Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 697-705, 2024 Apr 20.
Artículo en Chino | MEDLINE | ID: mdl-38708503

RESUMEN

OBJECTIVE: To explore the role of zinc finger protein 36(ZFP36) in regulating osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) and preosteoblasts. METHODS: ZFP36 expression was observed in primary mouse BMSCs and mouse preosteoblasts (MC3T3-E1 cells) during induced osteogenic differentiation. Zfp36-deficient cell models were constructed in the two cells using RNA interference technique and the changes in differentiation capacities of the transfected cells into osteoblasts were observed. Transcriptome sequencing was used to investigate the potential mechanisms of ZFP36 for regulating osteoblast differentiation of the two cells. U0126, a ERK/MAPK signal suppressor, was used to verify the regulatory mechanism of Zfp36 in osteogenic differentiation of Zfp36-deficient cells. RESULTS: During the 14-day induction of osteogenic differentiation, both mouse BMSCs and MC3T3-E1 cells exhibited increased expression of ZFP36, and its mRNA expression reached the peak level on Day 7(P < 0.0001). The Zfp36-deficient cell models showed reduced intensity of alkaline phosphatase (ALP) staining and alizarin red staining with significantly lowered expressions of the osteogenic marker genes including Alpl, Sp7, Bglap and Ibsp (P < 0.01). Transcriptome sequencing verified the reduction of bone mineralization-related gene expressions in Zfp36-deficient cells and indicated the involvement of ERK signaling in the potential regulatory mechanism of Zfp36. Immunoblotting showed that pERK protein expression increased significantly in Zfp36-deficient cells compared with the control cells. In Zfp36-deficient MC3T3-E1 cells, inhibition of activated ERK/MAPK signaling with U0126 resulted in obviously enhanced ALP staining and significantly increased expressions of osteoblast differentiation markers Runx2 and Bglap (P < 0.05). CONCLUSIONS: ZFP36 is involved in the regulation of osteoblast differentiation of mouse BMSCs and preosteoblasts, and ZFP36 deficiency causes inhibition of osteoblast differentiation of the cells by activating the ERK/MAPK signaling pathway.


Asunto(s)
Diferenciación Celular , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas , Osteoblastos , Osteogénesis , Animales , Ratones , Fosfatasa Alcalina/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Factor 1 de Respuesta al Butirato/metabolismo , Factor 1 de Respuesta al Butirato/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo
2.
Mol Cancer Res ; 22(1): 29-40, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-37801008

RESUMEN

Achaete-scute family bHLH transcription factor 1 (ASCL1) is a master transcription factor involved in neuroendocrine differentiation. ASCL1 is expressed in approximately 10% of lung adenocarcinomas (LUAD) and exerts tumor-promoting effects. Here, we explored miRNA profiles in ASCL1-positive LUADs and identified several miRNAs closely associated with ASCL1 expression, including miR-375, miR-95-3p/miR-95-5p, miR-124-3p, and members of the miR-17∼92 family. Similar to small cell lung cancer, Yes1 associated transcriptional regulator (YAP1), a representative miR-375 target gene, was suppressed in ASCL1-positive LUADs. ASCL1 knockdown followed by miRNA profiling in a cell culture model further revealed that ASCL1 positively regulates miR-124-3p and members of the miR-17∼92 family. Integrative transcriptomic analyses identified ZFP36 ring finger protein like 1 (ZFP36L1) as a target gene of miR-124-3p, and IHC studies demonstrated that ASCL1-positive LUADs are associated with low ZFP36L1 protein levels. Cell culture studies showed that ectopic ZFP36L1 expression inhibits cell proliferation, survival, and cell-cycle progression. Moreover, ZFP36L1 negatively regulated several genes including E2F transcription factor 1 (E2F1) and snail family transcriptional repressor 1 (SNAI1). In conclusion, our study revealed that suppression of ZFP36L1 via ASCL1-regulated miR-124-3p could modulate gene expression, providing evidence that ASCL1-mediated regulation of miRNAs shapes molecular features of ASCL1-positive LUADs. IMPLICATIONS: Our study revealed unique miRNA profiles of ASCL1-positive LUADs and identified ASCL1-regulated miRNAs with functional relevance.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , MicroARNs , Carcinoma Pulmonar de Células Pequeñas , Humanos , Línea Celular Tumoral , Adenocarcinoma del Pulmón/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Carcinoma Pulmonar de Células Pequeñas/genética , Proliferación Celular/genética , Neoplasias Pulmonares/patología , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor 1 de Respuesta al Butirato/genética , Factor 1 de Respuesta al Butirato/metabolismo
3.
Cell Rep ; 42(5): 112419, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37074914

RESUMEN

Potent T cell responses against infections and malignancies require a rapid yet tightly regulated production of toxic effector molecules. Their production level is defined by post-transcriptional events at 3' untranslated regions (3' UTRs). RNA binding proteins (RBPs) are key regulators in this process. With an RNA aptamer-based capture assay, we identify >130 RBPs interacting with IFNG, TNF, and IL2 3' UTRs in human T cells. RBP-RNA interactions show plasticity upon T cell activation. Furthermore, we uncover the intricate and time-dependent regulation of cytokine production by RBPs: whereas HuR supports early cytokine production, ZFP36L1, ATXN2L, and ZC3HAV1 dampen and shorten the production duration, each at different time points. Strikingly, even though ZFP36L1 deletion does not rescue the dysfunctional phenotype, tumor-infiltrating T cells produce more cytokines and cytotoxic molecules, resulting in superior anti-tumoral T cell responses. Our findings thus show that identifying RBP-RNA interactions reveals key modulators of T cell responses in health and disease.


Asunto(s)
Citocinas , Linfocitos T , Humanos , Linfocitos T/metabolismo , Regiones no Traducidas 3' , Citocinas/metabolismo , Proteínas de Unión al ARN/metabolismo , Factor 1 de Respuesta al Butirato/genética , Factor 1 de Respuesta al Butirato/metabolismo
4.
Signal Transduct Target Ther ; 8(1): 145, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37032379

RESUMEN

Genetic classification helps to disclose molecular heterogeneity and therapeutic implications in diffuse large B-cell lymphoma (DLBCL). Using whole exome/genome sequencing, RNA-sequencing, and fluorescence in situ hybridization in 337 newly diagnosed DLBCL patients, we established a simplified 38-gene algorithm (termed 'LymphPlex') based on the information on mutations of 35 genes and rearrangements of three genes (BCL2, BCL6, and MYC), identifying seven distinct genetic subtypes: TP53Mut (TP53 mutations), MCD-like (MYD88, CD79B, PIM1, MPEG1, BTG1, TBL1XR1, PRDM1, IRF4 mutations), BN2-like (BCL6 fusion, NOTCH2, CD70, DTX1, BTG2, TNFAIP3, CCND3 mutations), N1-like (NOTCH1 mutations), EZB-like (BCL2 fusion, EZH2, TNFRSF14, KMT2D, B2M, FAS, CREBBP, ARID1A, EP300, CIITA, STAT6, GNA13 mutations, with or without MYC rearrangement), and ST2-like (SGK1, TET2, SOCS1, DDX3X, ZFP36L1, DUSP2, STAT3, IRF8 mutations). Extended validation of 1001 DLBCL patients revealed clinical relevance and biological signature of each genetic subtype. TP53Mut subtype showed poor prognosis, characterized by p53 signaling dysregulation, immune deficiency, and PI3K activation. MCD-like subtype was associated with poor prognosis, activated B-cell (ABC) origin, BCL2/MYC double-expression, and NF-κB activation. BN2-like subtype showed favorable outcome within ABC-DLBCL and featured with NF-κB activation. N1-like and EZB-like subtypes were predominated by ABC-DLBCL and germinal center B-cell (GCB)-DLBCL, respectively. EZB-like-MYC+ subtype was characterized by an immunosuppressive tumor microenvironment, while EZB-like-MYC- subtype by NOTCH activation. ST2-like subtype showed favorable outcome within GCB-DLBCL and featured with stromal-1 modulation. Genetic subtype-guided targeted agents achieved encouraging clinical response when combined with immunochemotherapy. Collectively, LymphPlex provided high efficacy and feasibility, representing a step forward to the mechanism-based targeted therapy in DLBCL.


Asunto(s)
Proteínas Inmediatas-Precoces , Linfoma de Células B Grandes Difuso , Humanos , FN-kappa B/genética , Hibridación Fluorescente in Situ , Proteína 1 Similar al Receptor de Interleucina-1/genética , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/genética , Microambiente Tumoral , Factor 1 de Respuesta al Butirato/genética , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/uso terapéutico , Proteínas Supresoras de Tumor/genética
5.
Int J Mol Sci ; 24(6)2023 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-36982508

RESUMEN

The association between Porphyromonas gingivalis (P. gingivalis) and Alzheimer's disease (AD) remains unclear. The major aim of this study was to elucidate the role of genes and molecular targets in P. gingivalis-associated AD. Two Gene Expression Omnibus (GEO) datasets, GSE5281 for AD (n = 84 Alzheimer's, n = 74 control) and GSE9723 (n = 4 P. gingivalis, n = 4 control), were downloaded from the GEO database. Differentially expressed genes (DEGs) were obtained, and genes common to both diseases were drawn. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis was performed from the top 100 genes (50 upregulated and 50 downregulated genes). We then proceeded with CMap analysis to screen for possible small drug molecules targeting these genes. Subsequently, we performed molecular dynamics simulations. A total of 10 common genes (CALD1, HES1, ID3, PLK2, PPP2R2D, RASGRF1, SUN1, VPS33B, WTH3DI/RAB6A, and ZFP36L1) were identified with a p-value < 0.05. The PPI network of the top 100 genes showed UCHL1, SST, CHGB, CALY, and INA to be common in the MCC, DMNC, and MNC domains. Out of the 10 common genes identified, only 1 was mapped in CMap. We found three candidate small drug molecules to be a fit for PLK2, namely PubChem ID: 24971422, 11364421, and 49792852. We then performed molecular docking of PLK2 with PubChem ID: 24971422, 11364421, and 49792852. The best target, 11364421, was used to conduct the molecular dynamics simulations. The results of this study unravel novel genes to P. gingivalis-associated AD that warrant further validation.


Asunto(s)
Enfermedad de Alzheimer , Perfilación de la Expresión Génica , Humanos , Enfermedad de Alzheimer/genética , Porphyromonas gingivalis/genética , Simulación del Acoplamiento Molecular , Modelos Moleculares , Biología Computacional/métodos , Proteínas de Transporte Vesicular/genética , Factor 1 de Respuesta al Butirato/genética , Proteína Fosfatasa 2/genética
6.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(8): 721-726, 2022 Aug.
Artículo en Chino | MEDLINE | ID: mdl-35851086

RESUMEN

Objective To explore the role and mechanism of zinc finger protein 36 like 1 (ZFP36L1) in breast cancer. Methods Sixty breast cancer patients were enrolled in the study. Immunohistochemistry was performed to evaluate the ZFP36L1 expression. Clinicopathological parameters were observed. MCF-7 cells were transfected with overexpressed ZFP36L1 plasmid. The viability of MCF-7 cells was assayed by the 5-ethynyl-2-deoxyuridine (EdU) and MTS assay. The invasion of MCF-7 cells was assessed by TranswellTM assay. Western blot analysis was used to detect the expression of ß-catenin, vimentin, E-cadherin, signal transducer and activator of transcription 3 (STAT3), and phosphorylated STAT3 (p-STAT3). Results ZFP36L1-low expression has been found to be associated with poor prognosis in patients with breast cancer. Moreover, ZFP36L1 overexpression inhibited cell proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) in vitro. Accordingly, the expression of STAT3 and p-STAT3 increased significantly. Conclusion ZFP36L1, as a cancer suppressor gene, inhibits cell proliferation, invasion, and migration through EMT and STAT3 signaling pathway.


Asunto(s)
Neoplasias de la Mama , Transición Epitelial-Mesenquimal , Neoplasias de la Mama/patología , Factor 1 de Respuesta al Butirato/genética , Factor 1 de Respuesta al Butirato/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
7.
J Invest Dermatol ; 142(2): 402-413, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34333017

RESUMEN

Dermal fibroblasts are strategically positioned underneath the basal epidermis layer to support keratinocyte proliferation and extracellular matrix production. In inflammatory conditions, these fibroblasts produce cytokines and chemokines that promote the chemoattraction of immune cells into the dermis and the hyperplasia of the epidermis, two characteristic hallmarks of psoriasis. However, how dermal fibroblasts specifically contribute to psoriasis development remains largely uncharacterized. In this study, we investigated through which cytokines and signaling pathways dermal fibroblasts contribute to the inflammatory features of psoriatic skin. We show that dermal fibroblasts from lesional psoriatic skin are important producers of inflammatory mediators, including IL-6, CXCL8, and CXCL2. This increased cytokine production was found to be regulated by ZFP36 family members ZFP36, ZFP36L1, and ZFP36L2, RNA-binding proteins with mRNA-degrading properties. In addition, the expression of ZFP36 family proteins was found to be reduced in chronic inflammatory conditions that mimic psoriatic lesional skin. Collectively, these results indicate that dermal fibroblasts are important producers of cytokines in psoriatic skin and that reduced expression of ZFP36 members in psoriasis dermal fibroblasts contributes to their inflammatory phenotype.


Asunto(s)
Factor 1 de Respuesta al Butirato/metabolismo , Fibroblastos/metabolismo , Psoriasis/inmunología , Factores de Transcripción/metabolismo , Tristetraprolina/metabolismo , Biopsia , Factor 1 de Respuesta al Butirato/genética , Estudios de Casos y Controles , Epidermis/inmunología , Epidermis/metabolismo , Epidermis/patología , Técnicas de Silenciamiento del Gen , Voluntarios Sanos , Humanos , Mediadores de Inflamación/metabolismo , Queratinocitos/inmunología , Queratinocitos/metabolismo , Psoriasis/patología , Factores de Transcripción/genética , Tristetraprolina/genética
8.
Am J Pathol ; 192(2): 208-225, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34774847

RESUMEN

Zinc finger protein 36 like 1 (ZFP36L1) enhances the turnover of mRNAs containing AU-rich elements (AREs) in their 3'-untranslated regions (3'UTR). The physiological and pathological functions of ZFP36L1 in liver, however, remain largely unknown. Liver-specific ZFP36L1-deficient (Zfp36l1flox/flox/Cre+; L1LKO) mice were generated to investigate the role of ZFP36L1 in liver physiology and pathology. Under normal conditions, the L1LKO mice and their littermate controls (Zfp36l1flox/flox/Cre-; L1FLX) appeared normal. When fed a Lieber-DeCarli liquid diet containing alcohol, L1LKO mice were significantly protected from developing alcohol-induced hepatic steatosis, injury, and inflammation compared with L1FLX mice. Most importantly, fibroblast growth factor 21 (Fgf21) mRNA was significantly increased in the livers of alcohol diet-fed L1LKO mice compared with the alcohol diet-fed L1FLX group. The Fgf21 mRNA contains three AREs in its 3'UTR, and Fgf21 3'UTR was directly regulated by ZFP36L1 in luciferase reporter assays. Steady-state levels of Fgf21 mRNA were significantly decreased by wild-type ZFP36L1, but not by a non-binding zinc finger ZFP36L1 mutant. Finally, wild-type ZFP36L1, but not the ZFP36L1 mutant, bound to the Fgf21 3'UTR ARE RNA probe. These results demonstrate that ZFP36L1 inactivation protects against alcohol-induced hepatic steatosis and liver injury and inflammation, possibly by stabilizing Fgf21 mRNA. These findings suggest that the modulation of ZFP36L1 may be beneficial in the prevention or treatment of human alcoholic liver disease.


Asunto(s)
Regiones no Traducidas 3' , Factor 1 de Respuesta al Butirato/metabolismo , Hígado Graso Alcohólico/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hígado/metabolismo , Estabilidad del ARN , Animales , Factor 1 de Respuesta al Butirato/genética , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Factores de Crecimiento de Fibroblastos/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Hígado/patología , Ratones , Ratones Noqueados , Mutación
9.
Genome Med ; 13(1): 181, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34758847

RESUMEN

BACKGROUND: Genetic studies have been tremendously successful in identifying genomic regions associated with a wide variety of phenotypes, although the success of these studies in identifying causal genes, their variants, and their functional impacts has been more limited. METHODS: We identified 145 genes from IBD-associated genomic loci having endogenous expression within the intestinal epithelial cell compartment. We evaluated the impact of lentiviral transfer of the open reading frame (ORF) of these IBD genes into the HT-29 intestinal epithelial cell line via transcriptomic analyses. By comparing the genes in which expression was modulated by each ORF, as well as the functions enriched within these gene lists, we identified ORFs with shared impacts and their putative disease-relevant biological functions. RESULTS: Analysis of the transcriptomic data for cell lines expressing the ORFs for known causal genes such as HNF4a, IFIH1, and SMAD3 identified functions consistent with what is already known for these genes. These analyses also identified two major clusters of genes: Cluster 1 contained the known IBD causal genes IFIH1, SBNO2, NFKB1, and NOD2, as well as genes from other IBD loci (ZFP36L1, IRF1, GIGYF1, OTUD3, AIRE and PITX1), whereas Cluster 2 contained the known causal gene KSR1 and implicated DUSP16 from another IBD locus. Our analyses highlight how multiple IBD gene candidates can impact on epithelial structure and function, including the protection of the mucosa from intestinal microbiota, and demonstrate that DUSP16 acts a regulator of MAPK activity and contributes to mucosal defense, in part via its regulation of the polymeric immunoglobulin receptor, involved in the protection of the intestinal mucosa from enteric microbiota. CONCLUSIONS: This functional screen, based on expressing IBD genes within an appropriate cellular context, in this instance intestinal epithelial cells, resulted in changes to the cell's transcriptome that are relevant to their endogenous biological function(s). This not only helped in identifying likely causal genes within genetic loci but also provided insight into their biological functions. Furthermore, this work has highlighted the central role of intestinal epithelial cells in IBD pathophysiology, providing a scientific rationale for a drug development strategy that targets epithelial functions in addition to the current therapies targeting immune functions.


Asunto(s)
Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Factor 1 de Respuesta al Butirato/genética , Proteínas Portadoras/genética , Fosfatasas de Especificidad Dual/genética , Células Epiteliales/metabolismo , Microbioma Gastrointestinal , Células HEK293 , Humanos , Inmunoglobulinas , Factor 1 Regulador del Interferón/genética , Mucosa Intestinal/metabolismo , Intestinos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Factores de Transcripción Paired Box/genética , Proteínas Quinasas/genética , Factores de Transcripción/genética , Transcriptoma , Proteasas Ubiquitina-Específicas/genética , Proteína AIRE
10.
Exp Hematol ; 99: 54-64.e7, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34090970

RESUMEN

The mRNA-destabilizing proteins ZFP36L1 and ZFP36L2 are described as mediators of quiescence and play a pivotal role in hematopoietic malignancies. Both genes are mainly classified as tumor suppressor genes as they posttranscriptionally downregulate the expression of oncogenes and contribute to cellular quiescence. Here, we analyzed the role of ZFP36L1 and ZFP36L2 in chronic myeloid leukemia (CML). We found ZFP36L1 and ZFP36L2 expression to be deregulated in patients with CML. By use of in vitro models of tyrosine kinase inhibitor resistance, an increase in ZFP36L1 and ZFP36L2 expression was detected during the development of imatinib resistance. CRISPR/Cas9-derived knockout of ZFP36L1, but not of ZFP36L2, in imatinib-sensitive cells led to decreased proliferation rates in response to tyrosine kinase inhibitor treatment. This effect was also observed in untreated ZFP36L1 knockout cells, albeit to a lower extent. Genomewide gene expression analyses of ZFP36L1 knockout cells revealed differential expression of cell cycle regulators, in particular upregulation of the cell cycle inhibitor CDKN1A. In addition, the 3' untranslated region of CDKN1A was proven to be a direct target of ZFP36L1. This indicates that tumor suppressor genes can also be targeted by ZFP36L1. Hence, ZFP36L1 cannot unambiguously be regarded as a tumor suppressor gene.


Asunto(s)
Factor 1 de Respuesta al Butirato , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Regulación Leucémica de la Expresión Génica , Neoplasias Hematológicas , Leucemia Mielógena Crónica BCR-ABL Positiva , Adulto , Anciano , Anciano de 80 o más Años , Factor 1 de Respuesta al Butirato/biosíntesis , Factor 1 de Respuesta al Butirato/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Humanos , Mesilato de Imatinib/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Masculino , Persona de Mediana Edad
11.
Genome Biol ; 22(1): 7, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397444

RESUMEN

BACKGROUND: Crosstalk between genetic, epigenetic, and immune alterations in upper tract urothelial carcinomas and their role in shaping muscle invasiveness and patient outcome are poorly understood. RESULTS: We perform an integrative genome- and methylome-wide profiling of diverse non-muscle-invasive and muscle-invasive upper tract urothelial carcinomas. In addition to mutations of FGFR3 and KDM6A, we identify ZFP36L1 as a novel, significantly mutated tumor suppressor gene. Overall, mutations of ZFP36 family genes (ZFP36, ZFP36L1, and ZFP36L2) are identified in 26.7% of cases, which display a high mutational load. Unsupervised DNA methylation subtype classification identifies two epi-clusters associated with distinct muscle-invasive status and patient outcome, namely, EpiC-low and EpiC-high. While the former is hypomethylated, immune-depleted, and enriched for FGFR3-mutated, the latter is hypermethylated, immune-infiltrated, and tightly associated with somatic mutations of SWI/SNF genes. CONCLUSIONS: Our study delineates for the first time the key role for convergence between genetic and epigenetic alterations in shaping clinicopathological and immune upper tract urothelial carcinoma features.


Asunto(s)
Carcinoma de Células Transicionales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Vejiga Urinaria/genética , Factor 1 de Respuesta al Butirato/genética , Carcinoma de Células Transicionales/inmunología , Línea Celular Tumoral , Metilación de ADN , Epigenómica , Histona Demetilasas/genética , Humanos , Inmunidad , Mutación , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción/genética , Transcriptoma , Tristetraprolina/genética , Neoplasias de la Vejiga Urinaria/inmunología
12.
J Exp Med ; 218(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33306108

RESUMEN

Cell migration relies on coordinated activity of chemotactic and guidance receptors. Here, we report a specific role for the RNA-binding protein ZFP36L1 in limiting the abundance of molecules involved in the homing of antibody-secreting cells (ASCs) to the bone marrow (BM). In the absence of ZFP36L1, ASCs build up in the spleen and the liver and show diminished accumulation in the BM. ZFP36L1 facilitates migration by directly regulating G protein-coupled receptor kinase 2 (GRK2) and the integrin chains α4 and ß1 in splenic ASCs. Expression of CXCR4 and of the integrins α4 and ß1 is differentially regulated on ASCs produced at the early and late stages of the immune response. Consequently, deletion of the Zfp36l1 gene has a stronger effect on BM accumulation of high-affinity ASCs formed late in the response. Thus, ZFP36L1 is an integral part of the regulatory network controlling gene expression during ASC homing.


Asunto(s)
Células Productoras de Anticuerpos/metabolismo , Médula Ósea/metabolismo , Factor 1 de Respuesta al Butirato/metabolismo , Animales , Células Productoras de Anticuerpos/efectos de los fármacos , Antígenos/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Secuencia de Bases , Factor 1 de Respuesta al Butirato/genética , Recuento de Células , Muerte Celular/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Centro Germinal/citología , Inmunización , Integrinas/metabolismo , Lisofosfolípidos/farmacología , Ratones Endogámicos C57BL , Receptores CXCR4/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato/metabolismo , Bazo/metabolismo
13.
Oxid Med Cell Longev ; 2020: 2067959, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32685086

RESUMEN

Upregulation of Brf1 (TFIIB-related factor 1) and Pol III gene (RNA polymerase III-dependent gene, such as tRNAs and 5S rRNA) activities is associated with cell transformation and tumor development. Alcohol intake causes liver injury, such as steatosis, inflammation, fibrosis, and cirrhosis, which enhances the risk of HCC development. However, the mechanism of alcohol-promoted HCC remains to be explored. We have designed the complementary research system, which is composed of cell lines, an animal model, human samples, and experiments in vivo and in vitro, to carry out this project by using molecular biological, biochemical, and cellular biological approaches. It is a unique system to explore the mechanism of alcohol-associated HCC. Our results indicate that alcohol upregulates Brf1 and Pol III gene (tRNAs and 5S rRNA) transcription in primary mouse hepatocytes, immortalized mouse hepatocyte-AML-12 cells, and engineered human HepG2-ADH cells. Alcohol activates MSK1 to upregulate expression of Brf1 and Pol III genes, while inhibiting MSK1 reduces transcription of Brf1 and Pol III genes in alcohol-treated cells. The inhibitor of MSK1, SB-747651A, decreases the rates of cell proliferation and colony formation. Alcohol feeding promotes liver tumor development of the mouse. These results, for the first time, show the identification of the alcohol-response promoter fragment of the Pol III gene key transcription factor, Brf1. Our studies demonstrate that Brf1 expression is elevated in HCC tumor tissues of mice and humans. Alcohol increases cellular levels of Brf1, resulting in enhancement of Pol III gene transcription in hepatocytes through MSK1. Our mechanism analysis has demonstrated that alcohol-caused high-response fragment of the Brf1 promoter is at p-382/+109bp. The MSK1 inhibitor SB-747651A is an effective reagent to repress alcohol-induced cell proliferation and colony formation, which is a potential pharmaceutical agent. Developing this inhibitor as a therapeutic approach will benefit alcohol-associated HCC patients.


Asunto(s)
Factor 1 de Respuesta al Butirato/genética , Etanol/metabolismo , ARN de Transferencia/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/uso terapéutico , Transcripción Genética/efectos de los fármacos , Animales , Proliferación Celular , Humanos , Ratones , Proteínas Quinasas S6 Ribosómicas 90-kDa/farmacología , Transfección , Regulación hacia Arriba
14.
Nucleic Acids Res ; 48(13): 7371-7384, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32556261

RESUMEN

ZFP36L1, a CCCH-type zinc finger protein, is an RNA-binding protein that participates in controlling cellular mRNA abundance and turnover by posttranscriptional regulation. Here, we demonstrated that ZFP36L1 has an important role in host defense against influenza A virus (IAV) infection. Overexpression of ZFP36L1 reduced IAV replication via translational repression of HA, M and NS RNA segment transcripts. IAV infection upregulated cellular ZFP36L1 expression, and endogenous ZFP36L1 knockdown significantly enhanced IAV replication. ZFP36L1 directly binds to IAV NS1 mRNA in the cytoplasm and blocks the expression and function of NS1 protein. Mutation of CCCH-type zinc finger domains of ZFP36L1 lost its antiviral potential and NS1 mRNA binding. Thus, ZFP36L1 can act as a host innate defense by targeting HA, M and NS mRNA transcripts to suppress viral protein translation.


Asunto(s)
Factor 1 de Respuesta al Butirato/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas no Estructurales Virales/genética , Células A549 , Animales , Sitios de Unión , Factor 1 de Respuesta al Butirato/química , Factor 1 de Respuesta al Butirato/genética , Perros , Células HEK293 , Humanos , Virus de la Influenza A/metabolismo , Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de la Matriz Viral/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
15.
Sci Rep ; 10(1): 6515, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32296077

RESUMEN

DNA methylation could provide a link between environmental, genetic factors and weight control and can modify gene expression pattern. This study aimed to identify genes, which are differentially expressed and methylated depending on adiposity state by evaluating normal weight women and obese women before and after bariatric surgery (BS). We enrolled 24 normal weight (BMI: 22.5 ± 1.6 kg/m2) and 24 obese women (BMI: 43.3 ± 5.7 kg/m2) submitted to BS. Genome-wide methylation analysis was conducted using Infinium Human Methylation 450 BeadChip (threshold for significant CpG sites based on delta methylation level with a minimum value of 5%, a false discovery rate correction (FDR) of q < 0.05 was applied). Expression levels were measured using HumanHT-12v4 Expression BeadChip (cutoff of p ≤ 0.05 and fold change ≥2.0 was used to detect differentially expressed probes). The integrative analysis of both array data identified four genes (i.e. TPP2, PSMG6, ARL6IP1 and FAM49B) with higher methylation and lower expression level in pre-surgery women compared to normal weight women: and two genes (i.e. ZFP36L1 and USP32) that were differentially methylated after BS. These methylation changes were in promoter region and gene body. All genes are related to MAPK cascade, NIK/NF-kappaB signaling, cellular response to insulin stimulus, proteolysis and others. Integrating analysis of DNA methylation and gene expression evidenced that there is a set of genes relevant to obesity that changed after BS. A gene ontology analysis showed that these genes were enriched in biological functions related to adipogenesis, orexigenic, oxidative stress and insulin metabolism pathways. Also, our results suggest that although methylation plays a role in gene silencing, the majority of effects were not correlated.


Asunto(s)
Adiposidad/genética , Cirugía Bariátrica , Metilación de ADN , Obesidad/genética , Transcriptoma , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Factor 1 de Respuesta al Butirato/genética , Factor 1 de Respuesta al Butirato/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Redes y Vías Metabólicas , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Obesidad/cirugía , Periodo Posoperatorio , Periodo Preoperatorio , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
16.
Cancer Res ; 80(2): 219-233, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31551365

RESUMEN

ZFP36L1 is a tandem zinc-finger RNA-binding protein that recognizes conserved adenylate-uridylate-rich elements (ARE) located in 3'untranslated regions (UTR) to mediate mRNA decay. We hypothesized that ZFP36L1 is a negative regulator of a posttranscriptional hub involved in mRNA half-life regulation of cancer-related transcripts. Analysis of in silico data revealed that ZFP36L1 was significantly mutated, epigenetically silenced, and downregulated in a variety of cancers. Forced expression of ZFP36L1 in cancer cells markedly reduced cell proliferation in vitro and in vivo, whereas silencing of ZFP36L1 enhanced tumor cell growth. To identify direct downstream targets of ZFP36L1, systematic screening using RNA pull-down of wild-type and mutant ZFP36L1 as well as whole transcriptome sequencing of bladder cancer cells {plus minus} tet-on ZFP36L1 was performed. A network of 1,410 genes was identified as potential direct targets of ZFP36L1. These targets included a number of key oncogenic transcripts such as HIF1A, CCND1, and E2F1. ZFP36L1 specifically bound to the 3'UTRs of these targets for mRNA degradation, thus suppressing their expression. Dual luciferase reporter assays and RNA electrophoretic mobility shift assays showed that wild-type, but not zinc-finger mutant ZFP36L1, bound to HIF1A 3'UTR and mediated HIF1A mRNA degradation, leading to reduced expression of HIF1A and its downstream targets. Collectively, our findings reveal an indispensable role of ZFP36L1 as a posttranscriptional safeguard against aberrant hypoxic signaling and abnormal cell-cycle progression. SIGNIFICANCE: RNA-binding protein ZFP36L1 functions as a tumor suppressor by regulating the mRNA stability of a number of mRNAs involved in hypoxia and cell-cycle signaling.


Asunto(s)
Neoplasias de la Mama/genética , Factor 1 de Respuesta al Butirato/metabolismo , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias de la Vejiga Urinaria/genética , Regiones no Traducidas 3'/genética , Animales , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Factor 1 de Respuesta al Butirato/genética , Carcinogénesis/genética , Ciclo Celular/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral , Ciclina D1/genética , Factor de Transcripción E2F1/genética , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mutación , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Dedos de Zinc/genética
17.
Cell Stem Cell ; 24(5): 707-723.e8, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30982771

RESUMEN

The identity and degree of heterogeneity of glial progenitors and their contributions to brain tumor malignancy remain elusive. By applying lineage-targeted single-cell transcriptomics, we uncover an unanticipated diversity of glial progenitor pools with unique molecular identities in developing brain. Our analysis identifies distinct transitional intermediate states and their divergent developmental trajectories in astroglial and oligodendroglial lineages. Moreover, intersectional analysis uncovers analogous intermediate progenitors during brain tumorigenesis, wherein oligodendrocyte-progenitor intermediates are abundant, hyper-proliferative, and progressively reprogrammed toward a stem-like state susceptible to further malignant transformation. Similar actively cycling intermediate progenitors are prominent components in human gliomas with distinct driver mutations. We further unveil lineage-driving networks underlying glial fate specification and identify Zfp36l1 as necessary for oligodendrocyte-astrocyte lineage transition and glioma growth. Together, our results resolve the dynamic repertoire of common and divergent glial progenitors during development and tumorigenesis and highlight Zfp36l1 as a molecular nexus for balancing glial cell-fate decision and controlling gliomagenesis.


Asunto(s)
Glioma/genética , Células Madre Neoplásicas/fisiología , Neuroglía/fisiología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Células Madre/fisiología , Transcriptoma/genética , Animales , Biodiversidad , Factor 1 de Respuesta al Butirato/genética , Carcinogénesis , Diferenciación Celular , Reprogramación Celular , Desarrollo Fetal , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados
18.
Cell Death Differ ; 26(12): 2535-2550, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30858608

RESUMEN

RNA polymerase III (Pol-III) transcribes tRNAs and other small RNAs essential for protein synthesis and cell growth. Pol-III is deregulated during carcinogenesis; however, its role in vivo has not been studied. To address this issue, we manipulated levels of Brf1, a Pol-III transcription factor that is essential for recruitment of Pol-III holoenzyme at tRNA genes in vivo. Knockout of Brf1 led to embryonic lethality at blastocyst stage. In contrast, heterozygous Brf1 mice were viable, fertile and of a normal size. Conditional deletion of Brf1 in gastrointestinal epithelial tissues, intestine, liver and pancreas, was incompatible with organ homeostasis. Deletion of Brf1 in adult intestine and liver induced apoptosis. However, Brf1 heterozygosity neither had gross effects in these epithelia nor did it modify tumorigenesis in the intestine or pancreas. Overexpression of BRF1 rescued the phenotypes of Brf1 deletion in intestine and liver but was unable to initiate tumorigenesis. Thus, Brf1 and Pol-III activity are absolutely essential for normal homeostasis during development and in adult epithelia. However, Brf1 overexpression or heterozygosity are unable to modify tumorigenesis, suggesting a permissive, but not driving role for Brf1 in the development of epithelial cancers of the pancreas and gut.


Asunto(s)
Factor 1 de Respuesta al Butirato/deficiencia , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Páncreas/metabolismo , Animales , Factor 1 de Respuesta al Butirato/biosíntesis , Factor 1 de Respuesta al Butirato/genética , Homeostasis , Humanos , Ratones , Factores Asociados con la Proteína de Unión a TATA/biosíntesis , Factores Asociados con la Proteína de Unión a TATA/genética
19.
Haematologica ; 104(8): 1572-1579, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30655376

RESUMEN

In this study we interrogated the DNA methylome of myelofibrosis patients using high-density DNA methylation arrays. We detected 35,215 differentially methylated CpG, corresponding to 10,253 genes, between myelofibrosis patients and healthy controls. These changes were present both in primary and secondary myelofibrosis, which showed no differences between them. Remarkably, most differentially methylated CpG were located outside gene promoter regions and showed significant association with enhancer regions. This aberrant enhancer hypermethylation was negatively correlated with the expression of 27 genes in the myelofibrosis cohort. Of these, we focused on the ZFP36L1 gene and validated its decreased expression and enhancer DNA hypermethylation in an independent cohort of patients and myeloid cell-lines. In vitro reporter assay and 5'-azacitidine treatment confirmed the functional relevance of hyper-methylation of ZFP36L1 enhancer. Furthermore, in vitro rescue of ZFP36L1 expression had an impact on cell proliferation and induced apoptosis in SET-2 cell line indicating a possible role of ZFP36L1 as a tumor suppressor gene in myelofibrosis. Collectively, we describe the DNA methylation profile of myelofibrosis, identifying extensive changes in enhancer elements and revealing ZFP36L1 as a novel candidate tumor suppressor gene.


Asunto(s)
Factor 1 de Respuesta al Butirato/genética , Metilación de ADN , Elementos de Facilitación Genéticos/genética , Epigenómica/métodos , Mielofibrosis Primaria/genética , Apoptosis/efectos de los fármacos , Factor 1 de Respuesta al Butirato/metabolismo , Factor 1 de Respuesta al Butirato/farmacología , Estudios de Casos y Controles , Línea Celular , Proliferación Celular/efectos de los fármacos , Epigénesis Genética , Genes Supresores de Tumor , Humanos
20.
Nat Commun ; 10(1): 77, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622281

RESUMEN

Osteoarthritis (OA) is a whole-joint disease characterized by cartilage destruction and other whole-joint pathological changes. There is currently no effective disease-modifying therapy. Here we investigate the post-transcriptional mRNA regulation of OA-modulating proteins in chondrocytes and show that the ZFP36 family member, ZFP36L1, is specifically upregulated in OA chondrocytes and OA cartilage of humans and mice. Adenovirus-mediated overexpression of ZFP36L1 alone in mouse knee-joint tissue does not modulate OA pathogenesis. However, genetic ablation or silencing of Zfp36l1 significantly abrogates experimental OA in mice. Knockdown of Zfp36l1 increases the mRNA expression of two heat shock protein 70 (HSP70) family members, which act as its direct targets. Furthermore, overexpression of HSPA1A in joint tissues protects mice against experimental OA by inhibiting chondrocyte apoptosis. Our results indicate that the RNA-binding protein, ZFP36L1, regulates HSP70 family members that appear to protect against OA pathogenesis by inhibiting chondrocyte apoptosis.


Asunto(s)
Factor 1 de Respuesta al Butirato/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas Nucleares/metabolismo , Osteoartritis/patología , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis , Factor 1 de Respuesta al Butirato/genética , Cartílago Articular/citología , Cartílago Articular/patología , Células Cultivadas , Condrocitos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Articulación de la Rodilla , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Osteoartritis/genética , Osteoartritis/cirugía , Cultivo Primario de Células , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...