Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(17): e2401716121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38625937

RESUMEN

Serine phosphorylations on insulin receptor substrate 1 (IRS-1) by diverse kinases aoccur widely during obesity-, stress-, and inflammation-induced conditions in models of insulin resistance and type 2 diabetes. In this study, we define a region within the human IRS-1, which is directly C-terminal to the PTB domain encompassing numerous serine phosphorylation sites including Ser307 (mouse Ser302) and Ser312 (mouse 307) creating a phosphorylation insulin resistance (PIR) domain. We demonstrate that the IRS-1 PTB-PIR with its unphosphorylated serine residues interacts with the insulin receptor (IR) but loses the IR-binding when they are phosphorylated. Surface plasmon resonance studies further confirm that the PTB-PIR binds stronger to IR than just the PTB domain, and that phosphorylations at Ser307, Ser312, Ser315, and Ser323 within the PIR domain result in abrogating the binding. Insulin-responsive cells containing the mutant IRS-1 with all these four serines changed into glutamates to mimic phosphorylations show decreased levels of phosphorylations in IR, IRS-1, and AKT compared to the wild-type IRS-1. Hydrogen-deuterium exchange mass spectrometry experiments indicating the PIR domain interacting with the N-terminal lobe and the hinge regions of the IR kinase domain further suggest the possibility that the IRS-1 PIR domain protects the IR from the PTP1B-mediated dephosphorylation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Ratones , Humanos , Animales , Fosforilación , Serina/metabolismo , Receptor de Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Línea Celular , Fosfoproteínas/metabolismo , Insulina/metabolismo
2.
Semin Cancer Biol ; 86(Pt 2): 347-357, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35868515

RESUMEN

Several metabolic pathways for the supply of adenosine triphosphate (ATP) have been proposed; however, the major source of reducing power for ADP in cancer remains unclear. Although glycolysis is the source of ATP in tumors according to the Warburg effect, ATP levels do not differ between cancer cells grown in the presence and absence of glucose. Several theories have been proposed to explain the supply of ATP in cancer, including metabolic reprograming in the tumor microenvironment. However, these theories are based on the production of ATP by the TCA-OxPhos pathway, which is inconsistent with the Warburg effect. We found that blocking fatty acid oxidation (FAO) in the presence of glucose significantly decreased ATP production in various cancer cells. This suggests that cancer cells depend on fatty acids to produce ATP through FAO instead of glycolysis. We observed that cancer cell growth mainly relies on metabolic nutrients and oxygen systemically supplied through the bloodstream instead of metabolic reprogramming. In a spontaneous mouse tumor model (KrasG12D; Pdx1-cre), tumor growth was 2-fold higher in mice fed a high-fat diet (low-carbo diet) that caused obesity, whereas a calorie-balanced, low-fat diet (high-carbo diet) inhibited tumor growth by 3-fold compared with that in mice fed a control/normal diet. This 5-fold difference in tumor growth between mice fed low-fat and high-fat diets suggests that fat-induced obesity promotes cancer growth, and tumor growth depends on fatty acids as the primary source of energy.


Asunto(s)
Ácidos Grasos , Neoplasias , Ratones , Humanos , Animales , Ácidos Grasos/metabolismo , Adenosina Trifosfato/metabolismo , Dieta Alta en Grasa , Obesidad/complicaciones , Obesidad/metabolismo , Glucosa/metabolismo , Neoplasias/etiología , Microambiente Tumoral
3.
EMBO Rep ; 22(6): e51323, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33938112

RESUMEN

In eukaryotic cells, mitochondria are closely tethered to the endoplasmic reticulum (ER) at sites called mitochondria-associated ER membranes (MAMs). Ca2+ ion and phospholipid transfer occurs at MAMs to support diverse cellular functions. Unlike those in yeast, the protein complexes involved in phospholipid transfer at MAMs in humans have not been identified. Here, we determine the crystal structure of the tetratricopeptide repeat domain of PTPIP51 (PTPIP51_TPR), a mitochondrial protein that interacts with the ER-anchored VAPB protein at MAMs. The structure of PTPIP51_TPR shows an archetypal TPR fold, and an electron density map corresponding to an unidentified lipid-like molecule probably derived from the protein expression host is found in the structure. We reveal functions of PTPIP51 in phospholipid binding/transfer, particularly of phosphatidic acid, in vitro. Depletion of PTPIP51 in cells reduces the mitochondrial cardiolipin level. Additionally, we confirm that the PTPIP51-VAPB interaction is mediated by the FFAT-like motif of PTPIP51 and the MSP domain of VAPB. Our findings suggest that PTPIP51 is a phospholipid transfer protein with a MAM-tethering function.


Asunto(s)
Calcio , Fosfolípidos , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fosfolípidos/metabolismo , Proteínas Tirosina Fosfatasas
4.
Int J Mol Sci ; 24(15)2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37569345

RESUMEN

In previous work, we showed that cancer cells do not depend on glycolysis for ATP production, but they do on fatty acid oxidation. However, we found some cancer cells induced cell death after glucose deprivation along with a decrease of ATP production. We investigated the different response of glucose deprivation with two types of cancer cells including glucose insensitive cancer cells (GIC) which do not change ATP levels, and glucose sensitive cancer cells (GSC) which decrease ATP production in 24 h. Glucose deprivation-induced cell death in GSC by more than twofold after 12 h and by up to tenfold after 24 h accompanied by decreased ATP production to compare to the control (cultured in glucose). Glucose deprivation decreased the levels of metabolic intermediates of the pentose phosphate pathway (PPP) and the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) in both GSC and GIC. However, glucose deprivation increased reactive oxygen species (ROS) only in GSC, suggesting that GIC have a higher tolerance for decreased NADPH than GSC. The twofold higher ratio of reduced/oxidized glutathione (GSH/GSSG) in GIS than in GSC correlates closely with the twofold lower ROS levels under glucose starvation conditions. Treatment with N-acetylcysteine (NAC) as a precursor to the biologic antioxidant glutathione restored ATP production by 70% and reversed cell death caused by glucose deprivation in GSC. The present findings suggest that glucose deprivation-induced cancer cell death is not caused by decreased ATP levels, but rather triggered by a failure of ROS regulation by the antioxidant system. Conclusion is clear that glucose deprivation-induced cell death is independent from ATP depletion-induced cell death.


Asunto(s)
Adenosina Trifosfato , Glucosa , Neoplasias , Especies Reactivas de Oxígeno , Glucosa/deficiencia , Adenosina Trifosfato/metabolismo , Vía de Pentosa Fosfato , Especies Reactivas de Oxígeno/metabolismo , NADP/metabolismo , Glutatión/metabolismo , Acetilcisteína/metabolismo , Acetilcisteína/farmacología , Células PC-3 , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Muerte Celular
5.
Nucleic Acids Res ; 48(11): 6340-6352, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32383752

RESUMEN

API5 (APoptosis Inhibitor 5) and nuclear FGF2 (Fibroblast Growth Factor 2) are upregulated in various human cancers and are correlated with poor prognosis. Although their physical interaction has been identified, the function related to the resulting complex is unknown. Here, we determined the crystal structure of the API5-FGF2 complex and identified critical residues driving the protein interaction. These findings provided a structural basis for the nuclear localization of the FGF2 isoform lacking a canonical nuclear localization signal and identified a cryptic nuclear localization sequence in FGF2. The interaction between API5 and FGF2 was important for mRNA nuclear export through both the TREX and eIF4E/LRPPRC mRNA export complexes, thus regulating the export of bulk mRNA and specific mRNAs containing eIF4E sensitivity elements, such as c-MYC and cyclin D1. These data show the newly identified molecular function of API5 and nuclear FGF2, and provide a clue to understanding the dynamic regulation of mRNA export.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Transporte de ARN , ARN Mensajero/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Cristalografía por Rayos X , Ciclina D1/metabolismo , ARN Helicasas DEAD-box/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Humanos , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
6.
Nucleic Acids Res ; 46(13): 6544-6560, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-29901724

RESUMEN

Cyclin-dependent kinase 1 (Cdk1) is indispensable for embryonic stem cell (ESC) maintenance and embryo development. Even though some reports have described a connection between Cdk1 and Oct4, there is no evidence that Cdk1 activity is directly linked to the ESC pluripotency transcription program. We recently reported that Aurkb/PP1-mediated Oct4 resetting is important to cell cycle maintenance and pluripotency in mouse ESCs (mESCs). In this study, we show that Cdk1 is an upstream regulator of the Oct4 phosphorylation state during cell cycle progression, and it coordinates the chromatin associated state of Oct4 for pluripotency-related gene expression within the cell cycle. Upon entry into mitosis, Aurkb in the chromosome passenger complex becomes fully activated and PP1 activity is inhibited downstream of Cdk1 activation, leading to sustaining Oct4(S229) phosphorylation and dissociation of Oct4 from chromatin during the mitotic phase. Cdk1 inhibition at the mitotic phase abnormally results in Oct4 dephosphorylation, chromosome decondensation and chromatin association of Oct4, even in replicated chromosome. Our study results suggest a molecular mechanism by which Cdk1 directly links the cell cycle to the pluripotency transcription program in mESCs.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Ciclo Celular/genética , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Transcripción Genética , Animales , Aurora Quinasa B/metabolismo , Proteína Quinasa CDC2/antagonistas & inhibidores , División Celular/genética , Células Cultivadas , Fase G2/genética , Humanos , Ratones , Fosforilación , Proteína Fosfatasa 1/metabolismo
7.
Int J Mol Sci ; 21(21)2020 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-33114206

RESUMEN

Aberrant tyrosine-protein kinase Mer (MerTK) expression triggers prosurvival signaling and contributes to cell survival, invasive motility, and chemoresistance in many kinds of cancers. In addition, recent reports suggested that MerTK could be a primary target for abnormal platelet aggregation. Consequently, MerTK inhibitors may promote cancer cell death, sensitize cells to chemotherapy, and act as new antiplatelet agents. We screened an inhouse chemical library to discover novel small-molecule MerTK inhibitors, and identified AZD7762, which is known as a checkpoint-kinase (Chk) inhibitor. The inhibition of MerTK by AZD7762 was validated using an in vitro homogeneous time-resolved fluorescence (HTRF) assay and through monitoring the decrease in phosphorylated MerTK in two lung cancer cell lines. We also determined the crystal structure of the MerTK:AZD7762 complex and revealed the binding mode of AZD7762 to MerTK. Structural information from the MerTK:AZD7762 complex and its comparison with other MerTK:inhibitor structures gave us new insights for optimizing the development of inhibitors targeting MerTK.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Tiofenos/química , Tiofenos/farmacología , Urea/análogos & derivados , Tirosina Quinasa c-Mer/química , Tirosina Quinasa c-Mer/metabolismo , Células A549 , Línea Celular Tumoral , Cristalografía por Rayos X , Regulación hacia Abajo , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Modelos Moleculares , Fosforilación/efectos de los fármacos , Unión Proteica , Conformación Proteica , Dominios Proteicos , Relación Estructura-Actividad , Urea/química , Urea/farmacología
8.
FASEB J ; : fj201700837RRRR, 2018 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-29894668

RESUMEN

Unlike lower organisms, mammals have 2 C-terminal binding protein (Ctbp) isoforms, Ctbp1 and Ctbp2. Ctbp2 is revealed as a key factor involved in determining cell fate decisions by regulating the epigenetic state in active embryonic stem cell (ESC) genes. However, the molecular mechanism underlying how Ctbp1 and Ctbp2 have different roles remains elusive. Here we demonstrate that Ctbp isoform abundance is important for mouse embryonic ESCs (mESCs) to exit from pluripotency. Temporal expression patterns of Ctbp isoforms were quite different; Ctbp2 is more highly expressed in mESCs and decreases during differentiation, while Ctbp1 is constantly expressed at a lower level. Ctbp2 knockdown, but not Ctbp1 knockdown, in mESCs resulted in impaired exit from pluripotency. Interestingly, Ctbp1 and Ctbp2 overexpression in Ctbp2-knockdown mESCs leads to exiting from pluripotency in a manner similar to that of wild-type mESCs. Quantification of Ctbp1 and Ctbp2 revealed that differentiation ability correlates with abundance of Ctbp isoform in undifferentiated mESCs, suggesting that a sufficient amount of Ctbp isoform is a prerequisite for exiting from pluripotency. The results support the contention that 2 redundant Ctbp isoforms regulate elaborate differentiation via temporally distinctive regulatory patterns in mESCs.-Suh, M. Y., Kim, T. W., Lee, H.-T., Shin, J., Kim, J.-H., Jang, H., Kim, H. J., Kim, S.-T., Cho, E.-J., Youn, H.-D. Abundance of C-terminal binding protein isoform is a prerequisite for exit from pluripotency in mouse embryonic stem cells.

9.
Biochem Biophys Res Commun ; 503(3): 1980-1986, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30078675

RESUMEN

Germ cell tumours (GCTs) are one of the most threatening malignancies in young men and women. Although several reports have suggested the importance of OCT4 in human GCTs, its role has not been clearly investigated on a molecular level. In this study, we revealed GCT-specific direct transcriptional target genes of OCT4. Conditional knockdown of OCT4 in GCT cell lines reduced cell proliferation by affecting both cell cycle and death. Knockdown of OCT4 also reduced stemness of GCTs, as assessed by the expression of other stemness factors, alkaline phosphatase staining, and tumour sphere formation ability. Analysis of whole mRNA expression patterns among GCT cells harbouring endogenous, depleted, and rescued OCT4 revealed 1133 OCT4 target genes in GCT. Combined analysis of both the chromatin binding signature of OCT4 and the genes whose expression levels were changed by OCT4 revealed 258 direct target genes of OCT4 in GCTs. In a similar way, 594 direct target genes in normal embryonic stem cells (ESCs) were identified. Among these two sets of OCT4 direct target genes, 38 genes were common between GCTs and ESCs, most of which were related to regulation of pluripotency, and 220 genes were specific to GCTs, most of which were related to focal adhesion and extracellular matrix organisation. These results provide a molecular basis for how OCT4 regulates GCT stemness and will aid our understanding of the role of OCT4 in other cancers.


Asunto(s)
Matriz Extracelular/genética , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/patología , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosfatasa Alcalina/análisis , Fosfatasa Alcalina/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Doxiciclina/farmacología , Citometría de Flujo , Redes Reguladoras de Genes/genética , Humanos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores , Factor 3 de Transcripción de Unión a Octámeros/genética , Transcripción Genética/genética
10.
Nucleic Acids Res ; 43(9): 4505-16, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25813038

RESUMEN

Post-translational modifications of core histones affect various cellular processes, primarily through transcription. However, their relationship with the termination of transcription has remained largely unknown. In this study, we show that DNA damage-activated AKT phosphorylates threonine 45 of core histone H3 (H3-T45). By genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) analysis, H3-T45 phosphorylation was distributed throughout DNA damage-responsive gene loci, particularly immediately after the transcription termination site. H3-T45 phosphorylation pattern showed close-resemblance to that of RNA polymerase II C-terminal domain (CTD) serine 2 phosphorylation, which establishes the transcription termination signal. AKT1 was more effective than AKT2 in phosphorylating H3-T45. Blocking H3-T45 phosphorylation by inhibiting AKT or through amino acid substitution limited RNA decay downstream of mRNA cleavage sites and decreased RNA polymerase II release from chromatin. Our findings suggest that AKT-mediated phosphorylation of H3-T45 regulates the processing of the 3' end of DNA damage-activated genes to facilitate transcriptional termination.


Asunto(s)
Daño del ADN , Histonas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Terminación de la Transcripción Genética , Línea Celular , Células HeLa , Histonas/química , Humanos , Células MCF-7 , Fosforilación , Treonina/metabolismo , Sitio de Iniciación de la Transcripción
11.
Stem Cells ; 33(9): 2699-711, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26059508

RESUMEN

Pluripotent stem cells (PSCs) have distinct metabolic properties that support their metabolic and energetic needs and affect their stemness. In particular, high glycolysis is critical for the generation and maintenance of PSCs. However, it is unknown how PSCs maintain and acquire this metabolic signature. In this study, we found that core pluripotency factors regulate glycolysis directly by controlling the expression of glycolytic enzymes. Specifically, Oct4 directly governs Hk2 and Pkm2, which are important glycolytic enzymes that determine the rate of glycolytic flux. The overexpression of Hk2 and Pkm2 sustains high levels of glycolysis during embryonic stem cell (ESC) differentiation. Moreover, the maintenance of high glycolysis levels by Hk2 and Pkm2 overexpression hampers differentiation and preserves the pluripotency of ESCs in the absence of leukemia inhibitory factor. Overall, our study identifies a direct molecular connection between core pluripotency factors and ESC metabolic signatures and demonstrates the significance of metabolism in cell fate determination.


Asunto(s)
Proteínas Portadoras/biosíntesis , Células Madre Embrionarias/metabolismo , Glucólisis/fisiología , Hexoquinasa/biosíntesis , Proteínas de la Membrana/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Células Madre Pluripotentes/metabolismo , Hormonas Tiroideas/biosíntesis , Animales , Diferenciación Celular/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Unión a Hormona Tiroide
12.
Stem Cells ; 33(8): 2442-55, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25944056

RESUMEN

For cells to exit from pluripotency and commit to a lineage, the circuitry of a core transcription factor (CTF) network must be extinguished in an orderly manner through epigenetic modifications. However, how this choreographed epigenetic remodeling at active embryonic stem cell (ESC) genes occurs during differentiation is poorly understood. In this study, we demonstrate that C-terminal binding protein 2 (Ctbp2) regulates nucleosome remodeling and deacetylation (NuRD)-mediated deacetylation of H3K27 and facilitates recruitment of polycomb repressive complex 2 (PRC2)-mediated H3K27me3 in active ESC genes for exit from pluripotency during differentiation. By genomewide analysis, we found that Ctbp2 resides in active ESC genes and co-occupies regions with ESC CTFs in undifferentiated ESCs. Furthermore, ablation of Ctbp2 effects inappropriate gene silencing in ESCs by sustaining high levels of H3K27ac and impeding H3K27me3 in active ESC genes, thereby sustaining ESC maintenance during differentiation. Thus, Ctbp2 preoccupies regions in active genes with the NuRD complex in undifferentiated ESCs that are directed toward H3K27me3 by PRC2 to induce stable silencing, which is pivotal for natural lineage commitment.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/fisiología , Histonas/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Oxidorreductasas de Alcohol , Animales , Línea Celular , Ensamble y Desensamble de Cromatina/fisiología , Proteínas Co-Represoras , Proteínas de Unión al ADN/genética , Histonas/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Células Madre Embrionarias de Ratones/citología , Nucleosomas/genética , Nucleosomas/metabolismo , Fosfoproteínas/genética , Proteínas Represoras/genética
13.
Nucleic Acids Res ; 42(1): 224-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24078251

RESUMEN

Myocyte enhancer factor 2 (MEF2) is a family of transcription factors that regulates many processes, including muscle differentiation. Due to its many target genes, MEF2D requires tight regulation of transcription activity over time and by location. Epigenetic modifiers have been suggested to regulate MEF2-dependent transcription via modifications to histones and MEF2. However, the modulation of MEF2 activity by lysine methylation, an important posttranslational modification that alters the activities of transcription factors, has not been studied. We report the reversible lysine methylation of MEF2D by G9a and LSD1 as a regulatory mechanism of MEF2D activity and skeletal muscle differentiation. G9a methylates lysine-267 of MEF2D and represses its transcriptional activity, but LSD1 counteracts it. This residue is highly conserved between MEF2 members in mammals. During myogenic differentiation of C2C12 mouse skeletal muscle cells, the methylation of MEF2D by G9a decreased, on which MEF2D-dependent myogenic genes were upregulated. We have also identified lysine-267 as a methylation/demethylation site and demonstrate that the lysine methylation state of MEF2D regulates its transcriptional activity and skeletal muscle cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Lisina/metabolismo , Factores de Transcripción MEF2/metabolismo , Mioblastos Esqueléticos/metabolismo , Animales , Línea Celular , Cromatina/metabolismo , Células HEK293 , Histona Demetilasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Factores de Transcripción MEF2/antagonistas & inhibidores , Factores de Transcripción MEF2/química , Metilación , Ratones , Mioblastos Esqueléticos/citología , Oxidorreductasas N-Desmetilantes/metabolismo , Procesamiento Proteico-Postraduccional , Transcripción Genética
14.
Nucleic Acids Res ; 41(4): 2180-90, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23303793

RESUMEN

CABIN1 acts as a negative regulator of p53 by keeping p53 in an inactive state on chromatin. Genotoxic stress causes rapid dissociation of CABIN1 and activation of p53. However, its molecular mechanism is still unknown. Here, we reveal the phosphorylation- and ubiquitination-dependent degradation of CABIN1 upon DNA damage, releasing p53 for transcriptional activation. The DNA-damage-signaling kinases, ATM and CHK2, phosphorylate CABIN1 and increase the degradation of CABIN1 protein. Knockdown or overexpression of these kinases influences the stability of CABIN1 protein showing that their activity is critical for degradation of CABIN1. Additionally, CABIN1 was found to undergo ubiquitin-dependent proteasomal degradation mediated by the CRL4DDB2 ubiquitin ligase complex. Both phosphorylation and ubiquitination of CABIN1 appear to be relevant for controlling the level of CABIN1 protein upon genotoxic stress.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Daño del ADN , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2 , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Humanos , Mutágenos/toxicidad , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis , Estrés Fisiológico/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
Pak J Med Sci ; 31(5): 1207-12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26649015

RESUMEN

OBJECTIVES: To study about the blood count of a risk factor related to physical measurement and metabolic syndrome, and the area of epicardial fat for medical checkup patients. METHODS: From April 1(st) to November 15(th) in 2014, we measured the area of epicardial fat in the adult out patients under 60 years of age, who are in good health; and the patients took the blood test and low-dose lung CT. In order to identify the relationship between the area of epicardial fat and the risk factor of metabolic syndrome, we conducted correlation analysis. Then, we performed multiple regression analysis to evaluate an independent correlation of epicardial area. In addition, we computed the cut-off value of epicardial fat area by using ROC (Receiver Operating Characteristic) curve to foresee a metabolic syndrome factor that has the most proper sensitivity and specificity. RESULTS: Waist circumference, fasting blood sugar, triglyceride, high-density lipoprotein (HDL) cholesterol, systolic blood pressure, and diastolic blood pressure were shown to be the factors that affect the area of epicardial fat. Therefore, if waist circumference, fasting blood sugar, triglyceride, systolic blood pressure, and diastolic blood pressure were increased, the area of epicardial fat would be significantly increased (P<0.05); and if high-density lipoprotein cholesterol was increased, the area of epicardial fat would be significantly decreased (P<0.05). Out of metabolic syndrome factors, waist circumference's ROC curve area was 0.79 (Confidence Interval 0.73-0.84, P<0.05), which was the highest. The sensitivity was 83.7% when specificity was 70.1%, which proves that they are important factors for the diagnosis. In brief, metabolic syndrome is a disease that mostly appears in obesity patients, so we should try to monitor and cure the disease. CONCLUSION: The risk factors of metabolic syndrome can be managed through health care, and if we try to decrease the risk factors, we will be able to shrink epicardial fat area and decrease metabolic syndrome at the same time.

16.
J Phys Ther Sci ; 26(8): 1225-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25202186

RESUMEN

[Purpose] The aim of this study was to find biochemical markers related to low bone mineral density in Korean adults. [Subjects and Methods] From August 1 to September 15, 2013, subjects receiving medical checkups were classified as lumbar spine bone normal, osteopenic, or osteoporotic using a bone mineral densitometer. Next, age, body mass index, and biochemical parameter differences were compared among the three groups. [Results] The results revealed that, the relevant factors were maximum blood pressure, minimum blood pressure, bone mineral density, total bilirubin, alkaline phosphatase (ALP), fasting blood glucose, iron, neutrophils, monocytes, and eosinophils. The bone mineral density of patients with osteoporosis was 0.763 times lower than that of normal subjects. The total bilirubin level of patients with osteoporosis was 0.45 times lower than that of normal subjects. The alkaline phosphatase level of patients with osteopenia was 1.059 times higher than that of normal subjects, and that in patients with osteoporosis was 1.088 times higher than that in normal subjects. The fasting blood glucose level of patients with osteoporosis was 0.963 times lower than that of normal subjects. The iron level of patients with osteoporosis was 0.986 times lower than that of normal subjects. [Conclusion] In conclusion, osteoporosis is a representative disease in elderly women due to aging and menopause, and more active interest should be taken for prevention and treatment.

17.
Cells ; 13(3)2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38334608

RESUMEN

Effectively targeting cancer stemness is essential for successful cancer therapy. Recent studies have revealed that SOX2, a pluripotent stem cell factor, significantly contributes to cancer stem cell (CSC)-like characteristics closely associated with cancer malignancy. However, its contradictory impact on patient survival in specific cancer types, including lung adenocarcinoma (LUAD), underscores the need for more comprehensive research to clarify its functional effect on cancer stemness. In this study, we demonstrate that SOX2 is not universally required for the regulation of CSC-like properties in LUAD. We generated SOX2 knockouts in A549, H358, and HCC827 LUAD cells using the CRISPR/Cas9 system. Our results reveal unchanged CSC characteristics, including sustained proliferation, tumor sphere formation, invasion, migration, and therapy resistance, compared to normal cells. Conversely, SOX2 knockdown using conditional shRNA targeting SOX2, significantly reduced CSC traits. However, these loss-of-function effects were not rescued by SOX2 resistant to shRNA, underscoring the potential for SOX2 protein level-independent results in prior siRNA- or shRNA-based research. Ultimately, our findings demonstrate that SOX2 is not absolutely essential in LUAD cancer cells. This emphasizes the necessity of considering cancer subtype-dependent and context-dependent factors when targeting SOX2 overexpression as a potential therapeutic vulnerability in diverse cancers.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Células Madre Neoplásicas , Factores de Transcripción SOXB1 , Humanos , Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
18.
Exp Mol Med ; 54(9): 1450-1460, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36056187

RESUMEN

Myeloid epithelial reproductive proto-oncogene tyrosine kinase (MERTK) plays an essential role in modulating cancer immune tolerance by regulating macrophage efferocytosis. Studies are underway to develop small-molecule chemicals that inhibit MERTK as cancer immunotherapeutic agents, but these efforts are in their early stages. This study identified BMS794833, whose primary targets are MET and VEGFR2, as a potent MERTK inhibitor and developed a real-time efferocytosis monitoring system. The X-ray cocrystal structure revealed that BMS794833 was in contact with the ATP-binding pocket and the allosteric back pocket, rendering MERTK inactive. Homogeneous time-resolved fluorescence kinetic and Western blotting analyses showed that BMS794833 competitively inhibited MERTK activity in vitro and inhibited the autophosphorylation of MERTK in macrophages. We developed a system to monitor MERTK-dependent efferocytosis in real time, and using this system, we confirmed that BMS794833 significantly inhibited the efferocytosis of differentiated macrophages. Finally, BMS794833 significantly inhibited efferocytosis in vivo in a mouse model. These data show that BMS794833 is a type II MERTK inhibitor that regulates macrophage efferocytosis. In addition, the real-time efferocytosis monitoring technology developed in this study has great potential for future applications.


Asunto(s)
Proteínas Tirosina Quinasas , Proteínas Tirosina Quinasas Receptoras , Animales , Ratones , Adenosina Trifosfato/metabolismo , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/metabolismo , Macrófagos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proto-Oncogenes , Proteínas Tirosina Quinasas Receptoras/metabolismo
19.
Biomedicines ; 10(8)2022 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-36009455

RESUMEN

Breast cancer has a high risk of recurrence and distant metastasis after remission. Controlling distant metastasis is important for reducing breast cancer mortality, but accomplishing this goal remains elusive. In this study, we investigated the molecular pathways underlying metastasis using cells that mimic the breast cancer distant metastasis process. HCC1143 breast cancer cells were cultured under two-dimensional (2D)-adherent, tumor sphere (TS), and reattached (ReA) culture conditions to mimic primary tumors, circulating tumor cells, and metastasized tumors, respectively. ReA cells demonstrated increased TS formation and enhanced invasion capacity compared to the original 2D-cultured parental cells. In addition, ReA cells had a higher frequency of ESA+CD44+CD24- population, which represents a stem-cell-like cell population. RNA sequencing identified the cholesterol synthesis pathway as one of the most significantly increased pathways in TS and ReA cells compared to parental cells, which was verified by measuring intracellular cholesterol levels. Furthermore, the pharmacological inhibition of the cholesterol synthesis pathway decreased the ability of cancer cells to form TSs and invade. Our results suggest that the cholesterol synthesis pathway plays an important role in the distant metastasis of breast cancer cells by augmenting TS formation and invasion capacity.

20.
Biochem Biophys Res Commun ; 407(3): 541-7, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21414300

RESUMEN

Histone chaperones function in histone transfer and regulate the nucleosome occupancy and the activity of genes. HIRA is a replication-independent (RI) histone chaperone that is linked to transcription and various developmental processes. Here, we show that HIRA interacts with Mef2 and contributes to the activation of Mef2-target genes during muscle differentiation. Asf1 cooperated with HIRA and was indispensable for Mef2-dependent transcription. The HIRA R460A mutant, which is defective in Asf1 binding, lost the transcriptional co-activation. In addition, the role of Cabin1, previously reported as a Mef2 repressor and as one of the components of the HIRA-containing complex, was delineated in Mef2/HIRA-mediated transcription. Cabin1 associated with the C-terminus of HIRA via its N-terminal domain and suppressed Mef2/HIRA-mediated transcription. Expression of Cabin1 was dramatically reduced upon myoblast differentiation, which may allow Mef2 and HIRA/Asf1 to resume their transcriptional activity. HIRA led to more permeable chromatin structure marked by active histone modifications around the myogenin promoter. Our results suggest that histone chaperone complex components contribute to the regulation of Mef2 target genes for muscle differentiation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación de la Expresión Génica , Chaperonas de Histonas/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/crecimiento & desarrollo , Factores Reguladores Miogénicos/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Calcineurina/metabolismo , Proteínas de Ciclo Celular/genética , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona , Chaperonas de Histonas/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Factores de Transcripción MEF2 , Ratones , Proteína MioD/genética , Factores Reguladores Miogénicos/genética , Miogenina/genética , Fosfoproteínas/metabolismo , Factores de Transcripción/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA