Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
BMC Biol ; 22(1): 176, 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39183304

RESUMEN

BACKGROUND: Casein kinase 1α (CK1α), expressed in both ovarian germ and somatic cells, is involved in the initial meiosis and primordial follicle formation of mouse oocytes. Using in vitro and in vivo experiments in this study, we explored the function and mechanism of CK1α in estrogen synthesis in mice ovarian granulosa cells. METHODS: A CK1α knockout (cKO) mouse model, targeted specifically to ovarian granulosa cells (GCs), was employed to establish the influence of CK1α on in vivo estrogen synthesis. The influence of CK1α deficiency on GCs was determined in vivo and in vitro by immunofluorescence analysis and Western blot assay. Transcriptome profiling, differentially expressed genes and gene functional enrichment analyses, and computation protein-protein docking, were further employed to assess the CK1α pathway. Furthermore, wild-type female mice were treated with the CK1α antagonist D4476 to elucidate the CK1α's role in estrogen regulation. RESULTS: Ovarian GCs CK1α deficiency impaired fertility and superovulation of female mice; also, the average litter size and the estradiol (E2) level in the serum of cKO female mice were decreased by 57.3% and 87.4% vs. control mice, respectively. This deficiency disrupted the estrous cycle and enhanced the apoptosis in the GCs. We observed that CK1α mediated the secretion of estradiol in mouse ovarian GCs via the cytochrome P450 subfamily 19 member 1 (CYP19A1). CONCLUSIONS: These findings improve the existing understanding of the regulation mechanism of female reproduction and estrogen synthesis. TRIAL REGISTRATION: Not applicable.


Asunto(s)
Aromatasa , Estradiol , Células de la Granulosa , Ratones Noqueados , Animales , Femenino , Ratones , Aromatasa/metabolismo , Aromatasa/genética , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/genética , Estradiol/metabolismo , Células de la Granulosa/metabolismo
2.
Open Biol ; 14(7): 240075, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39043225

RESUMEN

Palmoplantar keratoderma (PPK) is a multi-faceted skin disorder characterized by the thickening of the epidermis and abrasions on the palms and soles of the feet. Among the genetic causes, biallelic pathogenic variants in the FAM83G gene have been associated with PPK in dogs and humans. Here, a novel homozygous variant (c.794G>C, p.Arg265Pro) in the FAM83G gene, identified by whole exome sequencing in a 60-year-old female patient with PPK, is reported. The patient exhibited alterations in the skin of both hands and feet, dystrophic nails, thin, curly and sparse hair, long upper eyelid eyelashes, and poor dental enamel. FAM83G activates WNT signalling through association with ser/thr protein kinase CK1α. When expressed in FAM83G-/- DLD1 colorectal cancer cells, the FAM83GR265P variant displayed poor stability, a loss of interaction with CK1α and attenuated WNT signalling response. These defects persisted in skin fibroblast cells derived from the patient. Our findings imply that the loss of FAM83G-CK1α interaction and subsequent attenuation of WNT signalling underlie the pathogenesis of PPK caused by the FAM83GR265P variant.


Asunto(s)
Caseína Quinasa Ialfa , Queratodermia Palmoplantar , Vía de Señalización Wnt , Humanos , Femenino , Queratodermia Palmoplantar/genética , Queratodermia Palmoplantar/patología , Persona de Mediana Edad , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/genética , Secuenciación del Exoma , Unión Proteica , Fibroblastos/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 , Animales , Humanos , Ratones , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/genética , Línea Celular Tumoral , Proliferación Celular , Células HCT116 , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología
4.
mBio ; 15(8): e0111724, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38940554

RESUMEN

Merkel cell polyomavirus (MCPyV) is a double-stranded tumor virus that is the main causative agent of Merkel cell carcinoma (MCC). The MCPyV large T antigen (LT), an essential viral DNA replication protein, maintains viral persistence by interacting with host Skp1-Cullin 1-F-box (SCF) E3 ubiquitin ligase complexes, which subsequently induces LT's proteasomal degradation, restricting MCPyV DNA replication. SCF E3 ubiquitin ligases require their substrates to be phosphorylated to bind them, utilizing phosphorylated serine residues as docking sites. The MCPyV LT unique region (MUR) is highly phosphorylated and plays a role in multiple host protein interactions, including SCF E3 ubiquitin ligases. Therefore, this domain highly governs LT stability. Though much work has been conducted to identify host factors that restrict MCPyV LT protein expression, the kinase(s) that cooperates with the SCF E3 ligase remains unknown. Here, we demonstrate that casein kinase 1 alpha (CK1α) negatively regulates MCPyV LT stability and LT-mediated replication by modulating interactions with the SCF ß-TrCP. Specifically, we show that numerous CK1 isoforms (α, δ, ε) localize in close proximity to MCPyV LT through in situ proximity ligation assays (PLA) and CK1α overexpression mainly resulted in decreased MCPyV LT protein expression. Inhibition of CK1α using short hairpin RNA (shRNA) and treatment of a CK1α inhibitor or an mTOR inhibitor, TORKinib, resulted in decreased ß-TrCP interaction with LT, increased LT expression, and enhanced MCPyV replication. The expression level of the CSNK1A1 gene transcripts is higher in MCPyV-positive MCC, suggesting a vital role of CK1α in limiting MCPyV replication required for establishing persistent infection. IMPORTANCE: Merkel cell polyomavirus (MCPyV) large tumor antigen is a polyphosphoprotein and the phosphorylation event is required to modulate various functions of LT, including viral replication. Therefore, cellular kinase pathways are indispensable for governing MCPyV polyomavirus infection and life cycle in coordinating with the immunosuppression environment at disease onset. Understanding the regulation mechanisms of MCPyV replication by viral and cellular factors will guide proper prevention strategies with targeted inhibitors for MCPyV-associated Merkel cell carcinoma (MCC) patients, who currently lack therapies.


Asunto(s)
Antígenos Virales de Tumores , Caseína Quinasa Ialfa , Poliomavirus de Células de Merkel , Proteínas con Repetición de beta-Transducina , Poliomavirus de Células de Merkel/genética , Poliomavirus de Células de Merkel/metabolismo , Humanos , Fosforilación , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/genética , Proteínas con Repetición de beta-Transducina/metabolismo , Proteínas con Repetición de beta-Transducina/genética , Antígenos Virales de Tumores/metabolismo , Antígenos Virales de Tumores/genética , Interacciones Huésped-Patógeno , Proteolisis , Replicación Viral , Unión Proteica , Antígenos Transformadores de Poliomavirus/metabolismo , Antígenos Transformadores de Poliomavirus/genética , Infecciones por Polyomavirus/virología , Infecciones por Polyomavirus/metabolismo , Infecciones por Polyomavirus/genética
5.
J Biol Chem ; 300(7): 107407, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38796065

RESUMEN

Members of the casein kinase 1 (CK1) family are important regulators of multiple signaling pathways. CK1α is a well-known negative regulator of the Wnt/ß-catenin pathway, which promotes the degradation of ß-catenin via its phosphorylation of Ser45. In contrast, the closest paralog of CK1α, CK1α-like, is a poorly characterized kinase of unknown function. In this study, we show that the deletion of CK1α, but not CK1α-like, resulted in a strong activation of the Wnt/ß-catenin pathway. Wnt-3a treatment further enhanced the activation, which suggests there are at least two modes, a CK1α-dependent and Wnt-dependent, of ß-catenin regulation. Rescue experiments showed that only two out of ten naturally occurring splice CK1α/α-like variants were able to rescue the augmented Wnt/ß-catenin signaling caused by CK1α deficiency in cells. Importantly, the ability to phosphorylate ß-catenin on Ser45 in the in vitro kinase assay was required but not sufficient for such rescue. Our compound CK1α and GSK3α/ß KO models suggest that the additional nonredundant function of CK1α in the Wnt pathway beyond Ser45-ß-catenin phosphorylation includes Axin phosphorylation. Finally, we established NanoBRET assays for the three most common CK1α splice variants as well as CK1α-like. Target engagement data revealed comparable potency of known CK1α inhibitors for all CK1α variants but not for CK1α-like. In summary, our work brings important novel insights into the biology of CK1α, including evidence for the lack of redundancy with other CK1 kinases in the negative regulation of the Wnt/ß-catenin pathway at the level of ß-catenin and Axin.


Asunto(s)
Caseína Quinasa Ialfa , Vía de Señalización Wnt , beta Catenina , Humanos , Empalme Alternativo , beta Catenina/metabolismo , beta Catenina/genética , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Células HEK293 , Fosforilación , Proteína Wnt3A/metabolismo , Proteína Wnt3A/genética
6.
Antiviral Res ; 226: 105895, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38679165

RESUMEN

Rift Valley fever virus (RVFV) is an arbovirus in the Phenuiviridae family identified initially by the large 'abortion storms' observed among ruminants; RVFV can also infect humans. In humans, there is a wide variation of clinical symptoms ranging from subclinical to mild febrile illness to hepatitis, retinitis, delayed-onset encephalitis, or even hemorrhagic fever. The RVFV is a tri-segmented negative-sense RNA virus consisting of S, M, and L segments. The L segment encodes the RNA-dependent RNA polymerase (RdRp), termed the L protein, which is responsible for both viral mRNA synthesis and genome replication. Phosphorylation of viral RdRps is known to regulate viral replication. This study shows that RVFV L protein is serine phosphorylated and identified Casein Kinase 1 alpha (CK1α) and protein phosphatase 1 alpha (PP1α) as L protein binding partners. Inhibition of CK1 and PP1 through small molecule inhibitor treatment, D4476 and 1E7-03, respectively, caused a change in the phosphorylated status of the L protein. Inhibition of PP1α resulted in increased L protein phosphorylation whereas inhibition of CK1α decreased L protein phosphorylation. It was also found that in RVFV infected cells, PP1α localized to the cytoplasmic compartment. Treatment of RVFV infected cells with CK1 inhibitors reduced virus production in both mammalian and mosquito cells. Lastly, inhibition of either CK1 or PP1 reduced viral genomic RNA levels. These data indicate that L protein is phosphorylated and that CK1 and PP1 play a crucial role in regulating the L protein phosphorylation cycle, which is critical to viral RNA production and viral replication.


Asunto(s)
Proteína Fosfatasa 1 , Virus de la Fiebre del Valle del Rift , Replicación Viral , Virus de la Fiebre del Valle del Rift/fisiología , Virus de la Fiebre del Valle del Rift/genética , Fosforilación , Humanos , Animales , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/genética , Genoma Viral , Proteínas Virales/metabolismo , Proteínas Virales/genética , Caseína Quinasa Ialfa/metabolismo , Caseína Quinasa Ialfa/genética , Chlorocebus aethiops , Línea Celular , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Células Vero , ARN Viral/genética , ARN Viral/metabolismo , Fiebre del Valle del Rift/virología
7.
Endocrinology ; 164(5)2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36929849

RESUMEN

Casein kinase 1α (CK1α) is a main component of the Wnt/ß-catenin signaling pathway, which participates in multiple biological processes. Our recent study demonstrated that CK1α is expressed in both germ cells and somatic cells of mouse testes and regulates spermatogenesis. However, little information is known about the role of CK1α in regulating the development of somatic cells in mouse testes. Our results demonstrated that conditional disruption of CK1α in murine Leydig cells sharply decreased testosterone levels; markedly affected testis development, sperm motility, and sperm morphology; and caused subfertility. The germ cell population was partially decreased in CK1α conditional knockout (cKO) mice, while the proliferation of Leydig cells and Sertoli cells was not affected. Furthermore, in vitro results verified that luteinizing hormone upregulates CK1α through the luteinizing hormone/protein kinase/Epidermal Growth Factor Receptor/extracellular regulated protein kinases/2 signaling pathway and that CK1α interacts with and phosphorylates EGFR, which subsequently activates the phosphorylation of ERK1/2, thereby promoting testosterone synthesis. In addition, high-dose testosterone propionate partially rescued the phenotype observed in cKO mice. This study provides new insights into the role of CK1α in steroidogenesis and male reproduction.


Asunto(s)
Caseína Quinasa Ialfa , Testículo , Ratones , Masculino , Animales , Testículo/metabolismo , Testosterona/metabolismo , Caseína Quinasa Ialfa/genética , Caseína Quinasa Ialfa/metabolismo , Semen/metabolismo , Motilidad Espermática , Células Intersticiales del Testículo/metabolismo , Hormona Luteinizante/metabolismo
8.
Cancer Cell ; 41(4): 726-739.e11, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36898380

RESUMEN

Acute myeloid leukemia (AML) is a hematologic malignancy for which several epigenetic regulators have been identified as therapeutic targets. Here we report the development of cereblon-dependent degraders of IKZF2 and casein kinase 1α (CK1α), termed DEG-35 and DEG-77. We utilized a structure-guided approach to develop DEG-35 as a nanomolar degrader of IKZF2, a hematopoietic-specific transcription factor that contributes to myeloid leukemogenesis. DEG-35 possesses additional substrate specificity for the therapeutically relevant target CK1α, which was identified through unbiased proteomics and a PRISM screen assay. Degradation of IKZF2 and CK1α blocks cell growth and induces myeloid differentiation in AML cells through CK1α-p53- and IKZF2-dependent pathways. Target degradation by DEG-35 or a more soluble analog, DEG-77, delays leukemia progression in murine and human AML mouse models. Overall, we provide a strategy for multitargeted degradation of IKZF2 and CK1α to enhance efficacy against AML that may be expanded to additional targets and indications.


Asunto(s)
Caseína Quinasa Ialfa , Leucemia Mieloide Aguda , Animales , Humanos , Ratones , Caseína Quinasa Ialfa/genética , Caseína Quinasa Ialfa/metabolismo , Hematopoyesis , Factor de Transcripción Ikaros/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Factores de Transcripción
9.
Am J Pathol ; 191(12): 2195-2202, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34809787

RESUMEN

The present study aimed to explore the roles of casein kinase 1α (CK1α) in endometriosis and its underlying mechanisms. Endometrial specimen were collected from the patients and healthy volunteers. The expression patterns of CK1α, phosphatase and tensin homolog (PTEN), and autophagy-related proteins were determined using immunohistochemistry staining, Western blot analysis, and quantitative RT-PCR. Besides, the CK1α-overexpressing cells and PTEN knockdown cells were constructed in the endometrial stromal cells isolated from endometriosis patients. In addition, the cells were transfected with pcDNA3.1-CK1α or pcDNA3.1-CK1α plus siRNA- PTEN. The expressions of CK1α, PTEN, and autophagy-related proteins were determined using Western blot and quantitative RT-PCR. The expressions of CK1α and autophagy-related 7 (Atg7) were significantly decreased in the ectopic endometrium compared with the eutopic endometrium. Spearman rank correlation analysis revealed positive correlations between CK1α and PTEN, CK1α and Atg7, and PTEN and Atg7. In addition, CK1α, PTEN, and autophagy-related proteins were down-regulated in ectopic endometrium. Interestingly, overexpression of CK1α significantly increased the expressions of autophagy-related proteins, whereas the protein expression of autophagy-related proteins was decreased with PTEN knock-down. CK1α regulated PTEN/Atg7-mediated autophagy in endometriosis.


Asunto(s)
Autofagia/fisiología , Caseína Quinasa Ialfa/genética , Endometriosis/genética , Enfermedades Uterinas/genética , Adulto , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/fisiología , Estudios de Casos y Controles , Caseína Quinasa Ialfa/fisiología , Regulación hacia Abajo/genética , Endometriosis/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Fosfohidrolasa PTEN/fisiología , Transducción de Señal/genética , Enfermedades Uterinas/patología , Adulto Joven
10.
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
11.
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
12.
Oncol Rep ; 44(5): 1895-1904, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32901886

RESUMEN

Enhancement of autophagy serves as a promising therapeutic strategy for cancer, including acute myeloid leukemia (AML). Casein kinase 1α (CK1α), encoded by CSNK1A1, regulates Wnt/ß­catenin, p53 and other key signaling pathways, and is critically involved in tumor progression. However, the relationship and mechanism of CK1α with autophagy in AML still remain unclear. In the present study, it was found that AML patients had higher expression of CSNK1A1 mRNA than healthy donors. Furthermore, we analyzed 163 cases of AML patients in the LAML database of TCGA and found that AML patients with high CSNK1A1 had shorter overall survival than those with low or medium CSNK1A1 expression. Furthermore, we demonstrated that CK1α was a negative regulator of autophagy and apoptosis. Pharmacologic inhibition of CK1α using D4476 or CK1α knockdown via lentivirus­mediated shRNA suppressed proliferation and the clone formation by enhancing autophagic flux and apoptosis in AML cell lines as well as in patient blast cells. Intriguingly, D4476­induced cell death was aggravated in combination with an autophagy inhibitor, Spautin­1, suggesting that autophagy may be a pro­survival signaling. CK1α interacted with murine double minute 2 (MDM2) and p53, and CK1α inhibitor D4476 significantly upregulated p53 and phosphorylated 5' AMP­activated protein kinase (AMPK), and substantially inhibited the phosphorylation of mammalian target of rapamycin (mTOR). Our findings indicate that CK1α promotes AML by suppressing p53 downstream of MDM2­mediated autophagy and apoptosis, suggesting that targeting CK1α provides a therapeutic opportunity to treat AML.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Apoptosis/fisiología , Autofagia/fisiología , Benzamidas/farmacología , Caseína Quinasa Ialfa/antagonistas & inhibidores , Caseína Quinasa Ialfa/genética , Línea Celular Tumoral , Humanos , Imidazoles/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
13.
Int J Mol Sci ; 21(16)2020 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-32824859

RESUMEN

Wnt signaling regulates numerous cellular processes during embryonic development and adult tissue homeostasis. Underscoring this physiological importance, deregulation of the Wnt signaling pathway is associated with many disease states, including cancer. Here, we review pivotal regulatory events in the Wnt signaling pathway that drive cancer growth. We then discuss the roles of the established negative Wnt regulator, casein kinase 1α (CK1α), in Wnt signaling. Although the study of CK1α has been ongoing for several decades, the bulk of such research has focused on how it phosphorylates and regulates its various substrates. We focus here on what is known about the mechanisms controlling CK1α, including its putative regulatory proteins and alternative splicing variants. Finally, we describe the discovery and validation of a family of pharmacological CK1α activators capable of inhibiting Wnt pathway activity. One of the important advantages of CK1α activators, relative to other classes of Wnt inhibitors, is their reduced on-target toxicity, overcoming one of the major impediments to developing a clinically relevant Wnt inhibitor. Therefore, we also discuss mechanisms that regulate CK1α steady-state homeostasis, which may contribute to the deregulation of Wnt pathway activity in cancer and underlie the enhanced therapeutic index of CK1α activators.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Neoplasias/metabolismo , Vía de Señalización Wnt , Animales , Antineoplásicos/uso terapéutico , Caseína Quinasa Ialfa/genética , Activadores de Enzimas/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico
14.
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
15.
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
16.
Nat Commun ; 11(1): 1141, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111827

RESUMEN

Osteosarcoma, an aggressive malignant cancer, has a high lung metastasis rate and lacks therapeutic target. Here, we reported that chromobox homolog 4 (CBX4) was overexpressed in osteosarcoma cell lines and tissues. CBX4 promoted metastasis by transcriptionally up-regulating Runx2 via the recruitment of GCN5 to the Runx2 promoter. The phosphorylation of CBX4 at T437 by casein kinase 1α (CK1α) facilitated its ubiquitination at both K178 and K280 and subsequent degradation by CHIP, and this phosphorylation of CBX4 could be reduced by TNFα. Consistently, CK1α suppressed cell migration and invasion through inhibition of CBX4. There was a reverse correlation between CK1α and CBX4 in osteosarcoma tissues, and CK1α was a valuable marker to predict clinical outcomes in osteosarcoma patients with metastasis. Pyrvinium pamoate (PP) as a selective activator of CK1α could inhibit osteosarcoma metastasis via the CK1α/CBX4 axis. Our findings indicate that targeting the CK1α/CBX4 axis may benefit osteosarcoma patients with metastasis.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Ligasas/antagonistas & inhibidores , Ligasas/metabolismo , Osteosarcoma/patología , Proteínas del Grupo Polycomb/antagonistas & inhibidores , Proteínas del Grupo Polycomb/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Caseína Quinasa Ialfa/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ligasas/genética , Ratones , Mutación , Metástasis de la Neoplasia , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/metabolismo , Fosforilación/efectos de los fármacos , Proteínas del Grupo Polycomb/genética , Regiones Promotoras Genéticas , Compuestos de Pirvinio/farmacología , Compuestos de Pirvinio/uso terapéutico , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos , Factores de Transcripción p300-CBP/metabolismo
17.
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
18.
Oncogene ; 39(1): 176-186, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31462704

RESUMEN

Somatic missense mutations of the CSNK1A1 gene encoding casein kinase 1 alpha (CK1α) occur in a subset of myelodysplastic syndrome (MDS) with del(5q) karyotype. The chromosomal deletion causes CSNK1A1 haplo-insufficiency. CK1α mutations have also been observed in a variety of solid and hematopoietic tumors at low frequency. The functional consequence of CK1α mutation remains unknown. Here we show that tumor-associated CK1α mutations exclusively localize to the substrate-binding cleft. Functional analysis of recurrent mutants E98K and D140A revealed enhanced binding to the p53 inhibitor MDMX, increased ability to stimulate MDMX-p53 binding, and increased suppression of p21 expression. Furthermore, E98K and D140A mutants have reduced ability to promote phosphorylation of ß-catenin, resulting in enhanced Wnt signaling. The results suggest that the CK1α mutations observed in tumors cause gain-of-function in cooperating with MDMX and inhibiting p53, and partial loss-of-function in suppressing Wnt signaling. These functional changes may promote expansion of abnormal myeloid progenitors in del(5q) MDS, and in rare cases drive the progression of other tumors.


Asunto(s)
Caseína Quinasa Ialfa/genética , Síndromes Mielodisplásicos/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/genética , Animales , Línea Celular Tumoral , Deleción Cromosómica , Haploinsuficiencia/genética , Xenoinjertos , Humanos , Ratones , Mutación Missense/genética , Síndromes Mielodisplásicos/patología , Fosforilación/genética , Unión Proteica/genética , Vía de Señalización Wnt/genética , beta Catenina/genética
19.
Anticancer Drugs ; 30(7): e0747, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31305293

RESUMEN

Pyrvinium tosylate (PT) is an anthelminthic drug that has recently been shown to suppress various human cancers. However, whether PT is effective in nasopharyngeal carcinoma (NPC) has not been determined to date. In this work, we show the selective efficacy of PT in NPC while sparing normal nasopharyngeal epithelial cells, and its ability to increase chemosensitivity. We show that PT at 100 and 500 nmol/l significantly inhibits growth and induces apoptosis in several NPC cell lines without affecting normal nasopharyngeal epithelial cells. Using cell culture and xenograft mouse models, PT markedly enhances cisplatin's efficacy in NPC and the combination leads to almost complete tumor inhibition. Mechanism studies show that PT suppresses active, nuclear ß-catenin level and activity and increases Axin level in NPC cells. ß-Catenin overexpression completely reverses the inhibitory effects of PT, confirming that ß-catenin is the molecular mechanism of PT's action in NPC. In addition, the effects of PT on ß-catenin and Axin levels and on Wnt signaling in NPC cells are mediated by its activation of casine kinase 1α. Our work is the first to suggest that Wnt/ß-catenin is a selective target for NPC treatment, and provides the preclinical evidence on the translational potential of PT as a useful addition to the treatment armamentarium for NPC.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Compuestos de Pirvinio/farmacología , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis , Caseína Quinasa Ialfa/genética , Proliferación Celular , Cisplatino/farmacología , Quimioterapia Combinada , Humanos , Ratones , Ratones SCID , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Cell Physiol ; 234(8): 14377-14388, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30644102

RESUMEN

Traumatic brain injury (TBI) is a common cause of death and acquired disability in adults and children. Identifying biomarkers for mild TBI (mTBI) that can predict functional impairments on neuropsychiatric and neurocognitive testing after head trauma is yet to be firmly established. Extracellular vesicles (EVs) are known to traffic from the brain to the oral cavity and can be detected in saliva. We hypothesize the genetic profile of salivary EVs in patients who have suffered head trauma will differ from normal healthy controls, thus constituting a unique expression signature for mTBI. We enrolled a total of 54 subjects including for saliva sampling, 23 controls with no history of head traumas, 16 patients enrolled from an outpatient concussion clinic, and 15 patients from the emergency department who had sustained a head trauma within 24 hr. We performed real-time PCR of the salivary EVs of the 54 subjects profiling 96 genes from the TaqMan Human Alzheimer's disease array. Real-time PCR analysis revealed 57 (15 genes, p < 0.05) upregulated genes in emergency department patients and 56 (14 genes, p < 0.05) upregulated genes in concussion clinic patients when compared with controls. Three genes were upregulated in both the emergency department patients and concussion clinic patients: CDC2, CSNK1A1, and CTSD ( p < 0.05). Our results demonstrate that salivary EVs gene expression can serve as a viable source of biomarkers for mTBI. This study shows multiple Alzheimer's disease genes present after an mTBI.


Asunto(s)
Biomarcadores , Lesiones Traumáticas del Encéfalo/genética , Proteína Quinasa CDC2/genética , Caseína Quinasa Ialfa/genética , Catepsina D/genética , Adolescente , Adulto , Anciano , Enfermedad de Alzheimer/genética , Conmoción Encefálica/genética , Conmoción Encefálica/patología , Lesiones Traumáticas del Encéfalo/diagnóstico , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Niño , Servicio de Urgencia en Hospital , Vesículas Extracelulares/genética , Femenino , Regulación de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Saliva/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA