Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
EMBO J ; 41(2): e110157, 2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34806788

RESUMEN

Organoids are self-organizing in vitro 3D cultures that are histologically similar to a variety of human organs. A recent study by Rosselot et al (2021) shows that mature intestinal organoids possess species-specific circadian clocks similar to their respective in vivo context, suggesting organoids as promising platforms to study circadian medicine.


Asunto(s)
Relojes Circadianos , Garrapatas , Animales , Relojes Circadianos/genética , Humanos , Intestinos , Organoides
2.
Cell ; 144(2): 268-81, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21236481

RESUMEN

Direct evidence for the requirement of delay in feedback repression in the mammalian circadian clock has been elusive. Cryptochrome 1 (Cry1), an essential clock component, displays evening-time expression and serves as a strong repressor at morning-time elements (E box/E' box). In this study, we reveal that a combination of day-time elements (D box) within the Cry1-proximal promoter and night-time elements (RREs) within its intronic enhancer gives rise to evening-time expression. A synthetic composite promoter produced evening-time expression, which was further recapitulated by a simple phase-vector model. Of note, coordination of day-time with night-time elements can modulate the extent of phase delay. A genetic complementation assay in Cry1(-/-):Cry2(-/-) cells revealed that substantial delay of Cry1 expression is required to restore circadian rhythmicity, and its prolonged delay slows circadian oscillation. Taken together, our data suggest that phase delay in Cry1 transcription is required for mammalian clock function.


Asunto(s)
Relojes Circadianos , Criptocromos/metabolismo , Retroalimentación , Animales , Ritmo Circadiano , Elementos de Facilitación Genéticos , Intrones , Ratones , Regiones Promotoras Genéticas , Elementos Reguladores de la Transcripción , Análisis de la Célula Individual
3.
Proc Natl Acad Sci U S A ; 120(40): e2214636120, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37769257

RESUMEN

Many mammalian proteins have circadian cycles of production and degradation, and many of these rhythms are altered posttranscriptionally. We used ribosome profiling to examine posttranscriptional control of circadian rhythms by quantifying RNA translation in the liver over a 24-h period from circadian-entrained mice transferred to constant darkness conditions and by comparing ribosome binding levels to protein levels for 16 circadian proteins. We observed large differences in ribosome binding levels compared to protein levels, and we observed delays between peak ribosome binding and peak protein abundance. We found extensive binding of ribosomes to upstream open reading frames (uORFs) in circadian mRNAs, including the core clock gene Period2 (Per2). An increase in the number of uORFs in the 5'UTR was associated with a decrease in ribosome binding in the main coding sequence and a reduction in expression of synthetic reporter constructs. Mutation of the Per2 uORF increased luciferase and fluorescence reporter expression in 3T3 cells and increased luciferase expression in PER2:LUC MEF cells. Mutation of the Per2 uORF in mice increased Per2 mRNA expression, enhanced ribosome binding on Per2, and reduced total sleep time compared to that in wild-type mice. These results suggest that uORFs affect mRNA posttranscriptionally, which can impact physiological rhythms and sleep.


Asunto(s)
Ritmo Circadiano , Perfilado de Ribosomas , Sueño , Animales , Ratones , Ritmo Circadiano/genética , Luciferasas/genética , Sistemas de Lectura Abierta/genética , ARN Mensajero/genética , Sueño/genética , Proteínas Circadianas Period/genética
4.
PLoS Biol ; 20(10): e3001813, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36194579

RESUMEN

The reduced sleep duration previously observed in Camk2b knockout mice revealed a role for Ca2+/calmodulin-dependent protein kinase II (CaMKII)ß as a sleep-promoting kinase. However, the underlying mechanism by which CaMKIIß supports sleep regulation is largely unknown. Here, we demonstrate that activation or inhibition of CaMKIIß can increase or decrease sleep duration in mice by almost 2-fold, supporting the role of CaMKIIß as a core sleep regulator in mammals. Importantly, we show that this sleep regulation depends on the kinase activity of CaMKIIß. A CaMKIIß mutant mimicking the constitutive-active (auto)phosphorylation state promotes the transition from awake state to sleep state, while mutants mimicking subsequent multisite (auto)phosphorylation states suppress the transition from sleep state to awake state. These results suggest that the phosphorylation states of CaMKIIß differently control sleep induction and maintenance processes, leading us to propose a "phosphorylation hypothesis of sleep" for the molecular control of sleep in mammals.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Calcio , Animales , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Mamíferos/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Sueño
5.
Mol Cell ; 67(5): 783-798.e20, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886336

RESUMEN

Temperature compensation is a striking feature of the circadian clock. Here we investigate biochemical mechanisms underlying temperature-compensated, CKIδ-dependent multi-site phosphorylation in mammals. We identify two mechanisms for temperature-insensitive phosphorylation at higher temperature: lower substrate affinity to CKIδ-ATP complex and higher product affinity to CKIδ-ADP complex. Inhibitor screening of ADP-dependent phosphatase activity of CKIδ identified aurintricarboxylic acid (ATA) as a temperature-sensitive kinase activator. Docking simulation of ATA and mutagenesis experiment revealed K224D/K224E mutations in CKIδ that impaired product binding and temperature-compensated primed phosphorylation. Importantly, K224D mutation shortens behavioral circadian rhythms and changes the temperature dependency of SCN's circadian period. Interestingly, temperature-compensated phosphorylation was evolutionary conserved in yeast. Molecular dynamics simulation and X-ray crystallography demonstrate that an evolutionally conserved CKI-specific domain around K224 can provide a structural basis for temperature-sensitive substrate and product binding. Surprisingly, this domain can confer temperature compensation on a temperature-sensitive TTBK1. These findings suggest the temperature-sensitive substrate- and product-binding mechanisms underlie temperature compensation.


Asunto(s)
Adenosina Trifosfato/metabolismo , Quinasa Idelta de la Caseína/metabolismo , Relojes Circadianos , Ritmo Circadiano , Núcleo Supraquiasmático/enzimología , Temperatura , Animales , Sitios de Unión , Quinasa Idelta de la Caseína/química , Quinasa Idelta de la Caseína/genética , Dominio Catalítico , Cristalografía por Rayos X , Genotipo , Células HEK293 , Humanos , Hidrólisis , Cinética , Locomoción , Ratones Transgénicos , Modelos Biológicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutación , Fenotipo , Fosforilación , Unión Proteica , Dominios Proteicos , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Serina , Relación Estructura-Actividad , Especificidad por Sustrato , Técnicas de Cultivo de Tejidos , Transfección
6.
Proc Natl Acad Sci U S A ; 114(20): 5069-5071, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28487482
7.
Nat Genet ; 38(3): 312-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16474406

RESUMEN

Direct evidence for the requirement of transcriptional feedback repression in circadian clock function has been elusive. Here, we developed a molecular genetic screen in mammalian cells to identify mutants of the circadian transcriptional activators CLOCK and BMAL1, which were uncoupled from CRYPTOCHROME (CRY)-mediated transcriptional repression. Notably, mutations in the PER-ARNT-SIM domain of CLOCK and the C terminus of BMAL1 resulted in synergistic insensitivity through reduced physical interactions with CRY. Coexpression of these mutant proteins in cultured fibroblasts caused arrhythmic phenotypes in population and single-cell assays. These data demonstrate that CRY-mediated repression of the CLOCK/BMAL1 complex activity is required for maintenance of circadian rhythmicity and provide formal proof that transcriptional feedback is required for mammalian clock function.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ritmo Circadiano/fisiología , Regulación de la Expresión Génica , Transactivadores/genética , Células 3T3 , Factores de Transcripción ARNTL , Animales , Proteínas CLOCK , Línea Celular , Retroalimentación , Genes Reporteros , Humanos , Luciferasas/análisis , Luciferasas/genética , Luminiscencia , Ratones , Plásmidos , Tiempo
8.
Proc Natl Acad Sci U S A ; 106(37): 15744-9, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19805222

RESUMEN

A striking feature of the circadian clock is its flexible yet robust response to various environmental conditions. To analyze the biochemical processes underlying this flexible-yet-robust characteristic, we examined the effects of 1,260 pharmacologically active compounds in mouse and human clock cell lines. Compounds that markedly (>10 s.d.) lengthened the period in both cell lines, also lengthened it in central clock tissues and peripheral clock cells. Most compounds inhibited casein kinase Iepsilon (CKIepsilon) or CKIdelta phosphorylation of the PER2 protein. Manipulation of CKIepsilon/delta-dependent phosphorylation by these compounds lengthened the period of the mammalian clock from circadian (24 h) to circabidian (48 h), revealing its high sensitivity to chemical perturbation. The degradation rate of PER2, which is regulated by CKIepsilon/delta-dependent phosphorylation, was temperature-insensitive in living clock cells, yet sensitive to chemical perturbations. This temperature-insensitivity was preserved in the CKIepsilon/delta-dependent phosphorylation of a synthetic peptide in vitro. Thus, CKIepsilon/delta-dependent phosphorylation is likely a temperature-insensitive period-determining process in the mammalian circadian clock.


Asunto(s)
Caseína Cinasa 1 épsilon/metabolismo , Quinasa Idelta de la Caseína/metabolismo , Ritmo Circadiano/fisiología , Animales , Evolución Biológica , Caseína Cinasa 1 épsilon/antagonistas & inhibidores , Caseína Cinasa 1 épsilon/genética , Quinasa Idelta de la Caseína/antagonistas & inhibidores , Quinasa Idelta de la Caseína/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/genética , Cianobacterias/genética , Cianobacterias/fisiología , Humanos , Cinética , Ratones , Modelos Biológicos , Células 3T3 NIH , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Circadianas Period , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Temperatura , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
9.
Cell Rep Methods ; 1(2): 100038, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35475238

RESUMEN

Recent advancements in tissue clearing technologies have offered unparalleled opportunities for researchers to explore the whole mouse brain at cellular resolution. With the expansion of this experimental technique, however, a scalable and easy-to-use computational tool is in demand to effectively analyze and integrate whole-brain mapping datasets. To that end, here we present CUBIC-Cloud, a cloud-based framework to quantify, visualize, and integrate mouse brain data. CUBIC-Cloud is a fully automated system where users can upload their whole-brain data, run analyses, and publish the results. We demonstrate the generality of CUBIC-Cloud by a variety of applications. First, we investigated the brain-wide distribution of five cell types. Second, we quantified Aß plaque deposition in Alzheimer's disease model mouse brains. Third, we reconstructed a neuronal activity profile under LPS-induced inflammation by c-Fos immunostaining. Last, we show brain-wide connectivity mapping by pseudotyped rabies virus. Together, CUBIC-Cloud provides an integrative platform to advance scalable and collaborative whole-brain mapping.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Ratones , Animales , Encéfalo/diagnóstico por imagen , Mapeo Encefálico , Enfermedad de Alzheimer/diagnóstico por imagen , Neuronas
10.
Front Neurosci ; 13: 1402, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32009883

RESUMEN

Rapid-eye movement (REM) sleep is a paradoxical sleep state characterized by brain activity similar to wakefulness, rapid-eye-movement, and lack of muscle tone. REM sleep is a fundamental brain function, evolutionary conserved across species, including human, mouse, bird, and even reptiles. The physiological importance of REM sleep is highlighted by severe sleep disorders incurred by a failure in REM sleep regulation. Despite the intense interest in the mechanism of REM sleep regulation, the molecular machinery is largely left to be investigated. In models of REM sleep regulation, acetylcholine has been a pivotal component. However, even newly emerged techniques such as pharmacogenetics and optogenetics have not fully clarified the function of acetylcholine either at the cellular level or neural-circuit level. Recently, we discovered that the G q type muscarinic acetylcholine receptor genes, Chrm1 and Chrm3, are essential for REM sleep. In this review, we develop the perspective of current knowledge on REM sleep from a molecular viewpoint. This should be a starting point to clarify the molecular and cellular machinery underlying REM sleep regulation and will provide insights to explore physiological functions of REM sleep and its pathological roles in REM-sleep-related disorders such as depression, PTSD, and neurodegenerative diseases.

11.
Anal Sci ; 35(10): 1141-1147, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31597873

RESUMEN

Microfluidic devices are important platforms to culture and observe biological tissues. Compared with conventional setups, microfluidic devices have advantages in perfusion, including an enhanced delivery of nutrients and gases to tissues. However, explanted tissues can maintain their functions for only hours to days in microfluidic devices, although their observations are desired for weeks. The suprachiasmatic nucleus (SCN) is a brain region composed of heterogeneous cells to control the biological clock system through synchronizing individual cells in this region. The synchronized and complicated cell-cell interactions of SCN cells are difficult to reproduce from seeded cells. Thus, the viability of explanted SCN contributes to the study of SCN functions. In this paper, we propose a new perfusion platform combining a PDMS microfluidic device with a porous membrane to culture an explanted SCN for 25 days. We expect that this platform will provide a universal interface for microfluidic manipulation of tissue explants.


Asunto(s)
Gases/metabolismo , Dispositivos Laboratorio en un Chip , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/metabolismo , Técnicas de Cultivo de Tejidos/instrumentación , Animales , Ratones , Supervivencia Tisular
12.
Nucleic Acids Res ; 34(5): e42, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16547197

RESUMEN

A systems-level understanding of a small but essential population of cells in development or adulthood (e.g. somatic stem cells) requires accurate quantitative monitoring of genome-wide gene expression, ideally from single cells. We report here a strategy to globally amplify mRNAs from single cells for highly quantitative high-density oligonucleotide microarray analysis that combines a small number of directional PCR cycles with subsequent linear amplification. Using this strategy, both the representation of gene expression profiles and reproducibility between individual experiments are unambiguously improved from the original method, along with high coverage and accuracy. The immediate application of this method to single cells in the undifferentiated inner cell masses of mouse blastocysts at embryonic day (E) 3.5 revealed the presence of two populations of cells, one with primitive endoderm (PE) expression and the other with pluripotent epiblast-like gene expression. The genes expressed differentially between these two populations were well preserved in morphologically differentiated PE and epiblast in the embryos one day later (E4.5), demonstrating that the method successfully detects subtle but essential differences in gene expression at the single-cell level among seemingly homogeneous cell populations. This study provides a strategy to analyze biophysical events in medicine as well as in neural, stem cell and developmental biology, where small numbers of distinctive or diseased cells play critical roles.


Asunto(s)
ADN Complementario , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/análisis , Animales , Secuencia de Bases , Blastocisto/metabolismo , ADN Complementario/química , Ratones , ARN Mensajero/metabolismo , Alineación de Secuencia
13.
Cell Rep ; 24(9): 2231-2247.e7, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30157420

RESUMEN

Sleep regulation involves interdependent signaling among specialized neurons in distributed brain regions. Although acetylcholine promotes wakefulness and rapid eye movement (REM) sleep, it is unclear whether the cholinergic pathway is essential (i.e., absolutely required) for REM sleep because of redundancy from neural circuits to molecules. First, we demonstrate that synaptic inhibition of TrkA+ cholinergic neurons causes a severe short-sleep phenotype and that sleep reduction is mostly attributable to a shortened sleep duration in the dark phase. Subsequent comprehensive knockout of acetylcholine receptor genes by the triple-target CRISPR method reveals that a similar short-sleep phenotype appears in the knockout of two Gq-type acetylcholine receptors Chrm1 and Chrm3. Strikingly, Chrm1 and Chrm3 double knockout chronically diminishes REM sleep to an almost undetectable level. These results suggest that muscarinic acetylcholine receptors, Chrm1 and Chrm3, are essential for REM sleep.


Asunto(s)
Acetilcolina/metabolismo , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/metabolismo , Sueño REM/genética , Animales , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
14.
J Biochem ; 154(4): 373-81, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23940085

RESUMEN

We screened circadian-regulated genes in rat cartilage by using a DNA microarray analysis. In rib growth-plate cartilage, numerous genes showed statistically significant circadian mRNA expression under both 12:12 h light-dark and constant darkness conditions. Type II collagen and aggrecan genes--along with several genes essential for post-translational modifications of collagen and aggrecan, including prolyl 4-hydroxylase 1, lysyl oxidase, lysyl oxidase-like 2 and 3'-phosphoadenosine 5'-phosphosulphate synthase 2--showed the same circadian phase. In addition, the mRNA level of SOX9, a master transcription factor for the synthesis of type II collagen and aggrecan, has a similar phase of circadian rhythms. The circadian expression of the matrix-related genes may be critical in the development and the growth of various cartilages, because similar circadian expression of the matrix-related genes was observed in hip joint cartilage. However, the circadian phase of the major matrix-related genes in the rib permanent cartilage was almost the converse of that in the rib growth-plate cartilage under light-dark conditions. We also found that half of the oscillating genes had conserved clock-regulatory elements, indicating contribution of the elements to the clock outputs. These findings suggest that the synthesis of the cartilage matrix macromolecules is controlled by cell-autonomous clocks depending upon the in vivo location of cartilage.


Asunto(s)
Cartílago/metabolismo , Relojes Circadianos , Proteínas Matrilinas/metabolismo , Fotoperiodo , Animales , Expresión Génica , Humanos , Masculino , Proteínas Matrilinas/genética , Ratas , Ratas Sprague-Dawley
15.
PLoS One ; 4(1): e4301, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19173005

RESUMEN

A new circadian variant was isolated by screening the intercross offspring of wild-caught mice (Mus musculus castaneus). This variant was characterized by an initial maintenance of damped oscillations and subsequent loss of rhythmicity after being transferred from light-dark (LD) cycles to constant darkness (DD). To map the genes responsible for the persistence of rhythmicity (circadian ratio) and the length of free-running period (tau), quantitative trait locus (QTL) analysis was performed using F(2) mice obtained from an F(1) cross between the circadian variant and C57BL/6J mice. As a result, a significant QTL with a main effect for circadian ratio (Arrhythmicity; Arrh-1) was mapped on Chromosome (Chr) 8. For tau, four significant QTLs, Short free-running period (Sfp-1) (Chr 1), Sfp-2 (Chr 6), Sfp-3 (Chr 8), Sfp-4 (Chr 11) were determined. An epistatic interaction was detected between Chr 3 (Arrh-2) and Chr 5 (Arrh-3). An in situ hybridization study of clock genes and mouse Period1::luciferase (mPer1::luc) real-time monitoring analysis in the suprachiasmatic nucleus (SCN) suggested that arrhythmicity in this variant might not be attributed to core circadian mechanisms in the SCN neurons. Our strategy using wild-derived variant mice may provide a novel opportunity to evaluate circadian and its related disorders in human that arise from the interaction between multiple variant genes.


Asunto(s)
Animales Salvajes/genética , Animales Salvajes/fisiología , Conducta Animal/fisiología , Ritmo Circadiano/genética , Sustitución de Aminoácidos , Animales , Proteínas CLOCK , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromosomas de los Mamíferos/genética , Ritmo Circadiano/fisiología , Cruzamientos Genéticos , Oscuridad , Epistasis Genética , Femenino , Regulación de la Expresión Génica , Escala de Lod , Mediciones Luminiscentes , Masculino , Ratones , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Circadianas Period , Sitios de Carácter Cuantitativo/genética , Análisis de Secuencia de ADN , Núcleo Supraquiasmático/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
16.
Genes Dev ; 21(13): 1687-700, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17578908

RESUMEN

The Drosophila circadian clock consists of integrated autoregulatory feedback loops, making the clock difficult to elucidate without comprehensively identifying the network components in vivo. Previous studies have adopted genome-wide screening for clock-controlled genes using high-density oligonucleotide arrays that identified hundreds of clock-controlled genes. In an attempt to identify the core clock genes among these candidates, we applied genome-wide functional screening using an RNA interference (RNAi) system in vivo. Here we report the identification of novel clock gene candidates including clockwork orange (cwo), a transcriptional repressor belonging to the basic helix-loop-helix ORANGE family. cwo is rhythmically expressed and directly regulated by CLK-CYC through canonical E-box sequences. A genome-wide search for its target genes using the Drosophila genome tiling array revealed that cwo forms its own negative feedback loop and directly suppresses the expression of other clock genes through the E-box sequence. Furthermore, this negative transcriptional feedback loop contributes to sustaining a high-amplitude circadian oscillation in vivo. Based on these results, we propose that the competition between cyclic CLK-CYC activity and the adjustable threshold imposed by CWO keeps E-box-mediated transcription within the controllable range of its activity, thereby rendering a Drosophila circadian clock capable of generating high-amplitude oscillation.


Asunto(s)
Relojes Biológicos/genética , Ritmo Circadiano/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Genómica , Proteínas Represoras/fisiología , Transcripción Genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas CLOCK , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Elementos E-Box , Regulación de la Expresión Génica , Genoma de los Insectos , Modelos Biológicos , Datos de Secuencia Molecular , Neuronas/metabolismo , Proteínas Represoras/genética , Homología de Secuencia de Aminoácido , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA