Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biosci Rep ; 41(1)2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33306121

RESUMO

BACKGROUND: The family with sequence similarity 20-member C (Fam20C) kinase plays important roles in physiopathological process and is responsible for majority of the secreted phosphoproteome, including substrates associated with tumor cell migration. However, it remains unclear whether Fam20C plays a role in cancers. Here, we aimed to analyze the expression and prognostic value of Fam20C in pan-cancer and to gain insights into the association between Fam20C and immune infiltration. METHODS: We analyzed Fam20C expression patterns and the associations between Fam20C expression levels and prognosis in pan-cancer via the ONCOMINE, TIMER (Tumor Immune Estimation Resource), PrognoScan, GEPIA (Gene Expression Profiling Interactive Analysis), and Kaplan-Meier Plotter databases. After that, GEPIA and TIMER databases were applied to investigate the relations between Fam20C expression and immune infiltration across different cancer types, especially BLCA (bladder urothelial carcinoma), LGG (brain lower grade glioma), and STAD (stomach adenocarcinoma). RESULTS: Compared with adjacent normal tissues, Fam20C was widely expressed across many cancers. In general, Fam20C showed a detrimental role in pan-cancer, it was positively associated with poor survival of BLCA, LGG, and STAD patients. Specifically, based on TCGA (The Cancer Genome Atlas) database, a high expression level of Fam20C was associated with worse prognostic value in stages T2-T4 and stages N0-N2 in the cohort of STAD patients. Moreover, Fam20C expression had positive associations with immune infiltration, including CD4+ T cells, macrophages, neutrophils, and dendritic cells, and other diverse immune cells in BLCA, LGG, and STAD. CONCLUSION: Fam20C may serve as a promising prognostic biomarker in pan-cancer and has positive associations with immune infiltrates.


Assuntos
Caseína Quinase I/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Neoplasias/imunologia , Caseína Quinase I/genética , Proteínas da Matriz Extracelular/genética , Humanos , Estimativa de Kaplan-Meier , Metástase Linfática , Neoplasias/genética , Neoplasias/patologia , Oncogenes , Prognóstico , RNA Mensageiro/genética
2.
Elife ; 92020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33295874

RESUMO

Circadian clocks in fungi and animals are driven by a functionally conserved transcription-translation feedback loop. In Neurospora crassa, negative feedback is executed by a complex of Frequency (FRQ), FRQ-interacting RNA helicase (FRH), and casein kinase I (CKI), which inhibits the activity of the clock's positive arm, the White Collar Complex (WCC). Here, we show that the prd-2 (period-2) gene, whose mutation is characterized by recessive inheritance of a long 26 hr period phenotype, encodes an RNA-binding protein that stabilizes the ck-1a transcript, resulting in CKI protein levels sufficient for normal rhythmicity. Moreover, by examining the molecular basis for the short circadian period of upf-1prd-6 mutants, we uncovered a strong influence of the Nonsense Mediated Decay pathway on CKI levels. The finding that circadian period defects in two classically derived Neurospora clock mutants each arise from disruption of ck-1a regulation is consistent with circadian period being exquisitely sensitive to levels of casein kinase I.


Assuntos
Caseína Quinase I/fisiologia , Relógios Circadianos/fisiologia , Proteínas Fúngicas/fisiologia , Neurospora crassa/fisiologia , Caseína Quinase I/metabolismo , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Genes Fúngicos/genética , Genes Fúngicos/fisiologia , Neurospora crassa/enzimologia , Neurospora crassa/genética
3.
J Physiol ; 593(19): 4439-52, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26174743

RESUMO

KEY POINTS: Increased NMDA receptor activity and excitability of presympathetic neurons in the hypothalamus can increase sympathetic nerve discharges leading to hypertension. In this study, we determined how protein kinases and phosphatases are involved in regulating NMDA receptor activity and firing activity of presympathetic neurons in the hypothalamus in normotensive and hypertensive rats. We show that casein kinase-1 inhibition increases NMDA receptor activity and excitability of presympathetic neurons in the hypothalamus and augments sympathetic nerve discharges in normotensive, but not in hypertensive, rats. Our data indicate that casein kinase-1 tonically regulates NMDA receptor activity by interacting with casein kinase-2 and protein phosphatases in the hypothalamus and that imbalance of NMDA receptor phosphorylation can augment the excitability of hypothalamic presympathetic neurons and sympathetic nerve discharges in hypertension. These findings help us understand the neuronal mechanism of hypertension, and reducing the NMDA receptor phosphorylation level may be effective for treating neurogenic hypertension. ABSTRACT: Increased N-methyl-d-aspartate receptor (NMDAR) activity in the paraventricular nucleus (PVN) of the hypothalamus is involved in elevated sympathetic outflow in hypertension. However, the molecular mechanisms underlying augmented NMDAR activity in hypertension remain unclear. In this study, we determined the role of casein kinase-1 (CK1) in regulating NMDAR activity in the PVN. NMDAR-mediated excitatory postsynaptic currents (EPSCs) and puff NMDA-elicited currents were recorded in spinally projecting PVN neurons in spontaneously hypertensive rats (SHRs) and Wistar-Kyoto (WKY) rats. The basal amplitudes of evoked NMDAR-EPSCs and puff NMDA currents were significantly higher in SHRs than in WKY rats. The CK1 inhibitor PF4800567 or PF670462 significantly increased the amplitude of NMDAR-EPSCs and puff NMDA currents in PVN neurons in WKY rats but not in SHRs. PF4800567 caused an NMDAR-dependent increase in the excitability of PVN neurons only in WKY rats. Also, the CK1ε protein level in the PVN was significantly lower in SHRs than in WKY rats. Furthermore, intracerebroventricular infusion of PF4800567 increased blood pressure and lumbar sympathetic nerve activity in WKY rats, and this effect was eliminated by microinjection of the NMDAR antagonist into the PVN. In addition, PF4800567 failed to increase NMDAR activity in brain slices of WKY rats pretreated with the protein phosphatase 1/2A, calcineurin, or casein kinase-2 inhibitor. Our findings suggest that CK1 tonically suppresses NMDAR activity in the PVN by reducing the NMDAR phosphorylation level. Diminished CK1 activity may contribute to potentiated glutamatergic synaptic input to PVN presympathetic neurons and elevated sympathetic vasomotor tone in neurogenic hypertension.


Assuntos
Caseína Quinase I/fisiologia , Hipertensão/fisiopatologia , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Masculino , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Sistema Nervoso Simpático/fisiologia
4.
J Cell Biol ; 208(6): 745-59, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25778921

RESUMO

Casein kinase 1δ/ε (CK1δ/ε) and their yeast homologue Hrr25 are essential for cell growth. Further, CK1δ is overexpressed in several malignancies, and CK1δ inhibitors have shown promise in several preclinical animal studies. However, the substrates of Hrr25 and CK1δ/ε that are necessary for cell growth and survival are unknown. We show that Hrr25 is essential for ribosome assembly, where it phosphorylates the assembly factor Ltv1, which causes its release from nascent 40S subunits and allows subunit maturation. Hrr25 inactivation or expression of a nonphosphorylatable Ltv1 variant blocked Ltv1 release in vitro and in vivo, and prevented entry into the translation-like quality control cycle. Conversely, phosphomimetic Ltv1 variants rescued viability after Hrr25 depletion. Finally, Ltv1 knockdown in human breast cancer cells impaired apoptosis induced by CK1δ/ε inhibitors, establishing that the antiproliferative activity of these inhibitors is due, at least in part, to disruption of ribosome assembly. These findings validate the ribosome assembly pathway as a novel target for the development of anticancer therapeutics.


Assuntos
Caseína Quinase I/fisiologia , Caseína Quinase Idelta/fisiologia , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/enzimologia , Linhagem Celular Tumoral , Humanos , Proteínas Nucleares/metabolismo , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Ribossômicas/metabolismo , Saccharomyces cerevisiae/citologia
5.
J Clin Invest ; 125(4): 1393-5, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25798614

RESUMO

RAS-driven cancers exhibit variable dependency on autophagy for survival; however, it is not fully understood how. In this issue of the JCI, Cheong and colleagues demonstrate that RAS-dependent elevation of casein kinase 1α (CK1α) negatively regulates autophagy at the level of autophagy gene transcription. Moreover, combined inhibition of both CK1α and autophagy reduced proliferation of RAS-driven tumors. The results of this study provide insight into the connection between mutant RAS and autophagy, and suggest targeting CK1α as a potential therapeutic strategy to modulate autophagy in RAS-driven cancers.


Assuntos
Autofagia/fisiologia , Caseína Quinase I/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/enzimologia , Proteína Oncogênica p21(ras)/fisiologia , Animais , Humanos
6.
J Clin Invest ; 125(4): 1401-18, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25798617

RESUMO

Activating mutations in the RAS oncogene are common in cancer but are difficult to therapeutically target. RAS activation promotes autophagy, a highly regulated catabolic process that metabolically buffers cells in response to diverse stresses. Here we report that casein kinase 1α (CK1α), a ubiquitously expressed serine/threonine kinase, is a key negative regulator of oncogenic RAS-induced autophagy. Depletion or pharmacologic inhibition of CK1α enhanced autophagic flux in oncogenic RAS-driven human fibroblasts and multiple cancer cell lines. FOXO3A, a master longevity mediator that transcriptionally regulates diverse autophagy genes, was a critical target of CK1α, as depletion of CK1α reduced levels of phosphorylated FOXO3A and increased expression of FOXO3A-responsive genes. Oncogenic RAS increased CK1α protein abundance via activation of the PI3K/AKT/mTOR pathway. In turn, elevated levels of CK1α increased phosphorylation of nuclear FOXO3A, thereby inhibiting transactivation of genes critical for RAS-induced autophagy. In both RAS-driven cancer cells and murine xenograft models, pharmacologic CK1α inactivation synergized with lysosomotropic agents to inhibit growth and promote tumor cell death. Together, our results identify a kinase feedback loop that influences RAS-dependent autophagy and suggest that targeting CK1α-regulated autophagy offers a potential therapeutic opportunity to treat oncogenic RAS-driven cancers.


Assuntos
Autofagia/fisiologia , Caseína Quinase I/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/enzimologia , Proteína Oncogênica p21(ras)/fisiologia , Sequência de Aminoácidos , Animais , Caseína Quinase I/antagonistas & inibidores , Retroalimentação Fisiológica , Fibroblastos/enzimologia , Fibroblastos/fisiologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/fisiologia , RNA Interferente Pequeno/farmacologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Serina-Treonina Quinases TOR/fisiologia , Ativação Transcricional
7.
Methods Enzymol ; 551: 175-95, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25662457

RESUMO

The circadian clock mechanism in organisms as diverse as cyanobacteria and humans involves both transcriptional and posttranslational regulation of key clock components. One of the roles for the posttranslational regulation is to time the degradation of the targeted clock proteins, so that their oscillation profiles are out of phase with respect to those of the mRNAs from which they are translated. In Drosophila, the circadian transcriptional regulator PERIOD (PER) is targeted for degradation by a kinase (DOUBLETIME or DBT) orthologous to mammalian kinases (CKIɛ and CKIδ) that also target mammalian PER. Since these kinases are not regulated by second messengers, the mechanism (if any) for their regulation is not known. We are investigating the possibility that regulation of DBT is conferred by other proteins that associate with DBT and PER. In this chapter, the methods we are employing to identify and analyze these factors are discussed. These methods include expression of wild type and mutant proteins with the GAL4/UAS binary expression approach, analysis of DBT in Drosophila S2 cells, in vitro kinase assays with DBT isolated from S2 cells, and proteomic analysis of DBT-containing complexes and of DBT phosphorylation with mass spectrometry. The work has led to the discovery of a previously unrecognized circadian rhythm component (Bride of DBT, a noncanonical FK506-binding protein) and the mapping of autophosphorylation sites within the DBT C-terminal domain with potential regulatory roles.


Assuntos
Caseína Quinase I/fisiologia , Relógios Circadianos , Drosophila melanogaster/enzimologia , Animais , Linhagem Celular , Drosophila melanogaster/fisiologia , Expressão Gênica , Fosforilação , Processamento de Proteína Pós-Traducional
8.
Oncogene ; 34(4): 474-84, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24469046

RESUMO

Sprouty2 (SPRY2) is a potent negative regulator of receptor tyrosine kinase signaling, and is implicated as a tumor suppressor. SPRY2 inhibits FGF-RAS-ERK signaling by binding to growth factor receptor bound protein 2 (GRB2) during fibroblast growth factor receptor (FGFR) activation, disrupting the GRB2-SOS (son of sevenless) complex that transduces signals from FGFR to RAS. SPRY2 binding to GRB2 is modulated by phosphorylation but the key regulatory kinase(s) are not known. Prior studies identified the frequent presence of CK1 phosphorylation motifs on SPRY2. We therefore tested if CK1 has a role in SPRY2 phosphorylation and function. Loss of CK1 binding and inhibition of CK1 activity by two structurally distinct small molecules abrogated SPRY2 inhibition of FGF-ERK signaling, leading to decreased SPRY2 interaction with GRB2. Moreover, CK1 activity and binding are necessary for SPRY2 inhibition of FGF-stimulated neurite outgrowth in PC12 cells. Consistent with its proposed role as an inhibitor of FGF signaling, we find that CSNK1E transcript abundance negatively correlates with FGF1/FGF7 message in human gastric cancer samples. Modulation of CK1 activity may be therapeutically useful in the treatment of FGF/SPRY2-related diseases.


Assuntos
Caseína Quinase I/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Proteína Adaptadora GRB2/fisiologia , Humanos , Neuritos/fisiologia , Fosforilação , Ratos , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Neoplasias Gástricas/metabolismo
9.
Cell Rep ; 9(5): 1692-1702, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25464840

RESUMO

The casein kinase 1 (CK1) family participates in various cellular processes in eukaryotes, but CK1 function in higher plants remains largely unknown. Here, we characterize the function of Arabidopsis CK1 in the regulation of ethylene biosynthesis. Etiolated seedlings of a CK1.8-deficient mutant, ck1.8-1, showed characteristic ethylene-specific constitutive responses due to overaccumulation of ethylene. Biochemical and physiological studies showed that CK1.8 phosphorylates ACS5, a key enzyme of ethylene biosynthesis, at threonine 463 to promote its interaction with the E3 ubiquitin ligase Ethylene Overproduction 1 (ETO1). Deficiency of CK1.8 leads to the accumulation of ACS5, and transgenic plants harboring a dephosphorylation-mimic ACS5(T463A) showed constitutive ethylene responses, confirming the role of CK1.8 in regulating ACS5 stability by phosphorylation and demonstrating that CK1.8 is an important regulator of ethylene biosynthesis. CK1.8 expression is feedback regulated by ethylene. Our studies provide insight into the regulation of ACS5 and ethylene biosynthesis.


Assuntos
Arabidopsis/enzimologia , Caseína Quinase I/fisiologia , Etilenos/biossíntese , Liases/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Proteólise , Plântula/enzimologia
10.
EMBO Rep ; 15(8): 862-70, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24968893

RESUMO

Autophagy is the major pathway for the delivery of cytoplasmic material to the vacuole or lysosome. Selective autophagy is mediated by cargo receptors, which link the cargo to the scaffold protein Atg11 and to Atg8 family proteins on the forming autophagosomal membrane. We show that the essential kinase Hrr25 activates the cargo receptor Atg19 by phosphorylation, which is required to link cargo to the Atg11 scaffold, allowing selective autophagy to proceed. We also find that the Atg34 cargo receptor is regulated in a similar manner, suggesting a conserved mechanism.


Assuntos
Caseína Quinase I/fisiologia , Receptores de Superfície Celular/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/enzimologia , Proteínas de Transporte Vesicular/metabolismo , Sequência de Aminoácidos , Autofagia , Proteínas Relacionadas à Autofagia , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Transporte Proteico , Receptores de Superfície Celular/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Transporte Vesicular/química
11.
Antimicrob Agents Chemother ; 58(3): 1501-15, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24366737

RESUMO

Protein kinase inhibitors have emerged as new drugs in various therapeutic areas, including leishmaniasis, an important parasitic disease. Members of the Leishmania casein kinase 1 (CK1) family represent promising therapeutic targets. Leishmania casein kinase 1 isoform 2 (CK1.2) has been identified as an exokinase capable of phosphorylating host proteins, thus exerting a potential immune-suppressive action on infected host cells. Moreover, its inhibition reduces promastigote growth. Despite these important properties, its requirement for intracellular infection and its chemical validation as a therapeutic target in the disease-relevant amastigote stage remain to be established. In this study, we used a multidisciplinary approach combining bioinformatics, biochemical, and pharmacological analyses with a macrophage infection assay to characterize and define Leishmania CK1.2 as a valid drug target. We show that recombinant and transgenic Leishmania CK1.2 (i) can phosphorylate CK1-specific substrates, (ii) is sensitive to temperature, and (iii) is susceptible to CK1-specific inhibitors. CK1.2 is constitutively expressed at both the promastigote insect stage and the vertebrate amastigote stage. We further demonstrated that reduction of CK1 activity by specific inhibitors, such as D4476, blocks promastigote growth, strongly compromises axenic amastigote viability, and decreases the number of intracellular Leishmania donovani and L. amazonensis amastigotes in infected macrophages. These results underline the potential role of CK1 kinases in intracellular survival. The identification of differences in structure and inhibition profiles compared to those of mammalian CK1 kinases opens new opportunities for Leishmania CK1.2 antileishmanial drug development. Our report provides the first chemical validation of Leishmania CK1 protein kinases, required for amastigote intracellular survival, as therapeutic targets.


Assuntos
Caseína Quinase I/efeitos dos fármacos , Leishmania donovani/efeitos dos fármacos , Animais , Benzamidas/farmacologia , Caseína Quinase I/antagonistas & inibidores , Caseína Quinase I/genética , Caseína Quinase I/fisiologia , Sequência Conservada/genética , Cricetinae/parasitologia , Feminino , Imidazóis/farmacologia , Indóis/farmacologia , Isoquinolinas/farmacologia , Leishmania donovani/enzimologia , Leishmania donovani/genética , Leishmania donovani/patogenicidade , Leishmania donovani/fisiologia , Leishmaniose Visceral/tratamento farmacológico , Leishmaniose Visceral/parasitologia , Macrófagos/parasitologia , Camundongos Endogâmicos C57BL , Floroglucinol/análogos & derivados , Floroglucinol/farmacologia , Alinhamento de Sequência , Tripanossomicidas/farmacologia
12.
Adv Exp Med Biol ; 740: 703-30, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22453966

RESUMO

Calcium/calmodulin-stimulated protein kinases can be classified as one of two types - restricted or multifunctional. This family of kinases contains several structural similarities: all possess a calmodulin binding motif and an autoinhibitory region. In addition, all of the calcium/calmodulin-stimulated protein kinases examined in this chapter are regulated by phosphorylation, which either activates or inhibits their kinase activity. However, as the multifunctional calcium/calmodulin-stimulated protein kinases are ubiquitously expressed, yet regulate a broad range of cellular functions, additional levels of regulation that control these cell-specific functions must exist. These additional layers of control include gene expression, signaling pathways, and expression of binding proteins and molecular targeting. All of the multifunctional calcium/calmodulin-stimulated protein kinases examined in this chapter appear to be regulated by these additional layers of control, however, this does not appear to be the case for the restricted kinases.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/química , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/química , Caseína Quinase I/síntese química , Caseína Quinase I/fisiologia , Humanos , Quinase de Cadeia Leve de Miosina/química , Quinase de Cadeia Leve de Miosina/fisiologia , Fosforilase Quinase/química , Fosforilase Quinase/fisiologia , Fosforilação
14.
Neuron ; 67(5): 810-20, 2010 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-20826312

RESUMO

Cyclic AMP signaling in Drosophila mushroom body neurons, anchored by the adenylyl cyclase encoded by the rutabaga gene, is indispensable for olfactory memory formation. From a screen for new memory mutants, we identified alleles of the gilgamesh (gish) gene, which encodes a casein kinase Iγ homolog that is preferentially expressed in the mushroom body neurons. The gish-encoded kinase participates in the physiology of these neurons underlying memory formation since the mutant memory deficit was rescued with expression of a gish cDNA in these neurons only during adulthood. A cellular memory trace, detected as increased calcium influx into the α'/ß' neuron processes in response to the odor used for conditioning, was disrupted in gish mutants. Epistasis experiments indicated a lack of genetic interactions between gish and rutabaga. Therefore, gish participates in a rutabaga-independent pathway for memory formation and accounts for some of the residual learning that occurs in rutabaga mutants.


Assuntos
Adenilil Ciclases/metabolismo , Aprendizagem da Esquiva/fisiologia , Caseína Quinase I/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Olfato/fisiologia , Adenilil Ciclases/genética , Animais , Animais Geneticamente Modificados , Comportamento Animal , Caseína Quinase I/deficiência , Caseína Quinase I/genética , Condicionamento Clássico/fisiologia , Drosophila , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Regulação da Expressão Gênica/genética , Transtornos da Memória/genética , Corpos Pedunculados/crescimento & desenvolvimento , Corpos Pedunculados/metabolismo , Mutação/genética , Odorantes
15.
Nat Cell Biol ; 12(5): 500-6, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20383139

RESUMO

During meiosis, the cohesin complexes that maintain sister chromatid cohesion are lost in a stepwise manner. At meiosis I the cohesin subunit Rec8 is cleaved only along the chromosome arms; until meiosis II it is protected at centromeres by the action of shugoshin (Sgo1)-protein phosphatase 2A (PP2A). Although this regulation hypothetically involves phosphorylation that is antagonized by Sgo1-PP2A, the kinase and substrate that are responsible are as yet unknown. Using a genetic screen for 'anti-shugoshin', we identify Hhp2, an orthologue of casein kinase 1delta/epsilon (CK1), as a factor required for Rec8 cleavage in fission yeast. We show that CK1, rather than a Polo-like kinase that is widely believed to do so, acts as the cohesin kinase to promote this cleavage during meiosis. Crucially, forced localization of excess Hhp2 at the pericentromeric region abrogates the ability of Sgo1-PP2A to protect centromeric Rec8. Thus, our studies prove the key notion that the balance between Rec8 phosphorylation and its dephosphorylation by Sgo1-PP2A regulates the step-wise loss of chromosomal cohesion in meiosis.


Assuntos
Caseína Quinase I/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Endopeptidases/metabolismo , Fosfoproteínas/metabolismo , Proteína Fosfatase 2/fisiologia , Proteínas de Schizosaccharomyces pombe/metabolismo , Centrômero , Cromátides , Hidrólise , Meiose , Fosforilação , Schizosaccharomyces , Separase
16.
BMC Cell Biol ; 11: 17, 2010 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-20205947

RESUMO

BACKGROUND: In yeast, glucose-dependent degradation of the Mth1 protein, a corepressor of the glucose transporter gene (HXT) repressor Rgt1, is a crucial event enabling expression of several HXT. This event occurs through a signaling pathway that involves the Rgt2 and Snf3 glucose sensors and yeast casein kinase 1 and 2 (Yck1/2). In this study, we examined whether the glucose sensors directly couple with Yck1/2 to convert glucose binding into an intracellular signal that leads to the degradation of Mth1. RESULTS: High levels of glucose induce degradation of Mth1 through the Rgt2/Snf3 glucose signaling pathway. Fluorescence microscopy analysis indicates that, under glucose-limited conditions, GFP-Mth1 is localized in the nucleus and does not shuttle between the nucleus and cytoplasm. If glucose-induced degradation is prevented due to disruption of the Rgt2/Snf3 pathway, GFP-Mth1 accumulates in the nucleus. When engineered to be localized to the cytoplasm, GFP-Mth1 is degraded regardless of the presence of glucose or the glucose sensors. In addition, removal of Grr1 from the nucleus prevents degradation of GFP-Mth1. These results suggest that glucose-induced, glucose sensor-dependent Mth1 degradation occurs in the nucleus. We also show that, like Yck2, Yck1 is localized to the plasma membrane via C-terminal palmitoylation mediated by the palmitoyl transferase Akr1. However, glucose-dependent degradation of Mth1 is not impaired in the absence of Akr1, suggesting that a direct interaction between the glucose sensors and Yck1/2 is not required for Mth1 degradation. CONCLUSION: Glucose-induced, glucose sensor-regulated degradation of Mth1 occurs in the nucleus and does not require direct interaction of the glucose sensors with Yck1/2.


Assuntos
Caseína Quinase I/fisiologia , Glucose/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/enzimologia , Aciltransferases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caseína Quinase I/metabolismo , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas F-Box/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
17.
J Biol Chem ; 284(47): 32384-94, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19759023

RESUMO

The ubiquitin ligase murine double minute clone 2 (MDM2) mediates ubiquitination and degradation of the tumor suppressor p53. The activation and stabilization of p53 by contrast is maintained by enzymes catalyzing p53 phosphorylation and acetylation. Casein kinase 1 (CK1) is one such enzyme; it stimulates p53 after transforming growth factor-beta treatment, irradiation, or DNA virus infection. We analyzed whether CK1 regulates p53 protein stability in unstressed conditions. Depletion of CK1 using small interfering RNA or inhibition of CK1 using the kinase inhibitor (D4476) activated p53 and destabilized E2F-1, indicating that steady-state levels of these proteins are controlled by CK1. Co-immunoprecipitation of endogenous CK1 with MDM2 occurred in undamaged cells, indicating the existence of a stable multiprotein complex, and as such, we evaluated whether the MDM2 Nutlin had similar pharmacological properties to the CK1 inhibitor D4476. Indeed, D4476 or Nutlin treatments resulted in the same p53 and E2F-1 steady-state protein level changes, indicating that the MDM2 x CK1 complex is both a negative regulator of p53 and a positive regulator of E2F-1 in undamaged cells. Although the treatment of cells with D4476 resulted in a partial p53-dependent growth arrest, the induction of p53-independent apoptosis by D4476 suggested a critical role for the MDM2 x CK1 complex in maintaining E2F-1 anti-apoptotic signaling. These data highlighting a pharmacological similarity between MDM2 and CK1 small molecule inhibitors and the fact that CK1 and MDM2 form a stable complex suggest that the MDM2 x CK1 complex is a component of a genetic pathway that co-regulates the stability of the p53 and E2F-1 transcription factors.


Assuntos
Caseína Quinase I/fisiologia , Fator de Transcrição E2F1/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose , Benzamidas/farmacologia , Caseína Quinase I/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Imidazóis/farmacologia , Modelos Biológicos , Fosforilação , Piperazinas/farmacologia , Isoformas de Proteínas , RNA Interferente Pequeno/metabolismo
18.
Brain Nerve ; 61(2): 161-6, 2009 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-19235466

RESUMO

In most neurodegenerative disorders, distinctive intracellular inclusion bodies are found in degenerative neurons, which are known to be neuropathological hallmarks of diseases. Recently, TAR DNA-binding protein of 43 KDa (TDP-43) has been identified as a major constituent protein of ubiquitin-positive inclusions in brains with frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). These disorders are now referred to as TDP-43 proteinopathy. TDP-43 deposited in brains with FTLD and ALS was found to be phosphorylated and ubiquitinated. To study the role of these posttranslational modifications in the formation of TDP-43 aggregates, we have produced polyclonal and monoclonal antibodies specific for TDP-43 phosphorylated at Ser409 and Ser 410. These antibodies specifically recognized abnormally phosphorylated TDP-43, but not normal TDP-43 in immunohistochemical analyses of brains of FTLD and ALS patients. Immunoblot analyses using these antibodies showed that phosphorylated and fragmented TDP-43 was deposited in diseased brains. Furthermore, we identified casein kinase 1 as a candidate protein kinase, which was responsible for abnormal phosphorylation of TDP-43. Phosphorylated recombinant TDP-43 proteins were demonstrated to be easier to fibrillate than wild-type TDP-43 in vitro. Recent discoveries of the missense mutations in the TDP-43 gene in familial or sporadic ALS cases prove a direct link between altered TDP-43 function and neurodegeneration. Elucidating the biochemical processes responsible for phosphorylation, fragmentation, and intracellular aggregation of TDP-43 may provide important insights into the pathogenesis of TDP-43 proteinopathy.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Doenças Neurodegenerativas/genética , Esclerose Lateral Amiotrófica/genética , Caseína Quinase I/fisiologia , Proteínas de Ligação a DNA/genética , Demência/genética , Humanos , Mutação de Sentido Incorreto , Fosforilação , Processamento de Proteína Pós-Traducional , Ubiquitinação
19.
Mol Biol Cell ; 20(5): 1592-604, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19116312

RESUMO

The evolutionarily conserved adaptor protein-3 (AP-3) complex mediates cargo-selective transport to lysosomes and lysosome-related organelles. To identify proteins that function in AP-3-mediated transport, we performed a genome-wide screen in Saccharomyces cerevisiae for defects in the vacuolar maturation of alkaline phosphatase (ALP), a cargo of the AP-3 pathway. Forty-nine gene deletion strains were identified that accumulated precursor ALP, many with established defects in vacuolar protein transport. Maturation of a vacuolar membrane protein delivered via a separate, clathrin-dependent pathway, was affected in all strains except those with deletions of YCK3, encoding a vacuolar type I casein kinase; SVP26, encoding an endoplasmic reticulum (ER) export receptor for ALP; and AP-3 subunit genes. Subcellular fractionation and fluorescence microscopy revealed ALP transport defects in yck3Delta cells. Characterization of svp26Delta cells revealed a role for Svp26p in ER export of only a subset of type II membrane proteins. Finally, ALP maturation kinetics in vac8Delta and vac17Delta cells suggests that vacuole inheritance is important for rapid generation of proteolytically active vacuolar compartments in daughter cells. We propose that the cargo-selective nature of the AP-3 pathway in yeast is achieved by AP-3 and Yck3p functioning in concert with machinery shared by other vacuolar transport pathways.


Assuntos
Complexo 3 de Proteínas Adaptadoras/fisiologia , Fosfatase Alcalina/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/metabolismo , Vacúolos/metabolismo , Complexo 3 de Proteínas Adaptadoras/genética , Fosfatase Alcalina/análise , Caseína Quinase I/genética , Caseína Quinase I/metabolismo , Caseína Quinase I/fisiologia , Deleção de Genes , Genoma Fúngico , Proteínas de Fluorescência Verde/análise , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Subunidades Proteicas/genética , Transporte Proteico/genética , Transporte Proteico/fisiologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Transporte Vesicular
20.
J Biol Chem ; 283(42): 28563-73, 2008 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-18669630

RESUMO

The tumor suppressor protein p53 is activated by distinct cellular stresses including radiation, hypoxia, type I interferon, and DNA/RNA virus infection. The transactivation domain of p53 contains a phosphorylation site at Ser20 whose modification stabilizes the binding of the transcriptional co-activator p300 and whose mutation in murine transgenics induces B-cell lymphoma. Although the checkpoint kinase CHK2 is implicated in promoting Ser20 site phosphorylation after irradiation, the enzyme that triggers this phosphorylation after DNA viral infection is undefined. Using human herpesvirus 6B (HHV-6B) as a virus that induces Ser20 site phosphorylation of p53 in T-cells, we sought to identify the kinase responsible for this virus-induced p53 modification. The p53 Ser20 kinase was fractionated and purified using cation, anion, and dye-ligand exchange chromatography. Mass spectrometry identified casein kinase 1 (CK1) and vaccinia-related kinase 1 (VRK1) as enzymes that coeluted with virus-induced Ser20 site kinase activity. Immunodepletion of CK1 but not VRK1 removed the kinase activity from the peak fraction, and bacterially expressed CK1 exhibited Ser20 site kinase activity equivalent to that of the virus-induced native CK1. CK1 modified p53 in a docking-dependent manner, which is similar to other known Ser20 site p53 kinases. Low levels of the CK1 inhibitor D4476 selectively inhibited HHV-6B-induced Ser20 site phosphorylation of p53. However, x-ray-induced Ser20 site phosphorylation of p53 was not blocked by D4476. These data highlight a central role for CK1 as the Ser20 site kinase for p53 in DNA virus-infected cells but also suggest that distinct stresses may selectively trigger different protein kinases to modify the transactivation domain of p53 at Ser20.


Assuntos
Caseína Quinase I/fisiologia , Herpesvirus Humano 6/metabolismo , Infecções por Roseolovirus/virologia , Serina/química , Proteína Supressora de Tumor p53/química , Sequência de Aminoácidos , Antivirais/farmacologia , Caseína Quinase I/química , Catálise , Linhagem Celular Tumoral , Humanos , Espectrometria de Massas/métodos , Dados de Sequência Molecular , Fosforilação , Estrutura Terciária de Proteína , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...