Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.096
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Cancer Res Clin Oncol ; 150(6): 294, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38842683

RESUMEN

BACKGROUND: Cervical cancer (CC) is a common malignancy amongst women globally. Ubiquitination plays a dual role in the occurrence and development of cancers. This study analyzed the mechanism of long noncoding RNA HOXC cluster antisense RNA 3 (lncRNA HOXC-AS3) in malignant proliferation of CC cells via mediating ubiquitination of lysine demethylase 5B (KDM5B/JARID1B). METHODS: The expression patterns of lncRNA HOXC-AS3 and KDM5B were measured by real-time quantitative polymerase chain reaction or Western blot analysis. After transfection with lncRNA HOXC-AS3 siRNA and pcDNA3.1-KDM5B, proliferation of CC cells was assessed by the cell counting kit-8, colony formation, and 5-Ethynyl-2'-deoxyuridine staining assays. The xenograft tumor model was established to confirm the impact of lncRNA HOXC-AS3 on CC cell proliferation in vivo by measuring tumor size and weight and the immunohistochemistry assay. The subcellular location of lncRNA HOXC-AS3 and the binding of lncRNA HOXC-AS3 to KDM5B were analyzed. After treatment of lncRNA HOXC-AS3 siRNA or MG132, the protein and ubiquitination levels of KDM5B were determined. Thereafter, the interaction and the subcellular co-location of tripartite motif-containing 37 (TRIM37) and KDM5B were analyzed by the co-immunoprecipitation and immunofluorescence assays. RESULTS: LncRNA HOXC-AS3 and KDM5B were upregulated in CC tissues and cells. Depletion of lncRNA HOXC-AS3 repressed CC cell proliferation and in vivo tumor growth. Mechanically, lncRNA HOXC-AS3 located in the nucleus directly bound to KDM5B, inhibited TRIM37-mediated ubiquitination of KDM5B, and upregulated the protein levels of KDM5B. KDM5B overexpression attenuated the inhibitory role of silencing lncRNA HOXC-AS3 in CC cell proliferation in vivo and in vitro. CONCLUSION: Nucleus-located lncRNA HOXC-AS3 facilitated malignant proliferation of CC cells via stabilization of KDM5B protein levels.


Asunto(s)
Proliferación Celular , Histona Demetilasas con Dominio de Jumonji , Ratones Desnudos , ARN Largo no Codificante , Neoplasias del Cuello Uterino , Humanos , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/metabolismo , ARN Largo no Codificante/genética , Femenino , Proliferación Celular/genética , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Ratones , Ubiquitinación , Línea Celular Tumoral , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Nucleares
2.
Nat Commun ; 15(1): 4790, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38839752

RESUMEN

Cancer cells are often addicted to serine synthesis to support growth. How serine synthesis is regulated in cancer is not well understood. We recently demonstrated protein arginine methyltransferase 1 (PRMT1) is upregulated in hepatocellular carcinoma (HCC) to methylate and activate phosphoglycerate dehydrogenase (PHGDH), thereby promoting serine synthesis. However, the mechanisms underlying PRMT1 upregulation and regulation of PRMT1-PHGDH axis remain unclear. Here, we show the E3 ubiquitin ligase F-box-only protein 7 (FBXO7) inhibits serine synthesis in HCC by binding PRMT1, inducing lysine 37 ubiquitination, and promoting proteosomal degradation of PRMT1. FBXO7-mediated PRMT1 downregulation cripples PHGDH arginine methylation and activation, resulting in impaired serine synthesis, accumulation of reactive oxygen species (ROS), and inhibition of HCC cell growth. Notably, FBXO7 is significantly downregulated in human HCC tissues, and inversely associated with PRMT1 protein and PHGDH methylation level. Overall, our study provides mechanistic insights into the regulation of cancer serine synthesis by FBXO7-PRMT1-PHGDH axis, and will facilitate the development of serine-targeting strategies for cancer therapy.


Asunto(s)
Carcinoma Hepatocelular , Proteínas F-Box , Neoplasias Hepáticas , Fosfoglicerato-Deshidrogenasa , Proteína-Arginina N-Metiltransferasas , Serina , Ubiquitinación , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Humanos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Proteínas F-Box/metabolismo , Proteínas F-Box/genética , Serina/metabolismo , Serina/biosíntesis , Fosfoglicerato-Deshidrogenasa/metabolismo , Fosfoglicerato-Deshidrogenasa/genética , Línea Celular Tumoral , Animales , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Ratones , Proliferación Celular , Metilación , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Masculino , Células HEK293 , Femenino , Células Hep G2
3.
Int J Biol Sci ; 20(7): 2622-2639, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725840

RESUMEN

Sorafenib is a standard first-line drug for advanced hepatocellular carcinoma, but the serious cardiotoxic effects restrict its therapeutic applicability. Here, we show that iron-dependent ferroptosis plays a vital role in sorafenib-induced cardiotoxicity. Remarkably, our in vivo and in vitro experiments demonstrated that ferroptosis inhibitor application neutralized sorafenib-induced heart injury. By analyzing transcriptome profiles of adult human sorafenib-treated cardiomyocytes, we found that Krüppel-like transcription factor 11 (KLF11) expression significantly increased after sorafenib stimulation. Mechanistically, KLF11 promoted ferroptosis by suppressing transcription of ferroptosis suppressor protein 1 (FSP1), a seminal breakthrough due to its ferroptosis-repressing properties. Moreover, FSP1 knockdown showed equivalent results to glutathione peroxidase 4 (GPX4) knockdown, and FSP1 overexpression counteracted GPX4 inhibition-induced ferroptosis to a substantial extent. Cardiac-specific overexpression of FSP1 and silencing KLF11 by an adeno-associated virus serotype 9 markedly improved cardiac dysfunction in sorafenib-treated mice. In summary, FSP1-mediated ferroptosis is a crucial mechanism for sorafenib-provoked cardiotoxicity, and targeting ferroptosis may be a promising therapeutic strategy for alleviating sorafenib-induced cardiac damage.


Asunto(s)
Cardiotoxicidad , Ferroptosis , Proteína de Unión al Calcio S100A4 , Sorafenib , Sorafenib/efectos adversos , Ferroptosis/efectos de los fármacos , Animales , Ratones , Cardiotoxicidad/metabolismo , Cardiotoxicidad/etiología , Humanos , Proteína de Unión al Calcio S100A4/metabolismo , Proteína de Unión al Calcio S100A4/genética , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Proteínas Represoras/metabolismo , Proteínas Represoras/genética
4.
Int J Biol Sci ; 20(7): 2422-2439, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725842

RESUMEN

Background & Aims: Reactive oxygen species (ROS) act as modulators triggering cellular dysfunctions and organ damage including liver fibrosis in which hepatic stellate cell (HSC) activation plays a key role. Previous studies suggest that microRNA-144 (miR-144) acts as a pro-oxidant molecule; however, whether and how miR-144 affects HSC activation and liver fibrosis remain unknown. Methods: Carbon tetrachloride (CCl4) and bile duct ligation (BDL)-induced experimental liver fibrosis models were used. Hepatic miR-144 expression was analyzed by miRNA in situ hybridization with RNAscope probe. The in vivo effects of silencing or overexpressing miR-144 were examined with an adeno-associated virus 6 (AAV6) carrying miR-144 inhibitor or mimics in fibrotic mouse experimental models. Results: In this study, we demonstrated that ROS treatment significantly upregulated miR-144 in HSCs, which further promoted HSC activation in vitro. Interestingly, miR-144 was preferentially elevated in HSCs of experimental liver fibrosis in mice and in human liver fibrotic tissues. Furthermore, in vivo loss or gain-of-function experiments via AAV6 carrying miR-144 antagomir or agomir revealed that blockade of miR-144 in HSCs mitigated, while overexpression of miR-144 in HSCs accelerated the development of experimental liver fibrosis. Mechanistically, SIN3 transcription regulator family member A (SIN3A), a transcriptional repressor, was identified to be the target of miR-144 in HSCs. MiR-144 downregulated Sin3A, and in line with this result, specific knockdown of Sin3a in HSCs remarkedly activated p38 MAPK signaling pathway to promote HSC activation, eventually exacerbating liver fibrosis. Conclusions: Oxidative stress-driven miR-144 fuels HSC activation and liver fibrogenesis by limiting the SIN3A-p38 axis. Thus, a specific inhibition of miR-144 in HSCs could be a novel therapeutic strategy for the treatment of liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas , Cirrosis Hepática , MicroARNs , Estrés Oxidativo , Especies Reactivas de Oxígeno , Complejo Correpresor Histona Desacetilasa y Sin3 , Proteínas Quinasas p38 Activadas por Mitógenos , MicroARNs/metabolismo , MicroARNs/genética , Animales , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones , Humanos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Tetracloruro de Carbono
5.
Cells ; 13(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38727269

RESUMEN

The histone deacetylase inhibitor (HDACi) valproic acid (VPA) has neuroprotective and anti-inflammatory effects in experimental traumatic brain injury (TBI), which have been partially attributed to the epigenetic disinhibition of the transcription repressor RE1-Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). Additionally, VPA changes post-traumatic brain injury (TBI) brain metabolism to create a neuroprotective environment. To address the interconnection of neuroprotection, metabolism, inflammation and REST/NRSF after TBI, we subjected C57BL/6N mice to experimental TBI and intraperitoneal VPA administration or vehicle solution at 15 min, 1, 2, and 3 days post-injury (dpi). At 7 dpi, TBI-induced an up-regulation of REST/NRSF gene expression and HDACi function of VPA on histone H3 acetylation were confirmed. Neurological deficits, brain lesion size, blood-brain barrier permeability, or astrogliosis were not affected, and REST/NRSF target genes were only marginally influenced by VPA. However, VPA attenuated structural damage in the hippocampus, microgliosis and expression of the pro-inflammatory marker genes. Analyses of plasma lipidomic and polar metabolomic patterns revealed that VPA treatment increased lysophosphatidylcholines (LPCs), which were inversely associated with interleukin 1 beta (Il1b) and tumor necrosis factor (Tnf) gene expression in the brain. The results show that VPA has mild neuroprotective and anti-inflammatory effects likely originating from favorable systemic metabolic changes resulting in increased plasma LPCs that are known to be actively taken up by the brain and function as carriers for neuroprotective polyunsaturated fatty acids.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Inflamación , Lisofosfatidilcolinas , Ratones Endogámicos C57BL , Neuronas , Ácido Valproico , Animales , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/sangre , Lesiones Traumáticas del Encéfalo/complicaciones , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Ratones , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/metabolismo , Inflamación/patología , Inflamación/tratamiento farmacológico , Lisofosfatidilcolinas/sangre , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Represoras/metabolismo , Proteínas Represoras/genética
6.
J Cell Biol ; 223(8)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38709169

RESUMEN

Histone H3 lysine36 dimethylation (H3K36me2) is generally distributed in the gene body and euchromatic intergenic regions. However, we found that H3K36me2 is enriched in pericentromeric heterochromatin in some mouse cell lines. We here revealed the mechanism of heterochromatin targeting of H3K36me2. Among several H3K36 methyltransferases, NSD2 was responsible for inducing heterochromatic H3K36me2. Depletion and overexpression analyses of NSD2-associating proteins revealed that NSD2 recruitment to heterochromatin was mediated through the imitation switch (ISWI) chromatin remodeling complexes, such as BAZ1B-SMARCA5 (WICH), which directly binds to AT-rich DNA via a BAZ1B domain-containing AT-hook-like motifs. The abundance and stoichiometry of NSD2, SMARCA5, and BAZ1B could determine the localization of H3K36me2 in different cell types. In mouse embryos, H3K36me2 heterochromatin localization was observed at the two- to four-cell stages, suggesting its physiological relevance.


Asunto(s)
Ensamble y Desensamble de Cromatina , Heterocromatina , N-Metiltransferasa de Histona-Lisina , Histonas , Proteínas Represoras , Animales , Humanos , Ratones , Adenosina Trifosfatasas , Proteínas que Contienen Bromodominio/genética , Proteínas que Contienen Bromodominio/metabolismo , Centrómero/metabolismo , Centrómero/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/genética , Heterocromatina/metabolismo , Heterocromatina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Histonas/genética , Metilación , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
7.
Front Immunol ; 15: 1293723, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690263

RESUMEN

T cells must adapt to variations in tissue microenvironments; these adaptations include the degree of oxygen availability. The hypoxia-inducible factor (HIF) transcription factors control much of this adaptation, and thus regulate many aspects of T cell activation and function. The HIFs are in turn regulated by oxygen-dependent hydroxylases: both the prolyl hydroxylases (PHDs) which interact with the VHL tumour suppressor and control HIF turnover, and the asparaginyl hydroxylase known as the Factor inhibiting HIF (FIH), which modulates HIF transcriptional activity. To determine the role of this latter factor in T cell function, we generated T cell-specific FIH knockout mice. We found that FIH regulates T cell fate and function in a HIF-dependent manner and show that the effects of FIH activity occur predominantly at physiological oxygen concentrations. T cell-specific loss of FIH boosts T cell cytotoxicity, augments T cell expansion in vivo, and improves anti-tumour immunotherapy in mice. Specifically inhibiting FIH in T cells may therefore represent a promising strategy for cancer immunotherapy.


Asunto(s)
Diferenciación Celular , Ratones Noqueados , Animales , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Activación de Linfocitos/inmunología , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Ratones Endogámicos C57BL
8.
Proc Natl Acad Sci U S A ; 121(23): e2318740121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38805275

RESUMEN

Repressor element-1 silencing transcription factor (REST) is required for the formation of mature neurons. REST dysregulation underlies a key mechanism of neurodegeneration associated with neurological disorders. However, the mechanisms leading to alterations of REST-mediated silencing of key neurogenesis genes are not known. Here, we show that BRCA1 Associated ATM Activator 1 (BRAT1), a gene linked to neurodegenerative diseases, is required for the activation of REST-responsive genes during neuronal differentiation. We find that INTS11 and INTS9 subunits of Integrator complex interact with BRAT1 as a distinct trimeric complex to activate critical neuronal genes during differentiation. BRAT1 depletion results in persistence of REST residence on critical neuronal genes disrupting the differentiation of NT2 cells into astrocytes and neuronal cells. We identified BRAT1 and INTS11 co-occupying the promoter region of these genes and pinpoint a role for BRAT1 in recruiting INTS11 to their promoters. Disease-causing mutations in BRAT1 diminish its association with INTS11/INTS9, linking the manifestation of disease phenotypes with a defect in transcriptional activation of key neuronal genes by BRAT1/INTS11/INTS9 complex. Finally, loss of Brat1 in mouse embryonic stem cells leads to a defect in neuronal differentiation assay. Importantly, while reconstitution with wild-type BRAT1 restores neuronal differentiation, the addition of a BRAT1 mutant is unable to associate with INTS11/INTS9 and fails to rescue the neuronal phenotype. Taken together, our study highlights the importance of BRAT1 association with INTS11 and INTS9 in the development of the nervous system.


Asunto(s)
Diferenciación Celular , Cromatina , Neurogénesis , Neuronas , Proteínas Represoras , Cromatina/metabolismo , Cromatina/genética , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Humanos , Neurogénesis/genética , Animales , Ratones , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas del Tejido Nervioso , Proteínas Co-Represoras
9.
Breast Cancer Res ; 26(1): 74, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702730

RESUMEN

The transcription factor TRPS1 is a context-dependent oncogene in breast cancer. In the mammary gland, TRPS1 activity is restricted to the luminal population and is critical during puberty and pregnancy. Its function in the resting state remains however unclear. To evaluate whether it could be a target for cancer therapy, we investigated TRPS1 function in the healthy adult mammary gland using a conditional ubiquitous depletion mouse model where long-term depletion does not affect fitness. Using transcriptomic approaches, flow cytometry and functional assays, we show that TRPS1 activity is essential to maintain a functional luminal progenitor compartment. This requires the repression of both YAP/TAZ and SRF/MRTF activities. TRPS1 represses SRF/MRTF activity indirectly by modulating RhoA activity. Our work uncovers a hitherto undisclosed function of TRPS1 in luminal progenitors intrinsically linked to mechanotransduction in the mammary gland. It may also provide new insights into the oncogenic functions of TRPS1 as luminal progenitors are likely the cells of origin of many breast cancers.


Asunto(s)
Glándulas Mamarias Animales , Proteínas Represoras , Factor de Respuesta Sérica , Células Madre , Factores de Transcripción , Animales , Femenino , Ratones , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Células Madre/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factor de Respuesta Sérica/metabolismo , Factor de Respuesta Sérica/genética , Humanos , Transactivadores/metabolismo , Transactivadores/genética
10.
Cancer Lett ; 592: 216926, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38714291

RESUMEN

Gastric cancer (GC) is one of the most common cancer worldwide. Neural invasion (NI) is considered as the symbiotic interaction between nerves and cancers, which strongly affects the prognosis of GC patients. Small extracellular vesicles (sEVs) play a key role in intercellular communication. However, whether sEVs mediate GC-NI remains unexplored. In this study, sEVs release inhibitor reduces the NI potential of GC cells. Muscarinic receptor M3 on GC-derived sEVs regulates their absorption by neuronal cells. The enrichment of sEV-circVAPA in NI-positive patients' serum is validated by serum high throughput sEV-circRNA sequencing and clinical samples. sEV-circVAPA promotes GC-NI in vitro and in vivo. Mechanistically, sEV-circVAPA decreases SLIT2 transcription by miR-548p/TGIF2 and inhibits SLIT2 translation via binding to eIF4G1, thereby downregulates SLIT2 expression in neuronal cells and finally induces GC-NI. Together, this work identifies the preferential absorption mechanism of GC-derived sEVs by neuronal cells and demonstrates a previously undefined role of GC-derived sEV-circRNA in GC-NI, which provides new insight into sEV-circRNA based diagnostic and therapeutic strategies for NI-positive GC patients.


Asunto(s)
Vesículas Extracelulares , Péptidos y Proteínas de Señalización Intercelular , Invasividad Neoplásica , Proteínas del Tejido Nervioso , Neuronas , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Animales , Neuronas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Línea Celular Tumoral , Ratones , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Masculino , Femenino , Proliferación Celular , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
11.
Biomolecules ; 14(5)2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38785968

RESUMEN

Plakophilin 1 (PKP1), a member of the p120ctn subfamily of the armadillo (ARM)-repeat-containing proteins, is an important structural component of cell-cell adhesion scaffolds although it can also be ubiquitously found in the cytoplasm and the nucleus. RYBP (RING 1A and YY1 binding protein) is a multifunctional intrinsically disordered protein (IDP) best described as a transcriptional regulator. Both proteins are involved in the development and metastasis of several types of tumors. We studied the binding of the armadillo domain of PKP1 (ARM-PKP1) with RYBP by using in cellulo methods, namely immunofluorescence (IF) and proximity ligation assay (PLA), and in vitro biophysical techniques, namely fluorescence, far-ultraviolet (far-UV) circular dichroism (CD), and isothermal titration calorimetry (ITC). We also characterized the binding of the two proteins by using in silico experiments. Our results showed that there was binding in tumor and non-tumoral cell lines. Binding in vitro between the two proteins was also monitored and found to occur with a dissociation constant in the low micromolar range (~10 µM). Finally, in silico experiments provided additional information on the possible structure of the binding complex, especially on the binding ARM-PKP1 hot-spot. Our findings suggest that RYBP might be a rescuer of the high expression of PKP1 in tumors, where it could decrease the epithelial-mesenchymal transition in some cancer cells.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Placofilinas , Unión Proteica , Humanos , Placofilinas/metabolismo , Placofilinas/genética , Placofilinas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas del Dominio Armadillo/metabolismo , Proteínas del Dominio Armadillo/química , Proteínas del Dominio Armadillo/genética , Dominios Proteicos , Dicroismo Circular
12.
Acta Neuropathol Commun ; 12(1): 72, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711090

RESUMEN

The RE1-silencing transcription factor (REST) acts either as a repressor or activator of transcription depending on the genomic and cellular context. REST is a key player in brain cell differentiation by inducing chromatin modifications, including DNA methylation, in a proximity of its binding sites. Its dysfunction may contribute to oncogenesis. Mutations in IDH1/2 significantly change the epigenome contributing to blockade of cell differentiation and glioma development. We aimed at defining how REST modulates gene activation and repression in the context of the IDH mutation-related phenotype in gliomas. We studied the effects of REST knockdown, genome wide occurrence of REST binding sites, and DNA methylation of REST motifs in IDH wild type and IDH mutant gliomas. We found that REST target genes, REST binding patterns, and TF motif occurrence proximal to REST binding sites differed in IDH wild-type and mutant gliomas. Among differentially expressed REST targets were genes involved in glial cell differentiation and extracellular matrix organization, some of which were differentially methylated at promoters or gene bodies. REST knockdown differently impacted invasion of the parental or IDH1 mutant glioma cells. The canonical REST-repressed gene targets showed significant correlation with the GBM NPC-like cellular state. Interestingly, results of REST or KAISO silencing suggested the interplay between these TFs in regulation of REST-activated and repressed targets. The identified gene regulatory networks and putative REST cooperativity with other TFs, such as KAISO, show distinct REST target regulatory networks in IDH-WT and IDH-MUT gliomas, without concomitant DNA methylation changes. We conclude that REST could be an important therapeutic target in gliomas.


Asunto(s)
Neoplasias Encefálicas , Metilación de ADN , Redes Reguladoras de Genes , Glioma , Isocitrato Deshidrogenasa , Mutación , Isocitrato Deshidrogenasa/genética , Glioma/genética , Glioma/patología , Glioma/metabolismo , Humanos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Regulación Neoplásica de la Expresión Génica/genética
13.
Med Oncol ; 41(7): 166, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38819709

RESUMEN

The full-length p200CUX1 protein encoded by the homology frame CUT-like protein (CUX1) plays an important role in tumors as a pro-oncogene or oncogene. However, its role and mechanism in acute myeloid leukemia remain unknown. p200CUX1 regulates several pathways, including the MAPK signaling pathway. Our data showed that p200CUX1 is lowly expressed in THP1 and U937 AML cell lines. Lentiviral overexpression of p200CUX1 reduced proliferation and promoted apoptosis and G0/G1 phase blockade, correlating with MAPK pathway suppression. Additionally, p200CUX1 regulated the expression of bone morphogenetic protein 8B (BMP8B), which is overexpressed in AML. Overexpression of p200CUX1 downregulated BMP8B expression and inhibited the MAPK pathway. Furthermore, BMP8B knockdown inhibited AML cell proliferation, enhanced apoptosis and the sensitivity of ATRA-induced cell differentiation, and blocked G0/G1 transition. Our findings demonstrate the pivotal function of the p200CUX1-BMP8B-MAPK axis in maintaining the viability of AML cells. Consequently, targeting p200CUX1 could represent a viable strategy in AML therapy.


Asunto(s)
Apoptosis , Proliferación Celular , Leucemia Mieloide Aguda , Sistema de Señalización de MAP Quinasas , Humanos , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/genética , Sistema de Señalización de MAP Quinasas/fisiología , Línea Celular Tumoral , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Progresión de la Enfermedad
14.
Genes (Basel) ; 15(5)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38790189

RESUMEN

BACKGROUND: Cervical cancer is among the highest-ranking types of cancer worldwide, with human papillomavirus (HPV) as the agent driving the malignant process. One aspect of the infection's evolution is given by epigenetic modifications, mainly DNA methylation and chromatin alteration. These processes are guided by several chromatin remodeling complexes, including NuRD. The purpose of this study was to evaluate the genome-wide binding patterns of the NuRD complex components (MBD2 and MBD3) in the presence of active HPV16 E6 and E7 oncogenes and to determine the potential of identified genes through an experimental model to differentiate between cervical precursor lesions, with the aim of establishing their utility as biomarkers. METHODS: The experimental model was built using the CaSki cell line and shRNA for E6 and E7 HPV16 silencing, ChIP-seq, qRT-PCR, and Western blot analyses. Selected genes' expression was also assessed in patients. RESULTS: Several genes have been identified to exhibit altered transcriptional activity due to the influence of HPV16 E6/E7 viral oncogenes acting through the MBD2/MBD3 NuRD complex, linking them to viral infection and cervical oncogenesis. CONCLUSIONS: The impacted genes primarily play roles in governing gene transcription, mRNA processing, and regulation of translation. Understanding these mechanisms offers valuable insights into the process of HPV-induced oncogenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Proteínas Oncogénicas Virales , Proteínas E7 de Papillomavirus , Proteínas Represoras , Neoplasias del Cuello Uterino , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Neoplasias del Cuello Uterino/virología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Ensamble y Desensamble de Cromatina/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Línea Celular Tumoral , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 16/patogenicidad , Carcinogénesis/genética , Infecciones por Papillomavirus/virología , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Regulación Neoplásica de la Expresión Génica
15.
Int J Mol Sci ; 25(10)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38791157

RESUMEN

The additional sex combs-like (ASXL) family, a mammalian homolog of the additional sex combs (Asx) of Drosophila, has been implicated in transcriptional regulation via chromatin modifications. Abnormal expression of ASXL family genes leads to myelodysplastic syndromes and various types of leukemia. De novo mutation of these genes also causes developmental disorders. Genes in this family and their neighbor genes are evolutionary conserved in humans and mice. This review provides a comprehensive summary of epigenetic regulations associated with ASXL family genes. Their expression is commonly regulated by DNA methylation at CpG islands preceding transcription starting sites. Their proteins primarily engage in histone tail modifications through interactions with chromatin regulators (PRC2, TrxG, PR-DUB, SRC1, HP1α, and BET proteins) and with transcription factors, including nuclear hormone receptors (RAR, PPAR, ER, and LXR). Histone modifications associated with these factors include histone H3K9 acetylation and methylation, H3K4 methylation, H3K27 methylation, and H2AK119 deubiquitination. Recently, non-coding RNAs have been identified following mutations in the ASXL1 or ASXL3 gene, along with circular ASXLs and microRNAs that regulate ASXL1 expression. The diverse epigenetic regulations linked to ASXL family genes collectively contribute to tumor suppression and developmental processes. Our understanding of ASXL-regulated epigenetics may provide insights into the development of therapeutic epigenetic drugs.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Humanos , Animales , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Histonas/metabolismo , Mutación
16.
Cell Rep ; 43(5): 114227, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38735044

RESUMEN

CUX1 is a homeodomain-containing transcription factor that is essential for the development and differentiation of multiple tissues. CUX1 is recurrently mutated or deleted in cancer, particularly in myeloid malignancies. However, the mechanism by which CUX1 regulates gene expression and differentiation remains poorly understood, creating a barrier to understanding the tumor-suppressive functions of CUX1. Here, we demonstrate that CUX1 directs the BAF chromatin remodeling complex to DNA to increase chromatin accessibility in hematopoietic cells. CUX1 preferentially regulates lineage-specific enhancers, and CUX1 target genes are predictive of cell fate in vivo. These data indicate that CUX1 regulates hematopoietic lineage commitment and homeostasis via pioneer factor activity, and CUX1 deficiency disrupts these processes in stem and progenitor cells, facilitating transformation.


Asunto(s)
Cromatina , Células Madre Hematopoyéticas , Proteínas de Homeodominio , Proteínas Represoras , Humanos , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Cromatina/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Animales , Ratones , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos/genética
17.
Cell Rep ; 43(5): 114176, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38691454

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) carries a dismal prognosis due to therapeutic resistance. We show that PDAC cells undergo global epigenetic reprogramming to acquire chemoresistance, a process that is driven at least in part by protein arginine methyltransferase 1 (PRMT1). Genetic or pharmacological PRMT1 inhibition impairs adaptive epigenetic reprogramming and delays acquired resistance to gemcitabine and other common chemo drugs. Mechanistically, gemcitabine treatment induces translocation of PRMT1 into the nucleus, where its enzymatic activity limits the assembly of chromatin-bound MAFF/BACH1 transcriptional complexes. Cut&Tag chromatin profiling of H3K27Ac, MAFF, and BACH1 suggests a pivotal role for MAFF/BACH1 in global epigenetic response to gemcitabine, which is confirmed by genetically silencing MAFF. PRMT1 and MAFF/BACH1 signature genes identified by Cut&Tag analysis distinguish gemcitabine-resistant from gemcitabine-sensitive patient-derived xenografts of PDAC, supporting the PRMT1-MAFF/BACH1 epigenetic regulatory axis as a potential therapeutic avenue for improving the efficacy and durability of chemotherapies in patients of PDAC.


Asunto(s)
Desoxicitidina , Resistencia a Antineoplásicos , Epigénesis Genética , Gemcitabina , Neoplasias Pancreáticas , Proteína-Arginina N-Metiltransferasas , Proteínas Represoras , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Humanos , Resistencia a Antineoplásicos/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Línea Celular Tumoral , Animales , Ratones , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/genética
18.
Int J Med Sci ; 21(6): 983-993, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38774750

RESUMEN

Previous studies have highlighted the protective effects of pyruvate kinase M2 (PKM2) overexpression in septic cardiomyopathy. In our study, we utilized cardiomyocyte-specific PKM2 knockout mice to further investigate the role of PKM2 in attenuating LPS-induced myocardial dysfunction, focusing on mitochondrial biogenesis and prohibitin 2 (PHB2). Our findings confirmed that the deletion of PKM2 in cardiomyocytes significantly exacerbated LPS-induced myocardial dysfunction, as evidenced by impaired contractile function and relaxation. Additionally, the deletion of PKM2 intensified LPS-induced myocardial inflammation. At the molecular level, LPS triggered mitochondrial dysfunction, characterized by reduced ATP production, compromised mitochondrial respiratory complex I/III activities, and increased ROS production. Intriguingly, the absence of PKM2 further worsened LPS-induced mitochondrial damage. Our molecular investigations revealed that LPS disrupted mitochondrial biogenesis in cardiomyocytes, a disruption that was exacerbated by the absence of PKM2. Given that PHB2 is known as a downstream effector of PKM2, we employed PHB2 adenovirus to restore PHB2 levels. The overexpression of PHB2 normalized mitochondrial biogenesis, restored mitochondrial integrity, and promoted mitochondrial function. Overall, our results underscore the critical role of PKM2 in regulating the progression of septic cardiomyopathy. PKM2 deficiency impeded mitochondrial biogenesis, leading to compromised mitochondrial integrity, increased myocardial inflammation, and impaired cardiac function. The overexpression of PHB2 mitigated the deleterious effects of PKM2 deletion. This discovery offers a novel insight into the molecular mechanisms underlying septic cardiomyopathy and suggests potential therapeutic targets for intervention.


Asunto(s)
Cardiomiopatías , Ratones Noqueados , Mitocondrias Cardíacas , Miocitos Cardíacos , Prohibitinas , Piruvato Quinasa , Sepsis , Animales , Cardiomiopatías/patología , Cardiomiopatías/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/etiología , Ratones , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Sepsis/metabolismo , Sepsis/patología , Sepsis/genética , Piruvato Quinasa/metabolismo , Piruvato Quinasa/genética , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Humanos , Biogénesis de Organelos , Lipopolisacáridos/toxicidad , Masculino , Modelos Animales de Enfermedad
19.
Int J Med Sci ; 21(7): 1194-1203, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38818468

RESUMEN

This study aims to elucidate the roles of Phosphoglycerate Mutase Family Member 5 (Pgam5) and Prohibitin 2 (Phb2) in the context of hyperglycemia-induced myocardial dysfunction, a critical aspect of diabetic cardiomyopathy. The research employed primary cardiomyocytes, which were then subjected to hyperglycemia treatment to mimic diabetic conditions. We used siRNA transfection to knock down Pgam5 and overexpressed Phb2 using adenovirus transfection to assess their individual and combined effects on cardiomyocyte health. Mitochondrial function was evaluated through measurements of mitochondrial membrane potential using the JC-1 probe, and levels of mitochondrial reactive oxygen species (ROS) were assessed. Additionally, the study involved qPCR analysis to quantify the transcriptional changes in genes related to mitochondrial fission and mitophagy. Our findings indicate that hyperglycemia significantly reduces cardiomyocyte viability and impairs mitochondrial function, as evidenced by decreased mitochondrial membrane potential and increased ROS levels. Pgam5 knockdown was observed to mitigate these adverse effects, preserving mitochondrial function and cardiomyocyte viability. On the molecular level, Pgam5 was found to regulate genes associated with mitochondrial fission (such as Drp1, Mff, and Fis1) and mitophagy (including Parkin, Bnip3, and Fundc1). Furthermore, overexpression of Phb2 countered the hyperglycemia-induced mitochondrial dysfunction and normalized the levels of key mitochondrial antioxidant enzymes. The combined data suggest a protective role for both Pgam5 knockdown and Phb2 overexpression against hyperglycemia-induced cellular and mitochondrial damage. The study elucidates the critical roles of Pgam5 and Phb2 in regulating mitochondrial dynamics in the setting of hyperglycemia-induced myocardial dysfunction. By modulating mitochondrial fission and mitophagy, Pgam5 and Phb2 emerge as key players in preserving mitochondrial integrity and cardiomyocyte health under diabetic conditions. These findings contribute significantly to our understanding of the molecular mechanisms underlying diabetic cardiomyopathy and suggest potential therapeutic targets for mitigating myocardial dysfunction in diabetes.


Asunto(s)
Cardiomiopatías Diabéticas , Hiperglucemia , Potencial de la Membrana Mitocondrial , Dinámicas Mitocondriales , Miocitos Cardíacos , Prohibitinas , Especies Reactivas de Oxígeno , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Dinámicas Mitocondriales/genética , Hiperglucemia/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/genética , Humanos , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/etiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Mitofagia/genética , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Ratas
20.
Nat Commun ; 15(1): 4450, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789411

RESUMEN

Histone deacetylases (HDACs) play a crucial role in transcriptional regulation and are implicated in various diseases, including cancer. They are involved in histone tail deacetylation and canonically linked to transcriptional repression. Previous studies suggested that HDAC recruitment to cell-cycle gene promoters via the retinoblastoma (RB) protein or the DREAM complex through SIN3B is essential for G1/S and G2/M gene repression during cell-cycle arrest and exit. Here we investigate the interplay among DREAM, RB, SIN3 proteins, and HDACs in the context of cell-cycle gene repression. Knockout of SIN3B does not globally derepress cell-cycle genes in non-proliferating HCT116 and C2C12 cells. Loss of SIN3A/B moderately upregulates several cell-cycle genes in HCT116 cells but does so independently of DREAM/RB. HDAC inhibition does not induce general upregulation of RB/DREAM target genes in arrested transformed or non-transformed cells. Our findings suggest that E2F:RB and DREAM complexes can repress cell-cycle genes without relying on HDAC activity.


Asunto(s)
Factores de Transcripción E2F , Histona Desacetilasas , Proteínas Represoras , Proteína de Retinoblastoma , Humanos , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Células HCT116 , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factores de Transcripción E2F/metabolismo , Factores de Transcripción E2F/genética , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/genética , Ratones , Animales , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3/genética , Proteínas de Interacción con los Canales Kv/metabolismo , Proteínas de Interacción con los Canales Kv/genética , Ciclo Celular/genética , Regiones Promotoras Genéticas/genética , Regulación de la Expresión Génica , Genes cdc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA