Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(3): 113951, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38508192

RESUMEN

Plasticity in daily timing of activity has been observed in many species, yet the underlying mechanisms driving nocturnality and diurnality are unknown. By regulating how much wheel-running activity will be rewarded with a food pellet, we can manipulate energy balance and switch mice to be nocturnal or diurnal. Here, we present the rhythmic transcriptome of 21 tissues, including 17 brain regions, sampled every 4 h over a 24-h period from nocturnal and diurnal male CBA/CaJ mice. Rhythmic gene expression across tissues comprised different sets of genes with minimal overlap between nocturnal and diurnal mice. We show that non-clock genes in the suprachiasmatic nucleus (SCN) change, and the habenula was most affected. Our results indicate that adaptive flexibility in daily timing of behavior is supported by gene expression dynamics in many tissues and brain regions, especially in the habenula, which suggests a crucial role for the observed nocturnal-diurnal switch.


Asunto(s)
Ritmo Circadiano , Transcriptoma , Ratones , Masculino , Animales , Ritmo Circadiano/genética , Transcriptoma/genética , Ratones Endogámicos CBA , Encéfalo , Núcleo Supraquiasmático/metabolismo
2.
iScience ; 27(3): 109051, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38384840

RESUMEN

The hypothalamic suprachiasmatic nucleus (SCN) is composed of heterogenous populations of neurons that express signaling peptides such as vasoactive intestinal polypeptide (VIP) and arginine vasopressin (AVP) and regulate circadian rhythms in behavior and physiology. SCN neurons acquire functional and morphological specializations from waves of transcription factors (TFs) that are expressed during neurogenesis. However, the in vitro generation of SCN neurons has never been achieved. Here we supplemented a highly efficient neuronal conversion protocol with TFs that are expressed during SCN neurogenesis, namely Six3, Six6, Dlx2, and Lhx1. Neurons induced from mouse and human fibroblasts predominantly exhibited neuronal properties such as bipolar or multipolar morphologies, GABAergic neurons with expression of VIP. Our study reveals a critical contribution of these TFs to the development of vasoactive intestinal peptide (Vip) expressing neurons in the SCN, suggesting the regenerative potential of neuronal subtypes contained in the SCN for future SCN regeneration and in vitro disease remodeling.

3.
Cell Rep Med ; 4(2): 100935, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36758547

RESUMEN

Transcription factor programs mediating the immune response to coronavirus disease 2019 (COVID-19) are not fully understood. Capturing active transcription initiation from cis-regulatory elements such as enhancers and promoters by capped small RNA sequencing (csRNA-seq), in contrast to capturing steady-state transcripts by conventional RNA-seq, allows unbiased identification of the underlying transcription factor activity and regulatory pathways. Here, we profile transcription initiation in critically ill COVID-19 patients, identifying transcription factor motifs that correlate with clinical lung injury and disease severity. Unbiased clustering reveals distinct subsets of cis-regulatory elements that delineate the cell type, pathway-specific, and combinatorial transcription factor activity. We find evidence of critical roles of regulatory networks, showing that STAT/BCL6 and E2F/MYB regulatory programs from myeloid cell populations are activated in patients with poor disease outcomes and associated with COVID-19 susceptibility genetic variants. More broadly, we demonstrate how capturing acute, disease-mediated changes in transcription initiation can provide insight into the underlying molecular mechanisms and stratify patient disease severity.


Asunto(s)
COVID-19 , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Regulación de la Expresión Génica , Leucocitos/metabolismo , Unidades de Cuidados Intensivos
4.
Nat Commun ; 14(1): 949, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36810287

RESUMEN

Obesity caused by genetic and environmental factors can lead to compromised skeletal muscle function. Time-restricted feeding (TRF) has been shown to prevent muscle function decline from obesogenic challenges; however, its mechanism remains unclear. Here we demonstrate that TRF upregulates genes involved in glycine production (Sardh and CG5955) and utilization (Gnmt), while Dgat2, involved in triglyceride synthesis is downregulated in Drosophila models of diet- and genetic-induced obesity. Muscle-specific knockdown of Gnmt, Sardh, and CG5955 lead to muscle dysfunction, ectopic lipid accumulation, and loss of TRF-mediated benefits, while knockdown of Dgat2 retains muscle function during aging and reduces ectopic lipid accumulation. Further analyses demonstrate that TRF upregulates the purine cycle in a diet-induced obesity model and AMPK signaling-associated pathways in a genetic-induced obesity model. Overall, our data suggest that TRF improves muscle function through modulations of common and distinct pathways under different obesogenic challenges and provides potential targets for obesity treatments.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Drosophila , Animales , Obesidad/metabolismo , Músculo Esquelético/metabolismo , Lípidos , Purinas , Dieta Alta en Grasa
5.
Obesity (Silver Spring) ; 31 Suppl 1: 63-74, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35912794

RESUMEN

OBJECTIVE: Time-restricted eating (TRE) restores circadian rhythms in mice, but the evidence to support this in humans is limited. The objective of this study was to investigate the effects of TRE on 24-hour profiles of plasma metabolites, glucoregulatory hormones, and the subcutaneous adipose tissue (SAT) transcriptome in humans. METHODS: Men (n = 15, age = 63 [4] years, BMI 30.5 [2.4] kg/m2 ) were recruited. A 35-hour metabolic ward stay was conducted at baseline and after 8 weeks of 10-hour TRE. Assessment included 24-hour profiles of plasma glucose, nonesterified fatty acid (NEFA), triglyceride, glucoregulatory hormones, and the SAT transcriptome. Dim light melatonin onset and cortisol area under the curve were calculated. RESULTS: TRE did not alter dim light melatonin onset but reduced morning cortisol area under the curve. TRE altered 24-hour profiles of insulin, NEFA, triglyceride, and glucose-dependent insulinotropic peptide and increased transcripts of circadian locomotor output cycles protein kaput (CLOCK) and nuclear receptor subfamily 1 group D member 2 (NR1D2) and decreased period circadian regulator 1 (PER1) and nuclear receptor subfamily 1 group D member 1 (NR1D1) at 12:00 am. The rhythmicity of 450 genes was altered by TRE, which enriched in transcripts for transcription corepressor activity, DNA-binding transcription factor binding, regulation of chromatin organization, and small GTPase binding pathways. Weighted gene coexpression network analysis revealed eigengenes that were correlated with BMI, insulin, and NEFA. CONCLUSIONS: TRE restored 24-hour profiles in hormones, metabolites, and genes controlling transcriptional regulation in SAT, which could underpin its metabolic health benefit.


Asunto(s)
Tejido Adiposo , Ritmo Circadiano , Ayuno Intermitente , Obesidad , Humanos , Masculino , Persona de Mediana Edad , Ritmo Circadiano/genética , Ácidos Grasos no Esterificados , Hidrocortisona , Insulinas , Melatonina , Obesidad/genética , Transcriptoma , Anciano
6.
Nutrition ; 96: 111583, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35150947

RESUMEN

OBJECTIVE: We sought to examine the effects of 8 wk of time-restricted eating (TRE) on glucose metabolism and the adipose tissue transcriptome during a metabolic ward stay in men with obesity. METHODS: In a single-arm, pre-post trial, 15 men (ages 63 ± 4 y, body mass index = 30.5 ± 2.4 kg/m2, waist circumference = 113 ± 4 cm) with obesity but no history of diabetes were enrolled and underwent 2 wk of baseline monitoring before they were instructed to eat their regular diets within a contiguous 10-h time frame each day for 8 wk. Metabolic testing was performed at baseline and week 8 during a 35-h metabolic ward stay, during which all food intake was strictly timed and controlled. Identical meal-tolerance tests were performed at breakfast and dinner. Blood glucose, glucoregulatory hormones, and subjective appetite score were measured. Subcutaneous adipose tissue biopsies were performed and the transcriptome was assessed. RESULTS: The primary outcome, plasma glucose area under the curve, was altered by TRE, being unchanged at breakfast but increased at dinner. However, TRE reduced fasting glucose, glycated hemoglobin, body weight, and body fat, and increased glucose-dependent insulinotropic peptide area under the curve at dinner. In subcutaneous adipose tissue, 117 genes were up-regulated and 202 genes down-regulated by TRE. Pathway analysis revealed down-regulation of genes involved in proteasome function and mitochondrial regulation. CONCLUSIONS: TRE had a net effect of reducing glycemia and dampening energy-consuming pathways in adipose tissue.


Asunto(s)
Ayuno , Control Glucémico , Tejido Adiposo/metabolismo , Anciano , Glucemia/metabolismo , Peso Corporal , Ayuno/fisiología , Humanos , Masculino , Persona de Mediana Edad , Obesidad/metabolismo
7.
bioRxiv ; 2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34462742

RESUMEN

The contribution of transcription factors (TFs) and gene regulatory programs in the immune response to COVID-19 and their relationship to disease outcome is not fully understood. Analysis of genome-wide changes in transcription at both promoter-proximal and distal cis-regulatory DNA elements, collectively termed the 'active cistrome,' offers an unbiased assessment of TF activity identifying key pathways regulated in homeostasis or disease. Here, we profiled the active cistrome from peripheral leukocytes of critically ill COVID-19 patients to identify major regulatory programs and their dynamics during SARS-CoV-2 associated acute respiratory distress syndrome (ARDS). We identified TF motifs that track the severity of COVID- 19 lung injury, disease resolution, and outcome. We used unbiased clustering to reveal distinct cistrome subsets delineating the regulation of pathways, cell types, and the combinatorial activity of TFs. We found critical roles for regulatory networks driven by stimulus and lineage determining TFs, showing that STAT and E2F/MYB regulatory programs targeting myeloid cells are activated in patients with poor disease outcomes and associated with single nucleotide genetic variants implicated in COVID-19 susceptibility. Integration with single-cell RNA-seq found that STAT and E2F/MYB activation converged in specific neutrophils subset found in patients with severe disease. Collectively we demonstrate that cistrome analysis facilitates insight into disease mechanisms and provides an unbiased approach to evaluate global changes in transcription factor activity and stratify patient disease severity.

8.
Nat Commun ; 11(1): 2521, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32415079

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Nat Commun ; 10(1): 2700, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221967

RESUMEN

Pathological obesity can result from genetic predisposition, obesogenic diet, and circadian rhythm disruption. Obesity compromises function of muscle, which accounts for a majority of body mass. Behavioral intervention that can counteract obesity arising from genetic, diet or circadian disruption and can improve muscle function holds untapped potential to combat the obesity epidemic. Here we show that Drosophila melanogaster (fruit fly) subject to obesogenic challenges exhibits metabolic disease phenotypes in skeletal muscle; sarcomere disorganization, mitochondrial deformation, upregulation of Phospho-AKT level, aberrant intramuscular lipid infiltration, and insulin resistance. Imposing time-restricted feeding (TRF) paradigm in which flies were fed for 12 h during the day counteracts obesity-induced dysmetabolism and improves muscle performance by suppressing intramuscular fat deposits, Phospho-AKT level, mitochondrial aberrations, and markers of insulin resistance. Importantly, TRF was effective even in an irregular lighting schedule mimicking shiftwork. Hence, TRF is an effective dietary intervention for combating metabolic dysfunction arising from multiple causes.


Asunto(s)
Trastornos Cronobiológicos/dietoterapia , Ayuno/fisiología , Síndrome Metabólico/dietoterapia , Músculo Esquelético/fisiopatología , Obesidad/dietoterapia , Animales , Animales Modificados Genéticamente , Trastornos Cronobiológicos/etiología , Trastornos Cronobiológicos/fisiopatología , Ritmo Circadiano/fisiología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Drosophila melanogaster , Metabolismo Energético/fisiología , Femenino , Humanos , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/patología , Síndrome Metabólico/fisiopatología , Músculo Esquelético/patología , Obesidad/etiología , Obesidad/patología , Obesidad/fisiopatología , Sarcómeros/patología , Horario de Trabajo por Turnos/efectos adversos , Resultado del Tratamiento
10.
Cell Metab ; 29(2): 303-319.e4, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30174302

RESUMEN

Increased susceptibility of circadian clock mutant mice to metabolic diseases has led to the idea that a molecular clock is necessary for metabolic homeostasis. However, these mice often lack a normal feeding-fasting cycle. We tested whether time-restricted feeding (TRF) could prevent obesity and metabolic syndrome in whole-body Cry1;Cry2 and in liver-specific Bmal1 and Rev-erbα/ß knockout mice. When provided access to food ad libitum, these mice rapidly gained weight and showed genotype-specific metabolic defects. However, when fed the same diet under TRF (food access restricted to 10 hr during the dark phase) they were protected from excessive weight gain and metabolic diseases. Transcriptome and metabolome analyses showed that TRF reduced the accumulation of hepatic lipids and enhanced cellular defenses against metabolic stress. These results suggest that the circadian clock maintains metabolic homeostasis by sustaining daily rhythms in feeding and fasting and by maintaining balance between nutrient and cellular stress responses.


Asunto(s)
Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Ayuno/metabolismo , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Animales , Relojes Circadianos/genética , Técnicas de Inactivación de Genes , Ratones , Ratones Noqueados , Aumento de Peso
11.
Science ; 359(6381)2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29439024

RESUMEN

Diurnal gene expression patterns underlie time-of-the-day-specific functional specialization of tissues. However, available circadian gene expression atlases of a few organs are largely from nocturnal vertebrates. We report the diurnal transcriptome of 64 tissues, including 22 brain regions, sampled every 2 hours over 24 hours, from the primate Papio anubis (baboon). Genomic transcription was highly rhythmic, with up to 81.7% of protein-coding genes showing daily rhythms in expression. In addition to tissue-specific gene expression, the rhythmic transcriptome imparts another layer of functional specialization. Most ubiquitously expressed genes that participate in essential cellular functions exhibit rhythmic expression in a tissue-specific manner. The peak phases of rhythmic gene expression clustered around dawn and dusk, with a "quiescent period" during early night. Our findings also unveil a different temporal organization of central and peripheral tissues between diurnal and nocturnal animals.


Asunto(s)
Encéfalo/fisiología , Relojes Circadianos/genética , Ritmo Circadiano/genética , Papio anubis/genética , Papio anubis/fisiología , Transcriptoma , Animales , Encéfalo/metabolismo , Genómica , Masculino
12.
Cell Metab ; 27(2): 404-418.e7, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29358041

RESUMEN

The mechanisms by which feeding and fasting drive rhythmic gene expression for physiological adaptation to daily rhythm in nutrient availability are not well understood. Here we show that, upon feeding, the RNA-binding protein NONO accumulates within speckle-like structures in liver cell nuclei. Combining RNA-immunoprecipitation and sequencing (RIP-seq), we find that an increased number of RNAs are bound by NONO after feeding. We further show that NONO binds and regulates the rhythmicity of genes involved in nutrient metabolism post-transcriptionally. Finally, we show that disrupted rhythmicity of NONO target genes has profound metabolic impact. Indeed, NONO-deficient mice exhibit impaired glucose tolerance and lower hepatic glycogen and lipids. Accordingly, these mice shift from glucose storage to fat oxidation, and therefore remain lean throughout adulthood. In conclusion, our study demonstrates that NONO post-transcriptionally coordinates circadian mRNA expression of metabolic genes with the feeding/fasting cycle, thereby playing a critical role in energy homeostasis.


Asunto(s)
Adaptación Fisiológica , Proteínas de Unión al ADN/metabolismo , Conducta Alimentaria , Hígado/metabolismo , Proteínas de Unión al ARN/metabolismo , Adiposidad/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Hepatocitos/metabolismo , Homeostasis/efectos de los fármacos , Intrones/genética , Ratones Endogámicos C57BL , Modelos Biológicos , Unión Proteica , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
Science ; 347(6227): 1265-9, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25766238

RESUMEN

Circadian clocks orchestrate periods of rest or activity and feeding or fasting over the course of a 24-hour day and maintain homeostasis. To assess whether a consolidated 24-hour cycle of feeding and fasting can sustain health, we explored the effect of time-restricted feeding (TRF; food access limited to daytime 12 hours every day) on neural, peripheral, and cardiovascular physiology in Drosophila melanogaster. We detected improved sleep, prevention of body weight gain, and deceleration of cardiac aging under TRF, even when caloric intake and activity were unchanged. We used temporal gene expression profiling and validation through classical genetics to identify the TCP-1 ring complex (TRiC) chaperonin, the mitochondrial electron transport chain complexes, and the circadian clock as pathways mediating the benefits of TRF.


Asunto(s)
Relojes Circadianos , Drosophila melanogaster/fisiología , Animales , Chaperoninas/genética , Chaperoninas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Transporte de Electrón , Ingestión de Energía , Conducta Alimentaria , Vuelo Animal , Corazón/fisiología , Masculino , Redes y Vías Metabólicas , Mitocondrias/metabolismo , Contracción Miocárdica , Sueño , Transcriptoma , Aumento de Peso
14.
Science ; 333(6051): 1881-5, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21960634

RESUMEN

In animals, circadian oscillators are based on a transcription-translation circuit that revolves around the transcription factors CLOCK and BMAL1. We found that the JumonjiC (JmjC) and ARID domain-containing histone lysine demethylase 1a (JARID1a) formed a complex with CLOCK-BMAL1, which was recruited to the Per2 promoter. JARID1a increased histone acetylation by inhibiting histone deacetylase 1 function and enhanced transcription by CLOCK-BMAL1 in a demethylase-independent manner. Depletion of JARID1a in mammalian cells reduced Per promoter histone acetylation, dampened expression of canonical circadian genes, and shortened the period of circadian rhythms. Drosophila lines with reduced expression of the Jarid1a homolog, lid, had lowered Per expression and similarly altered circadian rhythms. JARID1a thus has a nonredundant role in circadian oscillator function.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Proteínas CLOCK/metabolismo , Relojes Circadianos , Proteína 2 de Unión a Retinoblastoma/metabolismo , Acetilación , Animales , Proteínas de Unión al ADN , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Histona Demetilasas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji , Masculino , Ratones , Ratones Noqueados , Proteínas Circadianas Period/genética , Regiones Promotoras Genéticas , Transcripción Genética , Transfección
15.
Proc Natl Acad Sci U S A ; 106(50): 21453-8, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19940241

RESUMEN

In mammals, the circadian oscillator generates approximately 24-h rhythms in feeding behavior, even under constant environmental conditions. Livers of mice held under constant darkness exhibit circadian rhythm in abundance in up to 15% of expressed transcripts. Therefore, oscillations in hepatic transcripts could be driven by rhythmic food intake or sustained by the hepatic circadian oscillator, or a combination of both. To address this question, we used distinct feeding and fasting paradigms on wild-type (WT) and circadian clock-deficient mice. We monitored temporal patterns of feeding and hepatic transcription. Both food availability and the temporal pattern of feeding determined the repertoire, phase, and amplitude of the circadian transcriptome in WT liver. In the absence of feeding, only a small subset of transcripts continued to express circadian patterns. Conversely, temporally restricted feeding restored rhythmic transcription of hundreds of genes in oscillator-deficient mouse liver. Our findings show that both temporal pattern of food intake and the circadian clock drive rhythmic transcription, thereby highlighting temporal regulation of hepatic transcription as an emergent property of the circadian system.


Asunto(s)
Ritmo Circadiano/genética , Ingestión de Alimentos/genética , Regulación de la Expresión Génica , Hígado/metabolismo , Animales , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis , Factores de Tiempo
16.
Chromosoma ; 112(6): 269-76, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14767778

RESUMEN

Suppressors and enhancers of position effect variegation (PEV) have been linked to the establishment and maintenance of heterochromatin. The presence of centromeres and other inheritance elements in heterochromatic regions suggests that suppressors and enhancers of PEV, Su(var) s and E(var)s [collectively termed Mod(var)s], may be required for chromosome inheritance. In order to test this hypothesis, we screened 59 ethyl methanesulfonate-generated Drosophila Mod(var)s for dominant effects on the partially compromised inheritance of a minichromosome ( J21A) missing a portion of the genetically defined centromere. Nearly half of these Mod(var)s significantly increased or decreased the transmission of J21A. Analyses of homozygous mutant larval neuroblasts suggest that these mutations affect cell cycle progression and native chromosome morphology. Five out of six complementation groups tested displayed mitotic abnormalities, including phenotypes such as telomere fusions, overcondensed chromosomes, and low mitotic index. We conclude that Mod(var)s as a group are highly enriched for genes that encode essential inheritance functions. We propose that a primary function of Mod(var)s is to promote chromosome inheritance, and that the gene silencing phenotype associated with PEV may be a secondary consequence of the heterochromatic structures required to carry out these functions.


Asunto(s)
Drosophila melanogaster/genética , Genes Dominantes/fisiología , Genes de Insecto , Heterocromatina/genética , Neuronas/citología , Animales , Ciclo Celular , Centrómero , Transmisión de Enfermedad Infecciosa , Metanosulfonato de Etilo/toxicidad , Femenino , Silenciador del Gen , Prueba de Complementación Genética , Homocigoto , Larva/metabolismo , Masculino , Índice Mitótico , Mutación , Neuronas/metabolismo , Fenotipo , Supresión Genética , Telómero/metabolismo
17.
Genome Res ; 13(2): 182-94, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12566396

RESUMEN

Centromeres are the site for kinetochore formation and spindle attachment and are embedded in heterochromatin in most eukaryotes. The repeat-rich nature of heterochromatin has hindered obtaining a detailed understanding of the composition and organization of heterochromatic and centromeric DNA sequences. Here, we report the results of extensive sequence analysis of a fully functional centromere present in the Drosophila Dp1187 minichromosome. Approximately 8.4% (31 kb) of the highly repeated satellite DNA (AATAT and TTCTC) was sequenced, representing the largest data set of Drosophila satellite DNA sequence to date. Sequence analysis revealed that the orientation of the arrays is uniform and that individual repeats within the arrays mostly differ by rare, single-base polymorphisms. The entire complex DNA component of this centromere (69.7 kb) was sequenced and assembled. The 39-kb "complex island" Maupiti contains long stretches of a complex A+T rich repeat interspersed with transposon fragments, and most of these elements are organized as direct repeats. Surprisingly, five single, intact transposons are directly inserted at different locations in the AATAT satellite arrays. We find no evidence for centromere-specific sequences within this centromere, providing further evidence for sequence-independent, epigenetic determination of centromere identity and function in higher eukaryotes. Our results also demonstrate that the sequence composition and organization of large regions of centric heterochromatin can be determined, despite the presence of repeated DNA.


Asunto(s)
Centrómero/genética , Drosophila melanogaster/genética , Análisis de Secuencia de ADN/métodos , Animales , Composición de Base/genética , Cromosomas/genética , Clonación Molecular/métodos , Secuencia Conservada/genética , Mapeo Contig/métodos , Elementos Transponibles de ADN/genética , ADN Satélite/genética , Genes de Insecto/genética , Repeticiones de Minisatélite/genética , Datos de Secuencia Molecular , Mutagénesis Insercional/genética , Reacción en Cadena de la Polimerasa/métodos
18.
Genetics ; 161(1): 217-29, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12019236

RESUMEN

Approximately one-third of the human and Drosophila melanogaster genomes are heterochromatic, yet we know very little about the structure and function of this enigmatic component of eukaryotic genomes. To facilitate molecular and cytological analysis of heterochromatin we introduced a yellow(+) (y(+))-marked P element into centric heterochromatin by screening for variegated phenotypes, that is, mosaic gene inactivation. We recovered >110 P insertions with variegated yellow expression from approximately 3500 total mobilization events. FISH analysis of 71 of these insertions showed that 69 (97%) were in the centric heterochromatin, rather than telomeres or euchromatin. High-resolution banding analysis showed a wide but nonuniform distribution of insertions within centric heterochromatin; variegated insertions were predominantly recovered near regions of satellite DNA. We successfully used inverse PCR to clone and sequence the flanking DNA for approximately 63% of the insertions. BLAST analysis of the flanks demonstrated that either most of the variegated insertions could not be placed on the genomic scaffold, and thus may be inserted within novel DNA sequence, or that the flanking DNA hit multiple sites on the scaffold, due to insertions within different transposons. Taken together these data suggest that screening for yellow variegation is a very efficient method for recovering centric insertions and that a large-scale screen for variegated yellow P insertions will provide important tools for detailed analysis of centric heterochromatin structure and function.


Asunto(s)
Elementos Transponibles de ADN , Proteínas de Drosophila , Drosophila melanogaster/genética , Heterocromatina , Animales , ADN/aislamiento & purificación , Femenino , Hibridación Fluorescente in Situ , Proteínas de Insectos/genética , Masculino , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...