Mutación , Linaje , Polimorfismo de Nucleótido Simple , Conformación Proteica
11.
Am J Hum Genet ; 90(5): 871-8, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22521416

RESUMEN

Autosomal-recessive primary microcephaly (MCPH) is a rare congenital disorder characterized by intellectual disability, reduced brain and head size, but usually without defects in cerebral cortical architecture, and other syndromic abnormalities. MCPH is heterogeneous. The underlying genes of the seven known loci code for centrosomal proteins. We studied a family from northern Pakistan with two microcephalic children using homozygosity mapping and found suggestive linkage for regions on chromosomes 2, 4, and 9. We sequenced two positional candidate genes and identified a homozygous frameshift mutation in the gene encoding the 135 kDa centrosomal protein (CEP135), located in the linkage interval on chromosome 4, in both affected children. Post hoc whole-exome sequencing corroborated this mutation's identification as the causal variant. Fibroblasts obtained from one of the patients showed multiple and fragmented centrosomes, disorganized microtubules, and reduced growth rate. Similar effects were reported after knockdown of CEP135 through RNA interference; we could provoke them also by ectopic overexpression of the mutant protein. Our findings suggest an additional locus for MCPH at HSA 4q12 (MCPH8), further strengthen the role of centrosomes in the development of MCPH, and place CEP135 among the essential components of this important organelle in particular for a normal neurogenesis.


Asunto(s)
Proteínas Portadoras/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Mutación , Proteínas Portadoras/metabolismo , Centrosoma , Niño , Cromosomas Humanos Par 4/genética , Cromosomas Humanos Par 4/metabolismo , Exoma , Exones , Femenino , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Sitios Genéticos , Homocigoto , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Microcefalia/fisiopatología , Pakistán/epidemiología , Linaje , Polimorfismo de Nucleótido Simple , Interferencia de ARN , Análisis de Secuencia de ADN
12.
Am J Hum Genet ; 90(4): 661-74, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22482805

RESUMEN

Bone morphogenetic protein 1 (BMP1) is an astacin metalloprotease with important cellular functions and diverse substrates, including extracellular-matrix proteins and antagonists of some TGFß superfamily members. Combining whole-exome sequencing and filtering for homozygous stretches of identified variants, we found a homozygous causative BMP1 mutation, c.34G>C, in a consanguineous family affected by increased bone mineral density and multiple recurrent fractures. The mutation is located within the BMP1 signal peptide and leads to impaired secretion and an alteration in posttranslational modification. We also characterize a zebrafish bone mutant harboring lesions in bmp1a, demonstrating conservation of BMP1 function in osteogenesis across species. Genetic, biochemical, and histological analyses of this mutant and a comparison to a second, similar locus reveal that Bmp1a is critically required for mature-collagen generation, downstream of osteoblast maturation, in bone. We thus define the molecular and cellular bases of BMP1-dependent osteogenesis and show the importance of this protein for bone formation and stability.


Asunto(s)
Proteína Morfogenética Ósea 1/fisiología , Osteogénesis/genética , Osteogénesis/fisiología , Animales , Secuencia de Bases , Conservadores de la Densidad Ósea/uso terapéutico , Proteína Morfogenética Ósea 1/genética , Proteína Morfogenética Ósea 1/metabolismo , Huesos/metabolismo , Diferenciación Celular , Preescolar , Colágeno/biosíntesis , Difosfonatos/uso terapéutico , Exoma , Femenino , Fracturas Óseas/tratamiento farmacológico , Fracturas Óseas/prevención & control , Sitios Genéticos , Proteínas de Choque Térmico , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Fragmentos de Péptidos , Procesamiento Proteico-Postraduccional , Pez Cebra/genética , Pez Cebra/metabolismo
13.
Am J Hum Genet ; 91(6): 998-1010, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23200864

RESUMEN

Ubiquitination plays a crucial role in neurodevelopment as exemplified by Angelman syndrome, which is caused by genetic alterations of the ubiquitin ligase-encoding UBE3A gene. Although the function of UBE3A has been widely studied, little is known about its paralog UBE3B. By using exome and capillary sequencing, we here identify biallelic UBE3B mutations in four patients from three unrelated families presenting an autosomal-recessive blepharophimosis-ptosis-intellectual-disability syndrome characterized by developmental delay, growth retardation with a small head circumference, facial dysmorphisms, and low cholesterol levels. UBE3B encodes an uncharacterized E3 ubiquitin ligase. The identified UBE3B variants include one frameshift and two splice-site mutations as well as a missense substitution affecting the highly conserved HECT domain. Disruption of mouse Ube3b leads to reduced viability and recapitulates key aspects of the human disorder, such as reduced weight and brain size and a downregulation of cholesterol synthesis. We establish that the probable Caenorhabditis elegans ortholog of UBE3B, oxi-1, functions in the ubiquitin/proteasome system in vivo and is especially required under oxidative stress conditions. Our data reveal the pleiotropic effects of UBE3B deficiency and reinforce the physiological importance of ubiquitination in neuronal development and function in mammals.


Asunto(s)
Blefarofimosis/genética , Blefaroptosis/genética , Discapacidad Intelectual/genética , Ubiquitina-Proteína Ligasas/genética , Alelos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Blefarofimosis/diagnóstico , Blefaroptosis/diagnóstico , Encéfalo/patología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Sistema Nervioso Central , Niño , Preescolar , Exoma , Facies , Femenino , Genotipo , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Mutación , Estrés Oxidativo , Síndrome , Ubiquitina-Proteína Ligasas/deficiencia
14.
Biochim Biophys Acta ; 1833(5): 1190-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23391410

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer deaths, worldwide. MicroRNAs, inhibiting gene expression by targeting various transcripts, are involved in genomic dysregulation during hepatocellular tumorigenesis. In previous studies, microRNA-198 (miR-198) was shown to be significantly downregulated in HCV-positive hepatocellular carcinoma (HCC). Herein, the function of miR-198 in hepatocellular carcinoma cell growth and gene expression was studied. In hepatoma cell-types with low levels of liver-specific transcription factor HNF1α indicating a low differentiation grade, miR-198 expression was most downregulated. However, miR-198 treatment did not restore the expression of the liver-specific transcription factors HNF1α or HNF4α. Importantly, overexpression of miR-198 in Pop10 hepatoma cells markedly reduced cell growth. In agreement, comprehensive gene expression profiling by microarray hybridisation and real-time quantification revealed that central signal transducers of proliferation pathways were downregulated by miR-198. In contrast, genes mediating cellular adherence were highly upregulated by miR-198. Thus, the low expression of E-cadherin and claudin-1, involved in cell adhesion and cell-cell contacts, was abolished in hepatoma cells after miR-198 overexpression. This definite induction of both proteins by miR-198 was shown to be accompanied by a significantly impaired migration activity of hepatoma Pop10 cells. In conclusion, miR-198 acts as a tumor suppressor by repression of mitogenic and motogenic pathways diminishing cell growth and migration.


Asunto(s)
Carcinoma Hepatocelular , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , MicroARNs , Cadherinas/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Claudina-1/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo
15.
BMC Genomics ; 15: 776, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25199885

RESUMEN

BACKGROUND: Cyanobacteria constitute a serious threat to freshwater ecosystems by producing toxic secondary metabolites, e.g. microcystins. These microcystins have been shown to harm livestock, pets and humans and to affect ecosystem service and functioning. Cyanobacterial blooms are increasing worldwide in intensity and frequency due to eutrophication and global warming. However, Daphnia, the main grazer of planktonic algae and cyanobacteria, has been shown to be able to suppress bloom-forming cyanobacteria and to adapt to cyanobacteria that produce microcystins. Since Daphnia's genome was published only recently, it is now possible to elucidate the underlying molecular mechanisms of microcystin tolerance of Daphnia. RESULTS: Daphnia magna was fed with either a cyanobacterial strain that produces microcystins or its genetically engineered microcystin knock-out mutant. Thus, it was possible to distinguish between effects due to the ingestion of cyanobacteria and effects caused specifically by microcystins. By using RNAseq the differentially expressed genes between the different treatments were analyzed and affected KOG-categories were calculated. Here we show that the expression of transporter genes in Daphnia was regulated as a specific response to microcystins. Subsequent qPCR and dietary supplementation with pure microcystin confirmed that the regulation of transporter gene expression was correlated with the tolerance of several Daphnia clones. CONCLUSIONS: Here, we were able to identify new candidate genes that specifically respond to microcystins by separating cyanobacterial effects from microcystin effects. The involvement of these candidate genes in tolerance to microcystins was validated by correlating the difference in transporter gene expression with clonal tolerance. Thus, the prevention of microcystin uptake most probably constitutes a key mechanism in the development of tolerance and adaptation of Daphnia. With the availability of clear candidate genes, future investigations examining the process of local adaptation of Daphnia populations to microcystins are now possible.


Asunto(s)
Biotransformación/genética , Daphnia/genética , Daphnia/metabolismo , Microcistinas/metabolismo , Animales , Cianobacterias/metabolismo , Ecosistema , Perfilación de la Expresión Génica , Reproducibilidad de los Resultados , Transcriptoma
16.
Am J Hum Genet ; 88(5): 621-7, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21549336

RESUMEN

The fact that hereditary hearing loss is the most common sensory disorder in humans is reflected by, among other things, an extraordinary allelic and nonallelic genetic heterogeneity. X-chromosomal hearing impairment represents only a minor fraction of all cases. In a study of a Spanish family the locus for one of the X-chromosomal forms was assigned to Xp22 (DFNX4). We mapped the disease locus in the same chromosomal region in a large German pedigree with X-chromosomal nonsyndromic hearing impairment by using genome-wide linkage analysis. Males presented with postlingual hearing loss and onset at ages 3-7, whereas onset in female carriers was in the second to third decades. Targeted DNA capture with high-throughput sequencing detected a nonsense mutation in the small muscle protein, X-linked (SMPX) of affected individuals. We identified another nonsense mutation in SMPX in patients from the Spanish family who were previously analyzed to map DFNX4. SMPX encodes an 88 amino acid, cytoskeleton-associated protein that is responsive to mechanical stress. The presence of Smpx in hair cells and supporting cells of the murine cochlea indicates its role in the inner ear. The nonsense mutations detected in the two families suggest a loss-of-function mechanism underlying this form of hearing impairment. Results obtained after heterologous overexpression of SMPX proteins were compatible with this assumption. Because responsivity to physical force is a characteristic feature of the protein, we propose that long-term maintenance of mechanically stressed inner-ear cells critically depends on SMPX function.


Asunto(s)
Cromosomas Humanos X/genética , Codón sin Sentido , Pérdida Auditiva/genética , Proteínas Musculares/genética , Adolescente , Edad de Inicio , Alelos , Animales , Niño , Preescolar , Cóclea , Oído Interno/embriología , Oído Interno/metabolismo , Femenino , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Células Ciliadas Auditivas/metabolismo , Haplotipos , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Linaje
17.
Am J Hum Genet ; 89(5): 668-74, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22077972

RESUMEN

Urinary bladder malformations associated with bladder outlet obstruction are a frequent cause of progressive renal failure in children. We here describe a muscarinic acetylcholine receptor M3 (CHRM3) (1q41-q44) homozygous frameshift mutation in familial congenital bladder malformation associated with a prune-belly-like syndrome, defining an isolated gene defect underlying this sometimes devastating disease. CHRM3 encodes the M3 muscarinic acetylcholine receptor, which we show is present in developing renal epithelia and bladder muscle. These observations may imply that M3 has a role beyond its known contribution to detrusor contractions. This Mendelian disease caused by a muscarinic acetylcholine receptor mutation strikingly phenocopies Chrm3 null mutant mice.


Asunto(s)
Errores Innatos del Metabolismo/genética , Síndrome del Abdomen en Ciruela Pasa/genética , Receptor Muscarínico M3 , Vejiga Urinaria , Animales , Secuencia de Bases , Consanguinidad , Femenino , Mutación del Sistema de Lectura/genética , Humanos , Mutación INDEL/genética , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Síndrome del Abdomen en Ciruela Pasa/patología , Receptor Muscarínico M3/deficiencia , Receptor Muscarínico M3/genética , Homología de Secuencia de Ácido Nucleico , Factores Sexuales , Vejiga Urinaria/embriología , Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/genética , Obstrucción del Cuello de la Vejiga Urinaria/patología
18.
Nat Metab ; 6(6): 1053-1075, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38684889

RESUMEN

Promoting brown adipose tissue (BAT) activity innovatively targets obesity and metabolic disease. While thermogenic activation of BAT is well understood, the rheostatic regulation of BAT to avoid excessive energy dissipation remains ill-defined. Here, we demonstrate that adenylyl cyclase 3 (AC3) is key for BAT function. We identified a cold-inducible promoter that generates a 5' truncated AC3 mRNA isoform (Adcy3-at), whose expression is driven by a cold-induced, truncated isoform of PPARGC1A (PPARGC1A-AT). Male mice lacking Adcy3-at display increased energy expenditure and are resistant to obesity and ensuing metabolic imbalances. Mouse and human AC3-AT are retained in the endoplasmic reticulum, unable to translocate to the plasma membrane and lack enzymatic activity. AC3-AT interacts with AC3 and sequesters it in the endoplasmic reticulum, reducing the pool of adenylyl cyclases available for G-protein-mediated cAMP synthesis. Thus, AC3-AT acts as a cold-induced rheostat in BAT, limiting adverse consequences of cAMP activity during chronic BAT activation.


Asunto(s)
Adenilil Ciclasas , Tejido Adiposo Pardo , Frío , Adenilil Ciclasas/metabolismo , Adenilil Ciclasas/genética , Tejido Adiposo Pardo/metabolismo , Animales , Ratones , Masculino , Humanos , Termogénesis/genética , Metabolismo Energético , AMP Cíclico/metabolismo , Ratones Noqueados
19.
BMC Genomics ; 14: 559, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23947592

RESUMEN

BACKGROUND: Aging-related kidney diseases are a major health concern. Currently, models to study renal aging are lacking. Due to a reduced life-span progeroid models hold the promise to facilitate aging studies and allow examination of tissue-specific changes. Defects in genome maintenance in the Ercc1(-/Δ) progeroid mouse model result in premature aging and typical age-related pathologies. Here, we compared the glomerular transcriptome of young and aged Ercc1-deficient mice to young and aged WT mice in order to establish a novel model for research of aging-related kidney disease. RESULTS: In a principal component analysis, age and genotype emerged as first and second principal components. Hierarchical clustering of all 521 genes differentially regulated between young and old WT and young and old Ercc1(-/Δ) mice showed cluster formation between young WT and Ercc1(-/Δ) as well as old WT and Ercc1(-/Δ) samples. An unexpectedly high number of 77 genes were differentially regulated in both WT and Ercc1(-/Δ) mice (p < 0.0001). GO term enrichment analysis revealed these genes to be involved in immune and inflammatory response, cell death, and chemotaxis. In a network analysis, these genes were part of insulin signaling, chemokine and cytokine signaling and extracellular matrix pathways. CONCLUSION: Beyond insulin signaling, we find chemokine and cytokine signaling as well as modifiers of extracellular matrix composition to be subject to major changes in the aging glomerulus. At the level of the transcriptome, the pattern of gene activities is similar in the progeroid Ercc1(-/Δ) mouse model constituting a valuable tool for future studies of aging-associated glomerular pathologies.


Asunto(s)
Envejecimiento/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Perfilación de la Expresión Génica , Glomérulos Renales/metabolismo , Factores de Edad , Animales , Análisis por Conglomerados , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Enfermedades Renales/etiología , Masculino , Ratones , Ratones Noqueados , Anotación de Secuencia Molecular , Análisis de Componente Principal , Progeria/genética , Progeria/metabolismo , Transducción de Señal
20.
BMC Genomics ; 14: 923, 2013 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-24373391

RESUMEN

BACKGROUND: The genetics of development in the nematode Caenorhabditis elegans has been described in exquisite detail. The phylum Nematoda has two classes: Chromadorea (which includes C. elegans) and the Enoplea. While the development of many chromadorean species resembles closely that of C. elegans, enoplean nematodes show markedly different patterns of early cell division and cell fate assignment. Embryogenesis of the enoplean Romanomermis culicivorax has been studied in detail, but the genetic circuitry underpinning development in this species has not been explored. RESULTS: We generated a draft genome for R. culicivorax and compared its gene content with that of C. elegans, a second enoplean, the vertebrate parasite Trichinella spiralis, and a representative arthropod, Tribolium castaneum. This comparison revealed that R. culicivorax has retained components of the conserved ecdysozoan developmental gene toolkit lost in C. elegans. T. spiralis has independently lost even more of this toolkit than has C. elegans. However, the C. elegans toolkit is not simply depauperate, as many novel genes essential for embryogenesis in C. elegans are not found in, or have only extremely divergent homologues in R. culicivorax and T. spiralis. Our data imply fundamental differences in the genetic programmes not only for early cell specification but also others such as vulva formation and sex determination. CONCLUSIONS: Despite the apparent morphological conservatism, major differences in the molecular logic of development have evolved within the phylum Nematoda. R. culicivorax serves as a tractable system to contrast C. elegans and understand how divergent genomic and thus regulatory backgrounds nevertheless generate a conserved phenotype. The R. culicivorax draft genome will promote use of this species as a research model.


Asunto(s)
Evolución Biológica , Enóplidos/genética , Genoma de los Helmintos , Animales , Caenorhabditis elegans/genética , Enóplidos/crecimiento & desarrollo , Biblioteca de Genes , Transcriptoma , Tribolium/genética , Trichinella spiralis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA