Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(13)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35698877

RESUMEN

Casein kinase 1α (CK1α), acting as one member of the ß-catenin degradation complex, negatively regulates the Wnt/ß-catenin signaling pathway. CK1α knockout usually causes both Wnt/ß-catenin and p53 activation. Our results demonstrated that conditional disruption of CK1α in spermatogonia impaired spermatogenesis and resulted in male mouse infertility. The progenitor cell population was dramatically decreased in CK1α conditional knockout (cKO) mice, while the proliferation of spermatogonial stem cells (SSCs) was not affected. Furthermore, our molecular analyses identified that CK1α loss was accompanied by nuclear stability of p53 protein in mouse spermatogonia, and dual-luciferase reporter and chromatin immunoprecipitation assays revealed that p53 directly targeted the Sox3 gene. In addition, the p53 inhibitor pifithrin α (PFTα) partially rescued the phenotype observed in cKO mice. Collectively, our data suggest that CK1α regulates spermatogenesis and male fertility through p53-Sox3 signaling, and they deepen our understanding of the regulatory mechanism underlying the male reproductive system.


Asunto(s)
Caseína Quinasa Ialfa , Animales , Caseína Quinasa Ialfa/metabolismo , Masculino , Ratones , Factores de Transcripción SOXB1/metabolismo , Espermatogénesis/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
2.
EMBO J ; 39(14): e104410, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32511789

RESUMEN

Casein kinase 1 alpha (CK1α) is a serine/threonine kinase with numerous functions, including regulating the Wnt/ß-catenin and p53 pathways. CK1α has a well-established role in inhibiting the p53 tumor suppressor by binding to MDMX and stimulating MDMX-p53 interaction. MDMX purified from cells contains near-stoichiometric amounts of CK1α, suggesting that MDMX may in turn regulate CK1α function. We present evidence that MDMX is a potent competitive inhibitor of CK1α kinase activity (Ki  = 8 nM). Depletion of MDMX increases CK1α activity and ß-catenin S45 phosphorylation, whereas ectopic MDMX expression inhibits CK1α activity and ß-catenin phosphorylation. The MDMX acidic domain and zinc finger are necessary and sufficient for binding and inhibition of CK1α. P53 binding to MDMX disrupts an intramolecular auto-regulatory interaction and enhances its ability to inhibit CK1α. P53-null mice expressing the MDMXW200S/W201G mutant, defective in CK1α binding, exhibit reduced Wnt/ß-catenin target gene expression and delayed tumor development. Therefore, MDMX is a physiological inhibitor of CK1α and has a role in modulating cellular response to Wnt signaling. The MDMX-CK1α interaction may account for certain p53-independent functions of MDMX.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Vía de Señalización Wnt , Células A549 , Animales , Caseína Quinasa Ialfa/genética , Proteínas de Ciclo Celular/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
Biochem Biophys Res Commun ; 723: 150189, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38852281

RESUMEN

Casein kinase 1α (CK1α) is a serine/threonine protein kinase that acts in various cellular processes affecting cell division and signal transduction. CK1α is present as multiple splice variants that are distinguished by the presence or absence of a long insert (L-insert) and a short carboxyl-terminal insert (S-insert). When overexpressed, zebrafish CK1α splice variants exhibit different biological properties, such as subcellular localization and catalytic activity. However, whether endogenous, alternatively spliced CK1α gene products also differ in their biological functions has yet to be elucidated. Here, we identify a panel of splice variant specific CK1α antibodies and use them to show that four CK1α splice variants are expressed in mammals. We subsequently show that the relative abundance of CK1α splice variants varies across distinct mouse tissues and between various cancer cell lines. Furthermore, we identify pathways whose expression is noticeably altered in cell lines enriched with select splice variants of CK1α. Finally, we show that the S-insert of CK1α promotes the growth of HCT 116 cells as cells engineered to lack the S-insert display decreased cell growth. Together, we provide tools and methods to identify individual CK1α splice variants, which we use to begin to uncover the differential biological properties driven by specific splice variants of mammalian CK1α.


Asunto(s)
Empalme Alternativo , Caseína Quinasa Ialfa , Humanos , Animales , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/genética , Ratones , Línea Celular Tumoral , Proliferación Celular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Células HCT116 , Isoenzimas/genética , Isoenzimas/metabolismo
4.
Bioorg Chem ; 147: 107319, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38593529

RESUMEN

Reactivating p53 activity to restore its anticancer function is an attractive cancer treatment strategy. In this study, we designed and synthesized a series of novel PROTACs to reactivate p53 via the co-degradation of CK1α and CDK7/9 proteins. Bioactivity studies showed that the selected PROTAC 13i exhibited potency antiproliferative activity in MV4-11 (IC50 = 0.096 ± 0.012 µM) and MOLM-13 (IC50 = 0.072 ± 0.014 µM) cells, and induced apoptosis of MV4-11 cells. Western-blot analysis showed that PROTAC 13i triple CK1α and CDK7/9 protein degradation resulted in the significantly increased expression of p53. At the same time, the transcriptional repression due to the degradation significantly reduced downstream gene expression of MYC, MDM2, BCL-2 and MCL-1, and reduced the inflammatory cytokine levels of TNF-α, IL-1ß and IL-6 in PMBCs. These results indicate the beneficial impact of simultaneous CK1α and CDK7/9 degradation for acute myeloid leukemia therapy.


Asunto(s)
Antineoplásicos , Caseína Quinasa Ialfa , Proliferación Celular , Quinasa 9 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes , Ensayos de Selección de Medicamentos Antitumorales , Leucemia Mieloide Aguda , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/metabolismo , Relación Estructura-Actividad , Estructura Molecular , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Relación Dosis-Respuesta a Droga , Apoptosis/efectos de los fármacos , Descubrimiento de Drogas , Línea Celular Tumoral , Proteolisis/efectos de los fármacos , Células Tumorales Cultivadas , Quimera Dirigida a la Proteólisis , Quinasa Activadora de Quinasas Ciclina-Dependientes
5.
J Biol Chem ; 298(8): 102227, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35780831

RESUMEN

The Cullin-RING ligase 4 E3 ubiquitin ligase component Cereblon (CRBN) is a well-established target for a class of small molecules termed immunomodulatory drugs (IMiDs). These drugs drive CRBN to modulate the degradation of a number of neosubstrates required for the growth of multiple cancers. Whereas the mechanism underlying the activation of CRBN by IMiDs is well described, the normal physiological regulation of CRBN is poorly understood. We recently showed that CRBN is activated following exposure to Wnt ligands and subsequently mediates the degradation of a subset of physiological substrates. Among the Wnt-dependent substrates of CRBN is Casein kinase 1α (CK1α), a known negative regulator of Wnt signaling. Wnt-mediated degradation of CK1α occurs via its association with CRBN at a known IMiD binding pocket. Herein, we demonstrate that a small-molecule CK1α agonist, pyrvinium, directly prevents the Wnt-dependent interaction of CRBN with CK1α, attenuating the consequent CK1α degradation. We further show that pyrvinium disrupts the ability of CRBN to interact with CK1α at the IMiD binding pocket within the CRBN-CK1α complex. Of note, this function of pyrvinium is independent of its previously reported ability to enhance CK1α kinase activity. Furthermore, we also demonstrate that pyrvinium attenuates CRBN-induced Wnt pathway activation in vivo. Collectively, these results reveal a novel dual mechanism through which pyrvinium inhibits Wnt signaling by both attenuating the CRBN-mediated destabilization of CK1α and activating CK1α kinase activity.


Asunto(s)
Caseína Quinasa Ialfa , Compuestos de Pirvinio , Caseína Quinasa Ialfa/metabolismo , Compuestos de Pirvinio/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt
6.
Apoptosis ; 28(1-2): 1-19, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36308624

RESUMEN

Eryptosis is a coordinated non-lytic cell death of erythrocytes characterized by cell shrinkage, cell membrane scrambling, Ca2+ influx, ceramide accumulation, oxidative stress, activation of calpain and caspases. Physiologically, it aims at removing damaged or aged erythrocytes from circulation. A plethora of diseases are associated with enhanced eryptosis, including metabolic diseases, cardiovascular pathology, renal and hepatic diseases, hematological disorders, systemic autoimmune pathology, and cancer. This makes eryptosis and eryptosis-regulating signaling pathways a target for therapeutic interventions. This review highlights the eryptotic signaling machinery containing several protein kinases and its small molecular inhibitors with a special emphasis on casein kinase 1α (CK1α), a serine/threonine protein kinase with a broad spectrum of activity. In this review article, we provide a critical analysis of the regulatory role of CK1α in eryptosis, highlight triggers of CK1α-mediated suicidal death of red blood cells, cover the knowledge gaps in understanding CK1α-driven eryptosis and discover the opportunity of CK1α-targeted pharmacological modulation of eryptosis. Moreover, we discuss the directions of future research focusing on uncovering crosstalks between CK1α and other eryptosis-regulating kinases and pathways.


Asunto(s)
Caseína Quinasa Ialfa , Eriptosis , Humanos , Anciano , Caseína Quinasa Ialfa/metabolismo , Apoptosis , Eritrocitos/metabolismo , Estrés Oxidativo , Calcio/metabolismo , Fosfatidilserinas/metabolismo , Ceramidas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
J Cell Sci ; 134(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34730182

RESUMEN

The WAVE regulatory complex (WRC) is the main activator of the Arp2/3 complex, promoting lamellipodial protrusions in migrating cells. The WRC is basally inactive but can be activated by Rac1 and phospholipids, and through phosphorylation. However, the in vivo relevance of the phosphorylation of WAVE proteins remains largely unknown. Here, we identified casein kinase I alpha (CK1α) as a regulator of WAVE, thereby controlling cell shape and cell motility in Drosophila macrophages. CK1α binds and phosphorylates WAVE in vitro. Phosphorylation of WAVE by CK1α appears not to be required for activation but, rather, regulates its stability. Pharmacologic inhibition of CK1α promotes ubiquitin-dependent degradation of WAVE. Consistently, loss of Ck1α but not ck2 function phenocopies the depletion of WAVE. Phosphorylation-deficient mutations in the CK1α consensus sequences within the VCA domain of WAVE can neither rescue mutant lethality nor lamellipodium defects. By contrast, phosphomimetic mutations rescue all cellular and developmental defects. Finally, RNAi-mediated suppression of 26S proteasome or E3 ligase complexes substantially rescues lamellipodia defects in CK1α-depleted macrophages. Therefore, we conclude that basal phosphorylation of WAVE by CK1α protects it from premature ubiquitin-dependent degradation, thus promoting WAVE function in vivo. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Caseína Quinasa Ialfa , Caseína Quinasa Ialfa/genética , Caseína Quinasa Ialfa/metabolismo , Forma de la Célula , Humanos , Inmunidad , Fosforilación , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
8.
J Virol ; 96(7): e0010722, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35293767

RESUMEN

The propagation of the hepatitis C virus (HCV) is regulated in part by the phosphorylation of its nonstructural protein NS5A that undergoes sequential phosphorylation on several highly conserved serine residues and switches from a hypo- to a hyperphosphorylated state. Previous studies have shown that NS5A sequential phosphorylation requires NS3 encoded on the same NS3-NS4A-NS4B-NS5A polyprotein. Subtle mutations in NS3 without affecting its protease activity could affect NS5A phosphorylation. Given the ATPase domain in the NS3 COOH terminus, we tested whether NS3 participates in NS5A phosphorylation similarly to the nucleoside diphosphate kinase-like activity of the rotavirus NSP2 nucleoside triphosphatase (NTPase). Mutations in the NS3 ATP-binding motifs blunted NS5A hyperphosphorylation and phosphorylation at serines 225, 232, and 235, whereas a mutation in the RNA-binding domain did not. The phosphorylation events were not rescued with wild-type NS3 provided in trans. When provided with an NS3 ATPase-compatible ATP analog, N6-benzyl-ATP-γ-S, thiophosphorylated NS5A was detected in the cells expressing the wild-type NS3-NS5B polyprotein. The thiophosphorylation level was lower in the cells expressing NS3-NS5B with a mutation in the NS3 ATP-binding domain. In vitro assays with a synthetic peptide and purified wild-type NS3 followed by dot blotting and mass spectrometry found weak NS5A phosphorylation at serines 222 and 225 that was sensitive to an inhibitor of casein kinase Iα but not helicase. When casein kinase Iα was included in the assay, much stronger phosphorylation was observed at serines 225, 232, and 235. We concluded that NS5A sequential phosphorylation requires the ATP-binding domain of the NS3 helicase and that casein kinase Iα is a potent NS5A kinase. IMPORTANCE For more than 20 years, NS3 was known to participate in NS5A sequential phosphorylation. In the present study, we show for the first time that the ATP-binding domain of NS3 is involved in NS5A phosphorylation. In vitro assays showed that casein kinase Iα is a very potent kinase responsible for NS5A phosphorylation at serines 225, 232, and 235. Our data suggest that ATP binding by NS3 probably results in conformational changes that recruit casein kinase Iα to phosphorylate NS5A, initially at S225 and subsequently at S232 and S235. Our discovery reveals intricate requirements of the structural integrity of NS3 for NS5A hyperphosphorylation and HCV replication.


Asunto(s)
Hepacivirus , Hepatitis C , ARN Polimerasa Dependiente del ARN , Proteínas no Estructurales Virales , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Caseína Quinasa Ialfa/metabolismo , Hepacivirus/enzimología , Hepacivirus/genética , Hepatitis C/virología , Humanos , Fosforilación , Poliproteínas/metabolismo , Dominios Proteicos/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
9.
Cell Mol Life Sci ; 79(3): 184, 2022 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-35279748

RESUMEN

The pathogenesis of acute kidney injury (AKI) is associated with the activation of multiple signaling pathways, including Wnt/ß-catenin signaling. However, the mechanism of Wnt/ß-catenin pathway activation in renal interstitial fibroblasts during AKI is unclear. S100 calcium-binding protein A16 (S100A16), a new member of calcium-binding protein S100 family, is a multi-functional signaling factor involved in various pathogenies, including tumors, glycolipid metabolism disorder, and chronic kidney disease (CKD). We investigated the potential participation of S100A16 in Wnt/ß-catenin pathway activation during AKI by subjecting wild-type (WT) and S100A16 knockout (S100A16+/-) mice to the ischemia-reperfusion injury (IRI), and revealed S100A16 upregulation in this model, in which knockout of S100A16 impeded the Wnt/ß-catenin signaling pathway activation and recovered the expression of downstream hepatocyte growth factor (HGF). We also found that S100A16 was highly expressed in Platelet-derived growth factor receptor beta (PDGFRß) positive renal fibroblasts in vivo. Consistently, in rat renal interstitial fibroblasts (NRK-49F cells), both hypoxia/reoxygenation and S100A16 overexpression exacerbated fibroblasts apoptosis and inhibited HGF secretion; whereas S100A16 knockdown or Wnt/ß-catenin pathway inhibitor ICG-001 reversed these changes. Mechanistically, we showed that S100A16 promoted Wnt/ß-catenin signaling activation via the ubiquitylation and degradation of ß-catenin complex members, glycogen synthase kinase 3ß (GSK3ß) and casein kinase 1α (CK1α), mediated by E3 ubiquitin ligase, the HMG-CoA reductase degradation protein 1 (HRD1). Our study identified the S100A16 as a key regulator in the activation of Wnt/ß-catenin signaling pathway in AKI.


Asunto(s)
Lesión Renal Aguda/patología , Caseína Quinasa Ialfa/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas S100/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Lesión Renal Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/metabolismo , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Proteínas S100/antagonistas & inhibidores , Proteínas S100/deficiencia , Proteínas S100/genética , Ubiquitinación , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , Proteína X Asociada a bcl-2/metabolismo
10.
Xenobiotica ; 53(5): 445-453, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37590011

RESUMEN

1. Deguelin (DGN), a retinoid isolated from many plants, exhibits a potent anticancer activity against a wide spectrum of tumour cells. There is a dearth of evidence, however, regarding the toxicity of DGN to red blood cells (RBCs). This is relevant given the prevalent chemotherapy-associated anaemia observed in cancer patients.2. RBCs were exposed to 1-100 µM of DGN for 24 h at 37 °C. Haemolysis and related markers were photometrically measured while flow cytometry was employed to detect phosphatidylserine exposure through Annexin-V-FITC binding and light scatter properties. Additionally, cytosolic Ca2+ and reactive oxygen species were quantified using Fluo4/AM and H2DCFDA, respectively. DGN was also tested against specific signalling inhibitors in addition to vitamin C and ATP.3. DGN caused a significant increase in Annexin-V-positive cells which was accompanied by cell shrinkage without Ca2+ elevation or oxidative stress. DGN also elicited dose-responsive haemolysis which was ameliorated by preventing KCl efflux and in the presence of sucrose, D4476, and ATP. In whole blood, DGN significantly reduced the reticulocyte count and increased platelet distribution width and large cell count.4. DGN triggers premature RBC eryptosis and haemolysis through casein kinase 1α and ATP depletion, and exhibits a specific toxicity towards reticulocytes and platelets.


Asunto(s)
Caseína Quinasa Ialfa , Humanos , Caseína Quinasa Ialfa/metabolismo , Hemólisis , Eritrocitos/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo
11.
Proc Natl Acad Sci U S A ; 117(4): 1962-1970, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31932442

RESUMEN

Necroptosis is a regulated necrotic cell death pathway, mediated by a supermolecular complex called the necrosome, which contains receptor-interacting protein kinase 1 and 3 (RIPK1, RIPK3) and mixed-lineage kinase domain-like protein (MLKL). Phosphorylation of human RIPK3 at serine 227 (S227) has been shown to be required for downstream MLKL binding and necroptosis progression. Tandem immunoprecipitation of RIPK3 reveals that casein kinase 1 (CK1) family proteins associate with the necrosome upon necroptosis induction, and this interaction depends on the kinase activity of RIPK3. In addition, CK1 proteins colocalize with RIPK3 puncta during necroptosis. Importantly, CK1 proteins directly phosphorylate RIPK3 at S227 in vitro and in vivo. Loss of CK1 proteins abolishes S227 phosphorylation and blocks necroptosis. Furthermore, a RIPK3 mutant with mutations in the CK1 recognition motif fails to be phosphorylated at S227, does not bind or phosphorylate MLKL, and is unable to activate necroptosis. These results strongly suggest that CK1 proteins are necrosome components which are responsible for RIPK3-S227 phosphorylation.


Asunto(s)
Caseína Cinasa 1 épsilon/metabolismo , Caseína Quinasa Ialfa/metabolismo , Quinasa Idelta de la Caseína/metabolismo , Necroptosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Serina/metabolismo , Caseína Cinasa 1 épsilon/genética , Caseína Quinasa Ialfa/genética , Quinasa Idelta de la Caseína/genética , Células HeLa , Humanos , Fosforilación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Serina/genética
12.
Development ; 146(20)2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31511254

RESUMEN

Skin wound repair is essential to restore barrier function and prevent infection after tissue damage. Wound-edge epidermal cells migrate as a sheet to close the wound. However, it is still unclear how cell-cell junctions are regulated during wound closure (WC). To study this, we examined adherens junctions during WC in Drosophila larvae. ß-Catenin is reduced at the lateral cell-cell junctions of wound-edge epidermal cells in the early healing stages. Destruction complex components, including Ck1α, GSK3ß and ß-TrCP, suppress ß-catenin levels in the larval epidermis. Tissue-specific RNAi targeting these genes also caused severe WC defects. The Ck1αRNAi -induced WC defect is related to adherens junctions because loss of either ß-catenin or E-cadherin significantly rescued this WC defect. In contrast, TCFRNAi does not rescue the Ck1αRNAi -induced WC defect, suggesting that Wnt signaling is not related to this defect. Direct overexpression of ß-catenin recapitulates most of the features of Ck1α reduction during wounding. Finally, loss of Ck1α also blocked junctional E-cadherin reduction around the wound. Our results suggest that Ck1α and the destruction complex locally regulate cell adhesion to facilitate efficient wound repair.


Asunto(s)
Uniones Adherentes/metabolismo , Caseína Quinasa Ialfa/metabolismo , Larva/metabolismo , beta Catenina/metabolismo , Animales , Cadherinas/metabolismo , Drosophila , Células Epidérmicas/metabolismo , Epidermis/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo
13.
Nature ; 532(7597): 127-30, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26909574

RESUMEN

Thalidomide and its derivatives, lenalidomide and pomalidomide, are immune modulatory drugs (IMiDs) used in the treatment of haematologic malignancies. IMiDs bind CRBN, the substrate receptor of the CUL4-RBX1-DDB1-CRBN (also known as CRL4(CRBN)) E3 ubiquitin ligase, and inhibit ubiquitination of endogenous CRL4(CRBN) substrates. Unexpectedly, IMiDs also repurpose the ligase to target new proteins for degradation. Lenalidomide induces degradation of the lymphoid transcription factors Ikaros and Aiolos (also known as IKZF1 and IKZF3), and casein kinase 1α (CK1α), which contributes to its clinical efficacy in the treatment of multiple myeloma and 5q-deletion associated myelodysplastic syndrome (del(5q) MDS), respectively. How lenalidomide alters the specificity of the ligase to degrade these proteins remains elusive. Here we present the 2.45 Å crystal structure of DDB1-CRBN bound to lenalidomide and CK1α. CRBN and lenalidomide jointly provide the binding interface for a CK1α ß-hairpin-loop located in the kinase N-lobe. We show that CK1α binding to CRL4(CRBN) is strictly dependent on the presence of an IMiD. Binding of IKZF1 to CRBN similarly requires the compound and both, IKZF1 and CK1α, use a related binding mode. Our study provides a mechanistic explanation for the selective efficacy of lenalidomide in del(5q) MDS therapy. We anticipate that high-affinity protein-protein interactions induced by small molecules will provide opportunities for drug development, particularly for targeted protein degradation.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligasas/metabolismo , Sitios de Unión/efectos de los fármacos , Caseína Quinasa Ialfa/química , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Factor de Transcripción Ikaros/química , Factor de Transcripción Ikaros/metabolismo , Lenalidomida , Modelos Moleculares , Unión Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos , Talidomida/química , Talidomida/metabolismo , Talidomida/farmacología , Ubiquitina-Proteína Ligasas/química , Ubiquitinación/efectos de los fármacos
14.
J Cell Mol Med ; 25(15): 7395-7406, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34216174

RESUMEN

Glioblastoma multiforme (GBM), a fatal brain tumour with no available targeted therapies, has a poor prognosis. At present, radiotherapy is one of the main methods to treat glioma, but it leads to an obvious increase in inflammatory factors in the tumour microenvironment, especially IL-6 and CXCL1, which plays a role in tumour to resistance radiotherapy and tumorigenesis. Casein kinase 1 alpha 1 (CK1α) (encoded on chromosome 5q by Csnk1a1) is considered an attractive target for Tp53 wild-type acute myeloid leukaemia (AML) treatment. In this study, we evaluated the anti-tumour effect of Csnk1a1 suppression in GBM cells in vitro and in vivo. We found that down-regulation of Csnk1a1 or inhibition by D4476, a Csnk1a1 inhibitor, reduced GBM cell proliferation efficiently in both Tp53 wild-type and Tp53-mutant GBM cells. On the contrary, overexpression of Csnk1a1 promoted cell proliferation and colony formation. Csnk1a1 inhibition improved the sensitivity to radiotherapy. Furthermore, down-regulation of Csnk1a1 reduced the production and secretion of pro-inflammatory factors. In the preclinical GBM model, treatment with D4476 significantly inhibited the increase in pro-inflammatory factors caused by radiotherapy and improved radiotherapy sensitivity, thus inhibiting tumour growth and prolonging animal survival time. These results suggest targeting Csnk1a1 exert an anti-tumour role as an inhibitor of inflammatory factors, providing a new strategy for the treatment of glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Caseína Quinasa Ialfa/metabolismo , Glioma/metabolismo , Tolerancia a Radiación , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Caseína Quinasa Ialfa/antagonistas & inhibidores , Caseína Quinasa Ialfa/genética , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Glioma/patología , Glioma/radioterapia , Humanos , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína p53 Supresora de Tumor/genética
15.
Biochem J ; 477(18): 3583-3598, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32686824

RESUMEN

Estrogen sulfotransferase (SULT1E1) metabolically inactivates estrogen and SULT1E1 expression is tightly regulated by multiple nuclear receptors. Human fetal, but not adult, livers express appreciable amounts of SULT1E1 protein, which is mimicked in human hepatoma-derived HepG2 cells cultured in high glucose (450 mg/dl) medium. Here, we have investigated this glucose signal that leads to phosphorylation of nuclear receptor RORα (NR1F1) at Ser100 and the transcription mechanism by which phosphorylated RORα transduces this signal to nuclear receptor HNF4α, activating the SULT1E1 promoter. The promoter is repressed by non-phosphorylated RORα which binds a distal enhancer (-943/-922 bp) and interacts with and represses HNF4α-mediated transcription. In response to high glucose, RORα becomes phosphorylated at Ser100 and reverses its repression of HNF4α promoter activation. Moreover, the casein kinase CK1α, which is identified in an enhancer-bound nuclear protein complex, phosphorylates Ser100 in in vitro kinase assays. During these dynamic processes, both RORα and HNF4α remain on the enhancer. Thus, RORα utilizes phosphorylation to integrate HNF4α and transduces the glucose signal to regulate the SULT1E1 gene in HepG2 cells and this phosphorylation-mediated mechanism may also regulate SULT1E1 expressions in the human liver.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Estrógenos/metabolismo , Glucosa/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Transducción de Señal , Sulfotransferasas/metabolismo , Animales , Células COS , Caseína Quinasa Ialfa/genética , Chlorocebus aethiops , Estrógenos/genética , Glucosa/genética , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Fosforilación , Sulfotransferasas/genética
16.
Proc Natl Acad Sci U S A ; 115(51): E12015-E12023, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30509975

RESUMEN

The rotavirus (RV) genome is replicated and packaged into virus progeny in cytoplasmic inclusions called viroplasms, which require interactions between RV nonstructural proteins NSP2 and NSP5. How viroplasms form remains unknown. We previously found two forms of NSP2 in RV-infected cells: a cytoplasmically dispersed dNSP2, which interacts with hypophosphorylated NSP5; and a viroplasm-specific vNSP2, which interacts with hyperphosphorylated NSP5. Other studies report that CK1α, a ubiquitous cellular kinase, hyperphosphorylates NSP5, but requires NSP2 for reasons that are unclear. Here we show that silencing CK1α in cells before RV infection resulted in (i) >90% decrease in RV replication, (ii) disrupted vNSP2 and NSP5 interaction, (iii) dispersion of vNSP2 throughout the cytoplasm, and (iv) reduced vNSP2 protein levels. Together, these data indicate that CK1α directly affects NSP2. Accordingly, an in vitro kinase assay showed that CK1α phosphorylates serine 313 of NSP2 and triggers NSP2 octamers to form a lattice structure as demonstrated by crystallographic analysis. Additionally, a dual-specificity autokinase activity for NSP2 was identified and confirmed by mass spectrometry. Together, our studies show that phosphorylation of NSP2 involving CK1α controls viroplasm assembly. Considering that CK1α plays a role in the replication of other RNA viruses, similar phosphorylation-dependent mechanisms may exist for other virus pathogens that require cytoplasmic virus factories for replication.


Asunto(s)
Replicación del ADN/fisiología , Proteínas de Unión al ARN/metabolismo , Rotavirus/genética , Rotavirus/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología , Animales , Caseína Quinasa Ialfa/genética , Caseína Quinasa Ialfa/metabolismo , Línea Celular , Cristalografía por Rayos X , Citoplasma/metabolismo , Citoplasma/virología , Silenciador del Gen , Humanos , Cuerpos de Inclusión/metabolismo , Ratones , Modelos Moleculares , Fosforilación , Fosfotransferasas/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/genética , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/metabolismo , Proteínas no Estructurales Virales/genética
17.
Int J Mol Sci ; 22(7)2021 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-33918307

RESUMEN

Disturbance of protein kinase activity may result in dramatic consequences that often lead to cancer development and progression. In tumors of blood origin, both tyrosine kinases and serine/threonine kinases are altered by different types of mutations, critically regulating cancer hallmarks. CK1α and CK2 are highly conserved, ubiquitously expressed and constitutively active pleiotropic kinases, which participate in multiple biological processes. The involvement of these kinases in solid and blood cancers is well documented. CK1α and CK2 are overactive in multiple myeloma, leukemias and lymphomas. Intriguingly, they are not required to the same degree for the viability of normal cells, corroborating the idea of "druggable" kinases. Different to other kinases, mutations on the gene encoding CK1α and CK2 are rare or not reported. Actually, these two kinases are outside the paradigm of oncogene addiction, since cancer cells' dependency on these proteins resembles the phenomenon of "non-oncogene" addiction. In this review, we will summarize the general features of CK1α and CK2 and the most relevant oncogenic and stress-related signaling nodes, regulated by kinase phosphorylation, that may lead to tumor progression. Finally, we will report the current data, which support the positioning of these two kinases in the therapeutic scene of hematological cancers.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Caseína Quinasa Ialfa/metabolismo , Neoplasias Hematológicas/metabolismo , Animales , Quinasa de la Caseína II/antagonistas & inhibidores , Caseína Quinasa Ialfa/antagonistas & inhibidores , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Terapia Molecular Dirigida , Estrés Fisiológico
18.
J Struct Biol ; 212(2): 107602, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32798656

RESUMEN

DEPTOR is an inhibitor of the mTOR kinase which controls cell growth. DEPTOR consists of two DEP domains and a PDZ domain connected by an unstructured linker, and its stability is tightly regulated through post-translational modifications of its linker region that contains the 286SSGYFS291 degron. Based on the mTORC1 complex, our modelling suggests a possible spatial arrangement of DEPTOR which is characterised to form a dimer. Our model shows that the two PDZ domains of a DEPTOR dimer bind separately to the dimeric mTOR's FAT domains ~130 Å apart, while each of the two extended linkers is sufficiently long to span from the FAT domain to the kinase domain of mTOR and beyond to join a shared dimer of the DEP domains. This places the linker's S299 closest to the kinase's catalytic site, indicating that phosphorylation would start with it and successively upstream towards DEPTOR's degron. The CK1α kinase is reportedly responsible for the phosphorylation of the degron, and our docking analysis further reveals that CK1α contains sites to bind DEPTOR's pS286, pS287 and pT295, which may act as priming phosphates for the phosphorylation of the degron's S291. DEPTOR's linker can also be ubiquitylated by the UbcH5A-SCFß-TrCP complex without its PDZ dissociating from mTOR according to the modelling. As the catalytic cleft of mTOR's kinase is restricted, interactions between the kinase's unstructured segment surrounding the cleft and DEPTOR's linker, which may involve S293 and S299, may be critical to controlling DEPTOR's access to the catalytic cleft and hence its phosphorylation by mTOR in a manner dependent on mTOR's activation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Caseína Quinasa Ialfa/metabolismo , Simulación por Computador , Humanos , Fosforilación/fisiología , Dominios Proteicos/fisiología
19.
Hum Mol Genet ; 27(2): 373-384, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29177465

RESUMEN

Attempts have been made to treat nonsense-associated genetic disorders by chemical agents and hence an improved mechanistic insight into the decoding of readthrough signals is essential for the identification and characterisation of factors for the treatment of these disorders. To identify either novel compounds or genes that modulate translation readthrough, we have employed dual reporter-based high-throughput screens that use enzymatic and fluorescence activities and screened bioactive National Institute of Neurological Disease Syndrome (NINDS) compounds (n = 1000) and siRNA (n = 288) libraries. Whilst siRNAs targeting kinases such as CSNK1G3 and NME3 negatively regulate readthrough, neither the bioactive NINDS compounds nor PTC124 promote readthrough. Of note, PTC124 has previously been shown to promote readthrough. Furthermore, the impacts of G418 on the components of eukaryotic selenocysteine incorporation machinery have also been investigated. The selenocysteine machinery decodes the stop codon UGA specifying selenocysteine in natural selenoprotein genes. We have found that the eukaryotic SelC gene promotes the selenocysteine insertion sequence (SECIS)-mediated readthrough but inhibits the readthrough activity induced by G418. We have previously reported that SECIS-mediated readthrough at UGA codons follows a non-processive mechanism. Here, we show that G418-mediated promotion of readthrough also occurs through a non-processive mechanism which competes with translation termination. Based on our observations, we suggest that proteins generated through a non-processive mechanism may be therapeutically beneficial for the resolution of nonsense-associated genetic disorders.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Aminoglicósidos/metabolismo , Secuencia de Bases , Caseína Quinasa Ialfa/metabolismo , Codón sin Sentido , Codón de Terminación , Humanos , Nucleósido Difosfato Quinasas NM23/metabolismo , Conformación de Ácido Nucleico , Oxadiazoles/farmacología , Terminación de la Cadena Péptídica Traduccional , Biosíntesis de Proteínas , Inhibidores de la Síntesis de la Proteína , ARN Mensajero/genética , ARN Interferente Pequeño/genética
20.
EMBO Rep ; 19(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29514862

RESUMEN

The BMP and Wnt signalling pathways determine axis specification during embryonic development. Our previous work has shown that PAWS1 (also known as FAM83G) interacts with SMAD1 and modulates BMP signalling. Here, surprisingly, we show that overexpression of PAWS1 in Xenopus embryos activates Wnt signalling and causes complete axis duplication. Consistent with these observations in Xenopus, Wnt signalling is diminished in U2OS osteosarcoma cells lacking PAWS1, while BMP signalling is unaffected. We show that PAWS1 interacts and co-localises with the α isoform of casein kinase 1 (CK1), and that PAWS1 mutations incapable of binding CK1 fail both to activate Wnt signalling and to elicit axis duplication in Xenopus embryos.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Vía de Señalización Wnt , Animales , Proteína Axina/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Tumoral , Núcleo Celular , Expresión Génica Ectópica , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Complejos Multiproteicos/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA