Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39131317

RESUMEN

Casein kinase 1 δ (CK1δ) controls essential biological processes including circadian rhythms and Wnt signaling, but how its activity is regulated is not well understood. CK1δ is inhibited by autophosphorylation of its intrinsically disordered C-terminal tail. Two CK1 splice variants, δ 1 and δ 2 , are known to have very different effects on circadian rhythms. These variants differ only in the last 16 residues of the tail, referred to as the extreme C-termini (XCT), but with marked changes in potential phosphorylation sites. Here we test if the XCT of these variants have different effects in autoinhibition of the kinase. Using NMR and HDX-MS, we show that the δ 1 XCT is preferentially phosphorylated by the kinase and the δ 1 tail makes more extensive interactions across the kinase domain. Mutation of δ1 -specific XCT phosphorylation sites increases kinase activity both in vitro and in cells and leads to changes in circadian period, similar to what is reported in vivo. Mechanistically, loss of the phosphorylation sites in XCT disrupts tail interaction with the kinase domain. δ1 autoinhibition relies on conserved anion binding sites around the CK1 active site, demonstrating a common mode of product inhibition of CK1δ . These findings demonstrate how a phosphorylation cycle controls the activity of this essential kinase.

3.
Nat Cancer ; 5(4): 546-556, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38654103

RESUMEN

The circadian clock regulates daily rhythms of numerous physiological activities through tightly coordinated modulation of gene expression and biochemical functions. Circadian disruption is associated with enhanced tumor formation and metastasis via dysregulation of key biological processes and modulation of cancer stem cells (CSCs) and their specialized microenvironment. Here, we review how the circadian clock influences CSCs and their local tumor niches in the context of different stages of tumor metastasis. Identifying circadian therapeutic targets could facilitate the development of new treatments that leverage circadian modulation to ablate tumor-resident CSCs, inhibit tumor metastasis and enhance response to current therapies.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Metástasis de la Neoplasia , Neoplasias , Células Madre Neoplásicas , Microambiente Tumoral , Humanos , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/metabolismo , Ritmo Circadiano/fisiología , Relojes Circadianos/fisiología , Animales , Neoplasias/patología , Regulación Neoplásica de la Expresión Génica
4.
Mol Cell ; 83(10): 1677-1692.e8, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37207626

RESUMEN

PERIOD (PER) and Casein Kinase 1δ regulate circadian rhythms through a phosphoswitch that controls PER stability and repressive activity in the molecular clock. CK1δ phosphorylation of the familial advanced sleep phase (FASP) serine cluster embedded within the Casein Kinase 1 binding domain (CK1BD) of mammalian PER1/2 inhibits its activity on phosphodegrons to stabilize PER and extend circadian period. Here, we show that the phosphorylated FASP region (pFASP) of PER2 directly interacts with and inhibits CK1δ. Co-crystal structures in conjunction with molecular dynamics simulations reveal how pFASP phosphoserines dock into conserved anion binding sites near the active site of CK1δ. Limiting phosphorylation of the FASP serine cluster reduces product inhibition, decreasing PER2 stability and shortening circadian period in human cells. We found that Drosophila PER also regulates CK1δ via feedback inhibition through the phosphorylated PER-Short domain, revealing a conserved mechanism by which PER phosphorylation near the CK1BD regulates CK1 kinase activity.


Asunto(s)
Relojes Circadianos , Proteínas Circadianas Period , Animales , Humanos , Fosforilación , Retroalimentación , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Quinasa de la Caseína I/genética , Quinasa de la Caseína I/metabolismo , Ritmo Circadiano/genética , Drosophila/metabolismo , Serina/metabolismo , Mamíferos/metabolismo
5.
Mol Cell ; 81(6): 1133-1146, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33545069

RESUMEN

In our 24/7 well-lit world, it's easy to skip or delay sleep to work, study, and play. However, our circadian rhythms are not easily fooled; the consequences of jet lag and shift work are many and severe, including metabolic, mood, and malignant disorders. The internal clock that keeps track of time has at its heart the reversible phosphorylation of the PERIOD proteins, regulated by isoforms of casein kinase 1 (CK1). In-depth biochemical, genetic, and structural studies of these kinases, their mutants, and their splice variants have combined over the past several years to provide a robust understanding of how the core clock is regulated by a phosphoswitch whereby phosphorylation of a stabilizing site on PER blocks phosphorylation of a distant phosphodegron. The recent structure of a circadian mutant form of CK1 implicates an internal activation loop switch that regulates this phosphoswitch and points to new approaches to regulation of the clock.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Proteínas Circadianas Period/metabolismo , Animales , Quinasa de la Caseína I/genética , Humanos , Proteínas Circadianas Period/genética , Fosforilación/fisiología
6.
Proc Natl Acad Sci U S A ; 117(20): 10888-10896, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32354999

RESUMEN

Casein kinase 1 (CK1) plays a central role in regulating the period of the circadian clock. In mammals, PER2 protein abundance is regulated by CK1-mediated phosphorylation and proteasomal degradation. On the other hand, recent studies have questioned whether the degradation of the core circadian machinery is a critical step in clock regulation. Prior cell-based studies found that CK1 phosphorylation of PER2 at Ser478 recruits the ubiquitin E3 ligase ß-TrCP, leading to PER2 degradation. Creation of this phosphodegron is regulated by a phosphoswitch that is also implicated in temperature compensation. However, in vivo evidence that this phosphodegron influences circadian period is lacking. Here, we generated and analyzed PER2-Ser478Ala knock-in mice. The mice showed longer circadian period in behavioral analysis. Molecularly, mutant PER2 protein accumulated in both the nucleus and cytoplasm of the mouse liver, while Per2 messenger RNA (mRNA) levels were minimally affected. Nuclear PER1, CRY1, and CRY2 proteins also increased, probably due to stabilization of PER2-containing complexes. In mouse embryonic fibroblasts derived from PER2-Ser478Ala::LUC mice, three-phase decay and temperature compensation of the circadian period was perturbed. These data provide direct in vivo evidence for the importance of phosphorylation-regulated PER2 stability in the circadian clock and validate the phosphoswitch in a mouse model.


Asunto(s)
Relojes Circadianos/fisiología , Mutación , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Animales , Conducta Animal , Quinasa de la Caseína I/metabolismo , Núcleo Celular/metabolismo , Ritmo Circadiano/fisiología , Femenino , Regulación de la Expresión Génica , Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Fosforilación , ARN Mensajero/metabolismo , Transcriptoma , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo
7.
Elife ; 92020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32043967

RESUMEN

Post-translational control of PERIOD stability by Casein Kinase 1δ and ε (CK1) plays a key regulatory role in metazoan circadian rhythms. Despite the deep evolutionary conservation of CK1 in eukaryotes, little is known about its regulation and the factors that influence substrate selectivity on functionally antagonistic sites in PERIOD that directly control circadian period. Here we describe a molecular switch involving a highly conserved anion binding site in CK1. This switch controls conformation of the kinase activation loop and determines which sites on mammalian PER2 are preferentially phosphorylated, thereby directly regulating PER2 stability. Integrated experimental and computational studies shed light on the allosteric linkage between two anion binding sites that dynamically regulate kinase activity. We show that period-altering kinase mutations from humans to Drosophila differentially modulate this activation loop switch to elicit predictable changes in PER2 stability, providing a foundation to understand and further manipulate CK1 regulation of circadian rhythms.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Ritmo Circadiano , Proteínas Circadianas Period/metabolismo , Regulación Alostérica , Animales , Quinasa de la Caseína I/genética , Drosophila , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Fosforilación
8.
Proc Natl Acad Sci U S A ; 115(23): 5980-5985, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29784786

RESUMEN

The N6-methylation of internal adenosines (m6A) in mRNA has been quantified and localized throughout the transcriptome. However, the physiological significance of m6A in most highly methylated mRNAs is unknown. It was demonstrated previously that the circadian clock, based on transcription-translation negative feedback loops, is sensitive to the general inhibition of m6A. Here, we show that the Casein Kinase 1 Delta mRNA (Ck1δ), coding for a critical kinase in the control of circadian rhythms, cellular growth, and survival, is negatively regulated by m6A. Inhibition of Ck1δ mRNA methylation leads to increased translation of two alternatively spliced CK1δ isoforms, CK1δ1 and CK1δ2, uncharacterized until now. The expression ratio between these isoforms is tissue-specific, CK1δ1 and CK1δ2 have different kinase activities, and they cooperate in the phosphorylation of the circadian clock protein PER2. While CK1δ1 accelerates the circadian clock by promoting the decay of PER2 proteins, CK1δ2 slows it down by stabilizing PER2 via increased phosphorylation at a key residue on PER2 protein. These observations challenge the previously established model of PER2 phosphorylation and, given the multiple functions and targets of CK1δ, the existence of two isoforms calls for a re-evaluation of past research when CK1δ1 and CK1δ2 were simply CK1δ.


Asunto(s)
Quinasa Idelta de la Caseína/genética , Relojes Circadianos/genética , Metilación , Metiltransferasas/genética , ARN Mensajero/genética , Animales , Quinasa Idelta de la Caseína/metabolismo , Masculino , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas , Empalme del ARN/genética , ARN Mensajero/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(23): 5986-5991, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29784789

RESUMEN

Multisite phosphorylation of the PERIOD 2 (PER2) protein is the key step that determines the period of the mammalian circadian clock. Previous studies concluded that an unidentified kinase is required to prime PER2 for subsequent phosphorylation by casein kinase 1 (CK1), an essential clock component that is conserved from algae to humans. These subsequent phosphorylations stabilize PER2, delay its degradation, and lengthen the period of the circadian clock. Here, we perform a comprehensive biochemical and biophysical analysis of mouse PER2 (mPER2) priming phosphorylation and demonstrate, surprisingly, that CK1δ/ε is indeed the priming kinase. We find that both CK1ε and a recently characterized CK1δ2 splice variant more efficiently prime mPER2 for downstream phosphorylation in cells than the well-studied splice variant CK1δ1. While CK1 phosphorylation of PER2 was previously shown to be robust to changes in the cellular environment, our phosphoswitch mathematical model of circadian rhythms shows that the CK1 carboxyl-terminal tail can allow the period of the clock to be sensitive to cellular signaling. These studies implicate the extreme carboxyl terminus of CK1 as a key regulator of circadian timing.


Asunto(s)
Caseína Cinasa 1 épsilon/metabolismo , Quinasa Idelta de la Caseína/metabolismo , Ritmo Circadiano/fisiología , Proteínas Circadianas Period/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Proteínas Circadianas Period/genética , Fosforilación
10.
Mol Cell ; 66(4): 437-438, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28525736

RESUMEN

Circadian clocks signal and adapt to an ever-changing world by juggling a panoply of transcriptional and post-translational modifications. In this issue of Molecular Cell, Gustafson et al. (2017) report an additional requirement for accurate timekeeping, a cis/trans conformational flicker in the transcriptional activation domain of the core clock protein BMAL1.


Asunto(s)
Factores de Transcripción ARNTL/genética , Relojes Circadianos , Proteínas CLOCK/genética , Ritmo Circadiano , Procesamiento Proteico-Postraduccional
11.
Front Neurol ; 8: 161, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28496429

RESUMEN

An approximately 24-h biological timekeeping mechanism called the circadian clock is present in virtually all light-sensitive organisms from cyanobacteria to humans. The clock system regulates our sleep-wake cycle, feeding-fasting, hormonal secretion, body temperature, and many other physiological functions. Signals from the master circadian oscillator entrain peripheral clocks using a variety of neural and hormonal signals. Even centrally controlled internal temperature fluctuations can entrain the peripheral circadian clocks. But, unlike other chemical reactions, the output of the clock system remains nearly constant with fluctuations in ambient temperature, a phenomenon known as temperature compensation. In this brief review, we focus on recent advances in our understanding of the posttranslational modifications, especially a phosphoswitch mechanism controlling the stability of PER2 and its implications for the regulation of temperature compensation.

12.
Sci Adv ; 2(1): e1501292, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26824076

RESUMEN

Glioblastoma multiforme (GBM) is the most common and lethal form of intracranial tumor. We have established a lentivirus-induced mouse model of malignant gliomas, which faithfully captures the pathophysiology and molecular signature of mesenchymal human GBM. RNA-Seq analysis of these tumors revealed high nuclear factor κB (NF-κB) activation showing enrichment of known NF-κB target genes. Inhibition of NF-κB by either depletion of IκB kinase 2 (IKK2), expression of a IκBαM super repressor, or using a NEMO (NF-κB essential modifier)-binding domain (NBD) peptide in tumor-derived cell lines attenuated tumor proliferation and prolonged mouse survival. Timp1, one of the NF-κB target genes significantly up-regulated in GBM, was identified to play a role in tumor proliferation and growth. Inhibition of NF-κB activity or silencing of Timp1 resulted in slower tumor growth in both mouse and human GBM models. Our results suggest that inhibition of NF-κB activity or targeting of inducible NF-κB genes is an attractive therapeutic approach for GBM.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , FN-kappa B/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Quinasa I-kappa B/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Péptidos/genética , Transducción de Señal/genética , Regulación hacia Arriba/genética
13.
Proc Natl Acad Sci U S A ; 109(31): 12662-7, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22778400

RESUMEN

Chronic sleep deprivation perturbs the circadian clock and increases susceptibility to diseases such as diabetes, obesity, and cancer. Increased inflammation is one of the common underlying mechanisms of these diseases, thus raising a hypothesis that circadian-oscillator components may regulate immune response. Here we show that absence of the core clock component protein cryptochrome (CRY) leads to constitutive elevation of proinflammatory cytokines in a cell-autonomous manner. We observed a constitutive NF-κB and protein kinase A (PKA) signaling activation in Cry1(-/-);Cry2(-/-) cells. We further demonstrate that increased phosphorylation of p65 at S276 residue in Cry1(-/-);Cry2(-/-) cells is due to increased PKA signaling activity, likely induced by a significantly high basal level of cAMP, which we detected in these cells. In addition, we report that CRY1 binds to adenylyl cyclase and limits cAMP production. Based on these data, we propose that absence of CRY protein(s) might release its (their) inhibition on cAMP production, resulting in elevated cAMP and increased PKA activation, subsequently leading to NF-κB activation through phosphorylation of p65 at S276. These results offer a mechanistic framework for understanding the link between circadian rhythm disruption and increased susceptibility to chronic inflammatory diseases.


Asunto(s)
Ritmo Circadiano , Criptocromos/metabolismo , Citocinas/biosíntesis , Regulación de la Expresión Génica , Adenilil Ciclasas/genética , Adenilil Ciclasas/inmunología , Adenilil Ciclasas/metabolismo , Animales , Línea Celular , Criptocromos/genética , Criptocromos/inmunología , AMP Cíclico/genética , AMP Cíclico/inmunología , AMP Cíclico/metabolismo , Citocinas/genética , Citocinas/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Fosforilación/genética , Fosforilación/inmunología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Factor de Transcripción ReIA/metabolismo
14.
PLoS One ; 4(11): e7709, 2009 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-19888449

RESUMEN

BACKGROUND: The JNK pathway is a mitogen-activated protein (MAP) kinase pathway involved in the regulation of numerous physiological processes during development and in response to environmental stress. JNK activity is controlled by two MAPK kinases (MAPKK), Mkk4 and Mkk7. Mkk7 plays a prominent role upon Tumor Necrosis Factor (TNF) stimulation. Eiger, the unique TNF-superfamily ligand in Drosophila, potently activates JNK signaling through the activation of the MAPKKK Tak1. METHODOLOGY/PRINCIPAL FINDINGS: In a dominant suppressor screen for new components of the Eiger/JNK-pathway in Drosophila, we have identified an allelic series of the Mkk4 gene. Our genetic and biochemical results demonstrate that Mkk4 is dispensable for normal development and host resistance to systemic bacterial infection but plays a non-redundant role as a MAPKK acting in parallel to Hemipterous/Mkk7 in dTAK1-mediated JNK activation upon Eiger and Imd pathway activation. CONCLUSIONS/SIGNIFICANCE: In contrast to mammals, it seems that in Drosophila both MAPKKs, Hep/Mkk7 and Mkk4, are required to induce JNK upon TNF or pro-inflammatory stimulation.


Asunto(s)
Drosophila melanogaster/fisiología , Regulación Enzimológica de la Expresión Génica , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa 4/fisiología , MAP Quinasa Quinasa 7/fisiología , Quinasas Quinasa Quinasa PAM/fisiología , Alelos , Animales , Cruzamientos Genéticos , Drosophila melanogaster/enzimología , Inflamación , Mutación , Fenotipo , Células Fotorreceptoras de Invertebrados/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
16.
Genetics ; 171(4): 1683-94, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16079232

RESUMEN

Signaling by tumor necrosis factors (TNFs) plays a prominent role in mammalian development and disease. To fully understand this complex signaling pathway it is important to identify all regulators and transduction components. A single TNF family member, Eiger, is encoded in the Drosophila genome, offering the possibility of applying genetic approaches for pursuing this goal. Here we present a screen for the isolation of novel genes involved in the TNF/Eiger pathway. On the basis of Eiger's ability to potently activate Jun-N-terminal kinase (JNK) and trigger apoptosis, we used the Drosophila eye to establish an assay for dominant suppressors of this activity. In a large-scale screen the Drosophila homolog of TAB2/3 (dTAB2) was identified as an essential component of the Eiger-JNK pathway. Genetic epistasis and biochemical protein-protein interaction assays assign an adaptor role to dTAB2, linking dTRAF1 to the JNKKK dTAK1, demonstrating a conserved mechanism of TNF signal transduction in mammals and Drosophila. Thus, in contrast to morphogenetic processes, such as dorsal closure of the embryo, in which the JNK pathway is activated by the JNKKK Slipper, Eiger uses the dTAB2-dTAK1 module to induce JNK signaling activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Proteínas de la Membrana/metabolismo , Fenotipo , Transducción de Señal/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Pruebas Genéticas , Vectores Genéticos/genética , Immunoblotting , Inmunoprecipitación , Luciferasas , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas de la Membrana/genética , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis , Análisis de Secuencia de ADN , Supresión Genética/genética , Factor 1 Asociado a Receptor de TNF/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...