Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.938
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(17): 4579-4592.e24, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34297925

RESUMEN

Antibacterial agents target the products of essential genes but rarely achieve complete target inhibition. Thus, the all-or-none definition of essentiality afforded by traditional genetic approaches fails to discern the most attractive bacterial targets: those whose incomplete inhibition results in major fitness costs. In contrast, gene "vulnerability" is a continuous, quantifiable trait that relates the magnitude of gene inhibition to the effect on bacterial fitness. We developed a CRISPR interference-based functional genomics method to systematically titrate gene expression in Mycobacterium tuberculosis (Mtb) and monitor fitness outcomes. We identified highly vulnerable genes in various processes, including novel targets unexplored for drug discovery. Equally important, we identified invulnerable essential genes, potentially explaining failed drug discovery efforts. Comparison of vulnerability between the reference and a hypervirulent Mtb isolate revealed incomplete conservation of vulnerability and that differential vulnerability can predict differential antibacterial susceptibility. Our results quantitatively redefine essential bacterial processes and identify high-value targets for drug development.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Mycobacterium tuberculosis/genética , Aminoacil-ARNt Sintetasas/metabolismo , Antituberculosos/farmacología , Teorema de Bayes , Evolución Biológica , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/efectos de los fármacos , ARN Guía de Kinetoplastida/genética
2.
Nature ; 585(7825): 453-458, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908306

RESUMEN

Heterochromatin that depends on histone H3 lysine 9 methylation (H3K9me) renders embedded genes transcriptionally silent1-3. In the fission yeast Schizosaccharomyces pombe, H3K9me heterochromatin can be transmitted through cell division provided the counteracting demethylase Epe1 is absent4,5. Heterochromatin heritability might allow wild-type cells under certain conditions to acquire epimutations, which could influence phenotype through unstable gene silencing rather than DNA change6,7. Here we show that heterochromatin-dependent epimutants resistant to caffeine arise in fission yeast grown with threshold levels of caffeine. Isolates with unstable resistance have distinct heterochromatin islands with reduced expression of embedded genes, including some whose mutation confers caffeine resistance. Forced heterochromatin formation at implicated loci confirms that resistance results from heterochromatin-mediated silencing. Our analyses reveal that epigenetic processes promote phenotypic plasticity, letting wild-type cells adapt to unfavourable environments without genetic alteration. In some isolates, subsequent or coincident gene-amplification events augment resistance. Caffeine affects two anti-silencing factors: Epe1 is downregulated, reducing its chromatin association, and a shortened isoform of Mst2 histone acetyltransferase is expressed. Thus, heterochromatin-dependent epimutation provides a bet-hedging strategy allowing cells to adapt transiently to insults while remaining genetically wild type. Isolates with unstable caffeine resistance show cross-resistance to antifungal agents, suggesting that related heterochromatin-dependent processes may contribute to resistance of plant and human fungal pathogens to such agents.


Asunto(s)
Farmacorresistencia Fúngica/genética , Silenciador del Gen , Heterocromatina/genética , Heterocromatina/metabolismo , Schizosaccharomyces/genética , Cafeína/farmacología , Farmacorresistencia Fúngica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Heterocromatina/efectos de los fármacos , Histona Acetiltransferasas/metabolismo , Proteínas Nucleares/metabolismo , Fenotipo , Schizosaccharomyces/citología , Schizosaccharomyces/efectos de los fármacos , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo
3.
PLoS Biol ; 18(6): e3000722, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32569301

RESUMEN

Inflammation and infection can trigger local tissue Na+ accumulation. This Na+-rich environment boosts proinflammatory activation of monocyte/macrophage-like cells (MΦs) and their antimicrobial activity. Enhanced Na+-driven MΦ function requires the osmoprotective transcription factor nuclear factor of activated T cells 5 (NFAT5), which augments nitric oxide (NO) production and contributes to increased autophagy. However, the mechanism of Na+ sensing in MΦs remained unclear. High extracellular Na+ levels (high salt [HS]) trigger a substantial Na+ influx and Ca2+ loss. Here, we show that the Na+/Ca2+ exchanger 1 (NCX1, also known as solute carrier family 8 member A1 [SLC8A1]) plays a critical role in HS-triggered Na+ influx, concomitant Ca2+ efflux, and subsequent augmented NFAT5 accumulation. Moreover, interfering with NCX1 activity impairs HS-boosted inflammatory signaling, infection-triggered autolysosome formation, and subsequent antibacterial activity. Taken together, this demonstrates that NCX1 is able to sense Na+ and is required for amplifying inflammatory and antimicrobial MΦ responses upon HS exposure. Manipulating NCX1 offers a new strategy to regulate MΦ function.


Asunto(s)
Macrófagos/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Sodio/metabolismo , Empalme Alternativo/genética , Animales , Calcio/metabolismo , Espacio Extracelular/metabolismo , Silenciador del Gen/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Iones , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Óxido Nítrico/biosíntesis , Células RAW 264.7 , Cloruro de Sodio/farmacología
4.
Nature ; 547(7664): 463-467, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28682306

RESUMEN

Heterochromatic DNA domains have important roles in the regulation of gene expression and maintenance of genome stability by silencing repetitive DNA elements and transposons. From fission yeast to mammals, heterochromatin assembly at DNA repeats involves the activity of small noncoding RNAs (sRNAs) associated with the RNA interference (RNAi) pathway. Typically, sRNAs, originating from long noncoding RNAs, guide Argonaute-containing effector complexes to complementary nascent RNAs to initiate histone H3 lysine 9 di- and trimethylation (H3K9me2 and H3K9me3, respectively) and the formation of heterochromatin. H3K9me is in turn required for the recruitment of RNAi to chromatin to promote the amplification of sRNA. Yet, how heterochromatin formation, which silences transcription, can proceed by a co-transcriptional mechanism that also promotes sRNA generation remains paradoxical. Here, using Clr4, the fission yeast Schizosaccharomyces pombe homologue of mammalian SUV39H H3K9 methyltransferases, we design active-site mutations that block H3K9me3, but allow H3K9me2 catalysis. We show that H3K9me2 defines a functionally distinct heterochromatin state that is sufficient for RNAi-dependent co-transcriptional gene silencing at pericentromeric DNA repeats. Unlike H3K9me3 domains, which are transcriptionally silent, H3K9me2 domains are transcriptionally active, contain modifications associated with euchromatic transcription, and couple RNAi-mediated transcript degradation to the establishment of H3K9me domains. The two H3K9me states recruit reader proteins with different efficiencies, explaining their different downstream silencing functions. Furthermore, the transition from H3K9me2 to H3K9me3 is required for RNAi-independent epigenetic inheritance of H3K9me domains. Our findings demonstrate that H3K9me2 and H3K9me3 define functionally distinct chromatin states and uncover a mechanism for the formation of transcriptionally permissive heterochromatin that is compatible with its broadly conserved role in sRNA-mediated genome defence.


Asunto(s)
Silenciador del Gen , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/química , Histonas/metabolismo , Interferencia de ARN , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Silenciador del Gen/efectos de los fármacos , Heterocromatina/química , N-Metiltransferasa de Histona-Lisina , Ácidos Hidroxámicos/farmacología , Metilación/efectos de los fármacos , Metiltransferasas/metabolismo , Mutación , Proteínas Represoras/metabolismo , Schizosaccharomyces/efectos de los fármacos , Proteínas de Schizosaccharomyces pombe/metabolismo , Transcripción Genética/efectos de los fármacos
5.
J Am Chem Soc ; 144(7): 3174-3181, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35143189

RESUMEN

The therapeutic use of small interfering RNAs (siRNAs) as gene regulation agents has been limited by their poor stability and delivery. Although arranging siRNAs into a spherical nucleic acid (SNA) architecture to form siRNA-SNAs increases their stability and uptake, prototypical siRNA-SNAs consist of a hybridized architecture that causes guide strand dissociation from passenger strands, which limits the delivery of active siRNA duplexes. In this study, a new SNA design that directly attaches both siRNA strands to the SNA core through a single hairpin-shaped molecule to prevent guide strand dissociation is introduced and investigated. This hairpin-like architecture increases the number of siRNA duplexes that can be loaded onto an SNA by 4-fold compared to the original hybridized siRNA-SNA architecture. As a result, the hairpin-like siRNA-SNAs exhibit a 6-fold longer half-life in serum and decreased cytotoxicity. In addition, the hairpin-like siRNA-SNA produces more durable gene knockdown than the hybridized siRNA-SNA. This study shows how the chemistry used to immobilize siRNA on nanoparticles can markedly enhance biological function, and it establishes the hairpin-like architecture as a next-generation SNA construct that will be useful in life science and medical research.


Asunto(s)
Ácidos Nucleicos Inmovilizados/farmacología , ARN Interferente Pequeño/farmacología , Línea Celular Tumoral , Silenciador del Gen/efectos de los fármacos , Oro/química , Humanos , Ácidos Nucleicos Inmovilizados/química , Ácidos Nucleicos Inmovilizados/toxicidad , Secuencias Invertidas Repetidas , Nanopartículas del Metal/química , Nanopartículas del Metal/toxicidad , Hibridación de Ácido Nucleico , ARN Interferente Pequeño/química , ARN Interferente Pequeño/toxicidad
6.
Nature ; 537(7618): 63-68, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27533040

RESUMEN

Hypermethylation of the promoters of tumour suppressor genes represses transcription of these genes, conferring growth advantages to cancer cells. How these changes arise is poorly understood. Here we show that the activity of oxygen-dependent ten-eleven translocation (TET) enzymes is reduced by tumour hypoxia in human and mouse cells. TET enzymes catalyse DNA demethylation through 5-methylcytosine oxidation. This reduction in activity occurs independently of hypoxia-associated alterations in TET expression, proliferation, metabolism, hypoxia-inducible factor activity or reactive oxygen species, and depends directly on oxygen shortage. Hypoxia-induced loss of TET activity increases hypermethylation at gene promoters in vitro. In patients, tumour suppressor gene promoters are markedly more methylated in hypoxic tumour tissue, independent of proliferation, stromal cell infiltration and tumour characteristics. Our data suggest that up to half of hypermethylation events are due to hypoxia, with these events conferring a selective advantage. Accordingly, increased hypoxia in mouse breast tumours increases hypermethylation, while restoration of tumour oxygenation abrogates this effect. Tumour hypoxia therefore acts as a novel regulator of DNA methylation.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/deficiencia , Oxigenasas de Función Mixta/deficiencia , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Hipoxia Tumoral/fisiología , 5-Metilcitosina/metabolismo , Animales , Proliferación Celular , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Silenciador del Gen/efectos de los fármacos , Genes Supresores de Tumor , Humanos , Masculino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Oxidación-Reducción/efectos de los fármacos , Oxígeno/farmacología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células del Estroma/patología , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/genética
7.
Mol Cell ; 55(3): 347-60, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24999238

RESUMEN

Polycomb group (PcG) proteins are required for normal differentiation and development and are frequently deregulated in cancer. PcG proteins are involved in gene silencing; however, their role in initiation and maintenance of transcriptional repression is not well defined. Here, we show that knockout of the Polycomb repressive complex 2 (PRC2) does not lead to significant gene expression changes in mouse embryonic stem cells (mESCs) and that it is dispensable for initiating silencing of target genes during differentiation. Transcriptional inhibition in mESCs is sufficient to induce genome-wide ectopic PRC2 recruitment to endogenous PcG target genes found in other tissues. PRC2 binding analysis shows that it is restricted to nucleosome-free CpG islands (CGIs) of untranscribed genes. Our results show that it is the transcriptional state that governs PRC2 binding, and we propose that it binds by default to nontranscribed CGI genes to maintain their silenced state and to protect cell identity.


Asunto(s)
Islas de CpG , Células Madre Embrionarias/metabolismo , Silenciador del Gen , Nucleosomas/genética , Nucleosomas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Diclororribofuranosil Benzoimidazol/farmacología , Diterpenos/farmacología , Epigénesis Genética , Compuestos Epoxi/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Silenciador del Gen/efectos de los fármacos , Genoma , Ratones , Fenantrenos/farmacología , Unión Proteica/genética , Unión Proteica/fisiología
8.
Proc Natl Acad Sci U S A ; 116(1): 233-238, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30559205

RESUMEN

The composition of the gut microbiota is largely determined by environmental factors including the host diet. Dietary components are believed to influence the composition of the gut microbiota by serving as nutrients to a subset of microbes, thereby favoring their expansion. However, we now report that dietary fructose and glucose, which are prevalent in the Western diet, specifically silence a protein that is necessary for gut colonization, but not for utilization of these sugars, by the human gut commensal Bacteroides thetaiotaomicron Silencing by fructose and glucose requires the 5' leader region of the mRNA specifying the protein, designated Roc for regulator of colonization. Incorporation of the roc leader mRNA in front of a heterologous gene was sufficient for fructose and glucose to turn off expression of the corresponding protein. An engineered strain refractory to Roc silencing by these sugars outcompeted wild-type B. thetaiotaomicron in mice fed a diet rich in glucose and sucrose (a disaccharide composed of glucose and fructose), but not in mice fed a complex polysaccharide-rich diet. Our findings underscore a role for dietary sugars that escape absorption by the host intestine and reach the microbiota: regulation of gut colonization by beneficial microbes independently of supplying nutrients to the microbiota.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Bacteroides thetaiotaomicron/efectos de los fármacos , Carbohidratos de la Dieta/farmacología , Azúcares de la Dieta/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Proteínas Bacterianas/metabolismo , Fructosa/administración & dosificación , Fructosa/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Glucosa/administración & dosificación , Glucosa/farmacología , Ratones , Polisacáridos/administración & dosificación , Polisacáridos/farmacología , Simbiosis/efectos de los fármacos
9.
Int J Mol Sci ; 23(2)2022 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-35054871

RESUMEN

Glioblastoma (GBM) is the most malignant glioma with an extremely poor prognosis. It is characterized by high vascularization and its growth depends on the formation of new blood vessels. We have previously demonstrated that TRPML2 mucolipin channel expression increases with the glioma pathological grade. Herein by ddPCR and Western blot we found that the silencing of TRPML2 inhibits expression of the VEGFA/Notch2 angiogenic pathway. Moreover, the VEGFA/Notch2 expression increased in T98 and U251 cells stimulated with the TRPML2 agonist, ML2-SA1, or by enforced-TRPML2 levels. In addition, changes in TRPML2 expression or ML2-SA1-induced stimulation, affected Notch2 activation and VEGFA release. An increased invasion capability, associated with a reduced VEGF/VEGFR2 expression and increased vimentin and CD44 epithelial-mesenchymal transition markers in siTRPML2, but not in enforced-TRPML2 or ML2-SA1-stimulated glioma cells, was demonstrated. Furthermore, an increased sensitivity to Doxorubicin cytotoxicity was demonstrated in siTRPML2, whereas ML2-SA1-treated GBM cells were more resistant. The role of proteasome in Cathepsin B-dependent and -independent pRB degradation in siTRPML2 compared with siGLO cells was studied. Finally, through Kaplan-Meier analysis, we found that high TRPML2 mRNA expression strongly correlates with short survival in GBM patients, supporting TRPML2 as a negative prognostic factor in GBM patients.


Asunto(s)
Glioblastoma/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Receptor Notch2/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Canales de Potencial de Receptor Transitorio/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Catepsina B/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Pronóstico , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Canales de Potencial de Receptor Transitorio/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Carcinogenesis ; 42(1): 136-147, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32710611

RESUMEN

Hexavalent chromium [Cr(VI)] is a potent human lung carcinogen. Multiple mechanisms have been proposed that contribute to Cr(VI)-induced lung carcinogenesis including oxidative stress, DNA damage, genomic instability and epigenetic modulation. However, the molecular mechanisms and pathways mediating Cr(VI) carcinogenicity have not been fully elucidated. Hedgehog (Hh) signaling is a key pathway that plays important roles in the formation of multiple tissues during embryogenesis and in the maintenance of stem cell populations in adults. Dysregulation of Hh signaling pathway has been reported in many human cancers. Here, we report a drastic reduction in both mRNA and protein levels of hedgehog-interacting protein (HHIP), a downstream target and a negative regulator of Hh signaling, in Cr(VI)-transformed cells. These findings point to a potential role of Hh signaling in Cr(VI)-induced malignant transformation and lung carcinogenesis. Cr(VI)-transformed cells exhibited DNA hypermethylation and silencing histone marks in the promoter region of HHIP, indicating that an epigenetic mechanism mediates Cr(VI)-induced silencing of HHIP. In addition, the major targets of Hh signaling (GLI1-3 and PTCH1) were significantly increased in Cr(VI)-transformed cells, suggesting an aberrant activation of Hh signaling in these cells. Moreover, ectopically expressing HHIP not only suppressed Hh signaling but also inhibited cell proliferation and anchorage-independent growth in Cr(VI)-transformed cells. In conclusion, these findings establish a novel regulatory mechanism underlying Cr(VI)-induced lung carcinogenesis and provide new insights for developing a better diagnostic and prognostic strategy for Cr(VI)-related human lung cancer.


Asunto(s)
Proteínas Portadoras/genética , Transformación Celular Neoplásica/genética , Cromo/toxicidad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/inducido químicamente , Glicoproteínas de Membrana/genética , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/patología , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Transformación Celular Neoplásica/inducido químicamente , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Silenciador del Gen/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Transducción de Señal/efectos de los fármacos
11.
Plant J ; 102(1): 68-84, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31733119

RESUMEN

Repetitive DNA sequences and some genes are epigenetically repressed by transcriptional gene silencing (TGS). When genetic mutants are not available or problematic to use, TGS can be suppressed by chemical inhibitors. However, informed use of epigenetic inhibitors is partially hampered by the absence of any systematic comparison. In addition, there is emerging evidence that epigenetic inhibitors cause genomic instability, but the nature of this damage and its repair remain unclear. To bridge these gaps, we compared the effects of 5-azacytidine (AC), 2'-deoxy-5-azacytidine (DAC), zebularine and 3-deazaneplanocin A (DZNep) on TGS and DNA damage repair. The most effective inhibitor of TGS was DAC, followed by DZNep, zebularine and AC. We confirmed that all inhibitors induce DNA damage and suggest that this damage is repaired by multiple pathways with a critical role of homologous recombination and of the SMC5/6 complex. A strong positive link between the degree of cytidine analog-induced DNA demethylation and the amount of DNA damage suggests that DNA damage is an integral part of cytidine analog-induced DNA demethylation. This helps us to understand the function of DNA methylation in plants and opens the possibility of using epigenetic inhibitors in biotechnology.


Asunto(s)
Daño del ADN , Epigénesis Genética , Silenciador del Gen , Adenosina/análogos & derivados , Adenosina/farmacología , Arabidopsis/genética , Azacitidina/farmacología , Aberraciones Cromosómicas/efectos de los fármacos , Citidina/análogos & derivados , Citidina/farmacología , Daño del ADN/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Decitabina/farmacología , Epigénesis Genética/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Heterocromatina/efectos de los fármacos , Interferencia de ARN/efectos de los fármacos , Secuencias Repetidas en Tándem/efectos de los fármacos
12.
J Cell Physiol ; 236(9): 6520-6533, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33576499

RESUMEN

Pre-eclampsia (PE) is a pregnancy-related disorder that occurs after 20 weeks of gestation. It seriously affects the health of maternity and the fetus. However, the pathogenesis of PE is still unknown. Decidualization deficiency is considered a contributing factor to the development of PE. CTP synthetase (CTPS) which is the rate-limiting enzyme in the CTP de novo biosynthesis, is essential for nucleic acid synthesis and cellular energy metabolism, and often appears as cytoophidium in many cell types. Here, we found that the expression of CTPS was significantly downregulated in decidual tissues of patients with severe PE compared with healthy pregnant women. During in vitro decidualization, changes in CTPS were accompanied by opposite fluctuation of the AMPK signaling pathway. Moreover, the downregulation of CTPS by glutamine analogs or CTPS small interfering RNA inhibited the decidualization process and the AMPK signaling pathway. Investigating the underlying mechanism of action by co-immunoprecipitation coupled with mass spectrometry showed that CTPS interacted with ATP synthase (ATPS) and maintained the content of ATP on Day 3 of decidualization. However, when combined with mitochondrial stress protein STRESS-70 instead of ATPS, the concentration of ATP on Day 6 of induction was reduced. Corresponding to this, CTPS was mainly distributes in the cytoplasm on Day 3 of induction, while it appeared both in the cytoplasm and the nucleus on Day 6 in decidualized cells, which was similar to that in cells before induction. In summary, we believe that CTPS plays an important role in decidualization by participating in energy metabolism. Abnormal expression of CTPS in decidualization would lead to abnormal decidualization and consequently result in the occurrence of PE.


Asunto(s)
Ligasas de Carbono-Nitrógeno/metabolismo , Decidua/enzimología , Regulación hacia Abajo , Metabolismo Energético , Preeclampsia/enzimología , Adenilato Quinasa/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Diazooxonorleucina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Endometrio/patología , Metabolismo Energético/efectos de los fármacos , Femenino , Silenciador del Gen/efectos de los fármacos , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Embarazo , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
13.
J Cell Physiol ; 236(9): 6312-6327, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33561320

RESUMEN

Bcr-Abl independent resistance to tyrosine kinase inhibitor (TKI) is a crucial factor lead to relapse or acute leukemia transformation in chronic myeloid leukemia (CML). However, its mechanism is still unclear. Herein, we found that of nine common protein kinases C (PKCs), PKC-ß overexpression was significantly related with TKI resistance. Blockage of its expression in CD34+ cells and CML cell lines increased sensitivity to imatinib. Then, eighty-four leukemia related genes were compared between TKI-resistant CML cell lines with PKC-ß silenced or not. Gene Ontology term and Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that Arachidonate 5-lipoxygenase (Alox5) and its relative pathway mainly participated in the resistance induced by PKC-ß overexpression. It's also observed that Alox5 was increased not only in bone marrow biopsy but also in CD34+ cells derived from IM-resistant CML patients. The signaling pathway exploration indicated that ERK1/2 pathway mediates Alox5 upregulation by PKC-ß. Meanwhile, we also proved that Alox5 induces TKI-insensitivity in CML through inactivation of PTEN. In vivo experiment, PKC-ß elective inhibitor LY333531 prolonged survival time in CML-PDX mice model. In conclusion, targeted on PKC-ß overexpression might be a novel therapy mechanism to overcome TKI-resistance in CML.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteína Quinasa C beta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Proteínas de Fusión bcr-abl/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Mesilato de Imatinib/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Persona de Mediana Edad , Mutación/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C beta/antagonistas & inhibidores , Análisis de Supervivencia , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
14.
EMBO J ; 36(22): 3309-3324, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29046334

RESUMEN

The maxi-anion channels (MACs) are expressed in cells from mammals to amphibians with ~60% exhibiting a phenotype called Maxi-Cl. Maxi-Cl serves as the most efficient pathway for regulated fluxes of inorganic and organic anions including ATP However, its molecular entity has long been elusive. By subjecting proteins isolated from bleb membranes rich in Maxi-Cl activity to LC-MS/MS combined with targeted siRNA screening, CRISPR/Cas9-mediated knockout, and heterologous overexpression, we identified the organic anion transporter SLCO2A1, known as a prostaglandin transporter (PGT), as a key component of Maxi-Cl. Recombinant SLCO2A1 exhibited Maxi-Cl activity in reconstituted proteoliposomes. When SLCO2A1, but not its two disease-causing mutants, was heterologously expressed in cells which lack endogenous SLCO2A1 expression and Maxi-Cl activity, Maxi-Cl currents became activated. The charge-neutralized mutant became weakly cation-selective with exhibiting a smaller single-channel conductance. Slco2a1 silencing in vitro and in vivo, respectively, suppressed the release of ATP from swollen C127 cells and from Langendorff-perfused mouse hearts subjected to ischemia-reperfusion. These findings indicate that SLCO2A1 is an essential core component of the ATP-conductive Maxi-Cl channel.


Asunto(s)
Canales Iónicos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Fraccionamiento Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Dinoprostona/farmacología , Femenino , Eliminación de Gen , Silenciador del Gen/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Mutación/genética , Proteolípidos/efectos de los fármacos , Proteolípidos/metabolismo , Proteínas Recombinantes/metabolismo , Daño por Reperfusión/patología
15.
Biochem Biophys Res Commun ; 556: 163-170, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33845307

RESUMEN

Although efficient methods of gene silencing have been established in eukaryotes, many different techniques are still used in bacteria due to the lack of a standardized tool. Here, we developed a convenient and efficient method to downregulate the expression of a specific gene using ∼140 nucleotide RNA with a 24-nucleotide antisense region from an arabinose-inducible expression plasmid by taking Escherichia coli lacZ and phoA genes encoding ß-galactosidase and alkaline phosphatase, respectively, as target genes to evaluate the model. We examined the antisense RNA (asRNA) design, including targeting position, uORF stability elements at the 5'-end, and Hfq-binding module at the 3'-end, and inducer amount required to obtain effective experimental conditions for gene silencing. Furthermore, we constructed multiplexed dual-acting asRNA genes in the plasmid, which were transcribed as polycistronic RNA and were able to knockdown multiple target genes simultaneously. We observed the highest inhibition level of 98.6% when lacZ was targeted using the pMKN104 asRNA expression plasmid, containing a five times stronger PBAD -10 promoter sequence with no requirement of the Hfq protein for repression. These features allow the system to be utilized as an asRNA expression platform in many bacteria, besides E. coli, for gene regulation.


Asunto(s)
Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Silenciador del Gen , Genes/genética , ARN sin Sentido/genética , Arabinosa/metabolismo , Arabinosa/farmacología , Secuencia de Bases , Codón Iniciador/genética , Escherichia coli/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Genes/efectos de los fármacos , Genes Reporteros , Plásmidos/efectos de los fármacos , Plásmidos/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN sin Sentido/biosíntesis
16.
Biochem Biophys Res Commun ; 556: 72-78, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33839417

RESUMEN

Even though long non-coding RNA (lncRNA) MEG8 plays vital roles in carcinogenesis of malignances, its roles and mechanisms in hemangioma remain unknown. Therefore, we evaluate the oncogenic roles of MEG8 in hemangioma. Small interfering RNA (siRNA)-mediated depletion of MEG8 inhibited the proliferation and increased MDA level in human hemangioma endothelial cells (HemECs). The inhibitors of ferroptosis (ferrostatin-1 and liproxstatin-1) abolished the MEG8 silence induced cell viability loss. Knockdown of MEG8 increased the miR-497-5p expression and reduced the mRNA and protein levels of NOTCH2. Using a dual-luciferase assay, we confirmed the binding between MEG8 and miR-497-5p, and between the miR-497-5p and 3'UTR of NOTCH2. We further found that silencing MEG8 significantly decreased the expressions of SLC7A11 and GPX4 both in mRNA and protein level and had no effect on the level of AIFM2. Importantly, blocking miR-497-5p abrogated the effects of MEG8 loss on cell viability, MDA level and expression levels of NOTCH2, SLC7A11 and GPX4 in HemECs. Taken together, our results suggested that knockdown of long non-coding RNA MEG8 inhibited the proliferation and induced the ferroptosis of hemangioma endothelial cells by regulating miR-497-5p/NOTCH2 axis.


Asunto(s)
Células Endoteliales/metabolismo , Ferroptosis/genética , Silenciador del Gen , Hemangioma/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Receptor Notch2/genética , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Secuencia de Bases , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Ciclohexilaminas/farmacología , Regulación hacia Abajo , Células Endoteliales/patología , Ferroptosis/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , MicroARNs/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fenilendiaminas/farmacología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Quinoxalinas/farmacología , ARN Largo no Codificante/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Receptor Notch2/biosíntesis , Receptor Notch2/metabolismo , Compuestos de Espiro/farmacología
17.
Plant Physiol ; 183(3): 1126-1144, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32332089

RESUMEN

Ovule formation is essential for realizing crop yield because it determines seed number. The underlying molecular mechanism, however, remains elusive. Here, we show that cell wall invertase (CWIN) functions as a positive regulator of ovule initiation in Arabidopsis (Arabidopsis thaliana). In situ hybridization revealed that CWIN2 and CWIN4 were expressed at the placenta region where ovule primordia initiated. Specific silencing of CWIN2 and CWIN4 using targeted artificial microRNA driven by an ovule-specific SEEDSTICK promoter (pSTK) resulted in a substantial reduction of CWIN transcript and activity, which blocked ovule initiation and aggravated ovule abortion. There was no induction of carbon (C) starvation genes in the transgenic lines, and supplementing newly forming floral buds with extra C failed to recover the ovule phenotype. This indicates that suppression of CWIN did not lead to C starvation. A group of hexose transporters was downregulated in the transgenic plants. Among them, two representative ones were spatially coexpressed with CWIN2 and CWIN4, suggesting a coupling between CWIN and hexose transporters for ovule initiation. RNA-sequencing analysis identified differentially expressed genes encoding putative extracellular receptor-like kinases, MADS-box transcription factors, including STK, and early auxin response genes in response to CWIN-silencing. Our data demonstrate the essential role of CWIN in ovule initiation, which is most likely to occur through sugar signaling instead of C nutrient contribution. We propose that CWIN-mediated sugar signaling may be perceived by, and transmitted through, hexose transporters or receptor-like kinases to regulate ovule formation by modulating downstream auxin signaling and MADS-box transcription factors.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Carbono/metabolismo , Pared Celular/enzimología , Óvulo Vegetal/crecimiento & desarrollo , Transducción de Señal , Azúcares/metabolismo , beta-Fructofuranosidasa/metabolismo , Arabidopsis/citología , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Pared Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Genes de Plantas , Ácidos Indolacéticos/farmacología , Inflorescencia/efectos de los fármacos , Inflorescencia/enzimología , Meristema/efectos de los fármacos , Meristema/enzimología , Óvulo Vegetal/efectos de los fármacos , Óvulo Vegetal/enzimología , Óvulo Vegetal/genética , Fenotipo , Plantas Modificadas Genéticamente , ARN Mensajero/genética , ARN Mensajero/metabolismo , Semillas/genética , Transducción de Señal/efectos de los fármacos
18.
Cytokine ; 142: 155495, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33765653

RESUMEN

MicroRNA (MiR)-135b and its mediated Wnt/ß-catenin signaling pathway are involved in human malignancies. However, their roles in multiple myeloma (MM) remained poorly understood. Our study aimed to uncover their roles in MM. MiR-135b and Versican expressions were measured using quantitative real-time polymerase chain reaction (qRT-PCR). MM cell proliferation, apoptosis, migration and invasion were detected by cell counting kit-8 (CCK-8) assay, flow cytometry, wound healing assay and transwell assay, respectively. Relative expression of Wnt/ß-catenin signaling pathway-related protein was quantified by Western blot. MiR-135b was upregulated in the serum of MM patients, and miR-135b upregulation promoted MM cell proliferation, migration and invasion but suppressed apoptosis. Also, miR-135b upregulation promoted activation of Wnt/ß-catenin signaling pathway. However, downregulation of miR-135b caused an opposite effect. After incubating cells with miR-135b inhibitor and Wnt/ß-catenin signaling pathway agonist Lithium chloride (LiCl), which reversed the effects of downregulating miR-135b. Versican is the downstream effector of the Wnt/ß-catenin signaling pathway, and its silencing reversed the effects of LiCl on MM cells. In conclusion, miR-135b and its mediated Wnt/ß-catenin signaling pathway promoted proliferation, migration and invasion but suppressed apoptosis of MM cells through regulating Versican, providing a possible treatment for MM.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Mieloma Múltiple/genética , Versicanos/metabolismo , Vía de Señalización Wnt , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Cloruro de Litio/farmacología , MicroARNs/genética , Mieloma Múltiple/patología , Invasividad Neoplásica , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
19.
J Recept Signal Transduct Res ; 41(3): 245-254, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32878540

RESUMEN

BACKGROUND: Cytokine therapies show promise in treating renal cell carcinoma (RCC). Transforming growth factor beta 1 (TGF-ß1) is a cytokine whose downstream Smad2/3 signaling activity is inhibited by the protein phosphatase Mg2+/Mn2+-dependent 1 A (PPM1A). Here, we hypothesized that PPM1A may be involved in suppressing RCC cell aggressiveness through its negative regulation of Smad2/3. METHODS: We quantified PPM1A expression from RCC tumors and matching healthy tissue and performed a Kaplan-Meier survival analysis. In silico analysis on PPM1A was performed using Cancer Genome Atlas-Kidney Renal Clear Cell Carcinoma and Clinical Proteomic Tumor Analysis Consortium RCC cohort data. We tested four RCC cell lines and selected the ACNH and A498 cells lines as expressing the greatest PPM1A levels. We assayed the effects of RNAi-mediated PPM1A silencing on invasiveness, proliferation, colony formation, and Smad2/3 phosphorylation in untreated and TGF-ß1-stimulated ACNH and A498 cells. A nude mouse A498 xenograft tumor model was constructed to validate PPM1A's effects in vivo. RESULTS: PPM1A levels are reduced in RCC tumors and are negatively correlated with RCC grade and stage. Below-median PPM1A expression is associated with reduced overall survival in RCC patients. PPM1A silencing promoted cellular invasiveness, proliferation, colony formation, and Smad2/3 phosphorylation under TGF-ß1-stimulated conditions but not under untreated conditions. These effects of PPM1A were shown to be dependent on Smad2/3. Intratumor PPM1A overexpression inhibited A498 xenograft tumor growth. CONCLUSIONS: This study establishes a direct link between PPM1A's suppression of Smad2/3 signaling and RCC cell aggressiveness. PPM1A could potentially serve as a biomarker for RCC cell aggressiveness.


Asunto(s)
Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Proteína Fosfatasa 2C/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo/genética , Silenciador del Gen/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Análisis de Supervivencia , Factor de Crecimiento Transformador beta1/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Toxicol Appl Pharmacol ; 433: 115773, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34688701

RESUMEN

Carcinogenicity of hexavalent chromium [Cr (VI)] has been supported by a number of epidemiological and animal studies; however, its carcinogenic mode of action is still incompletely understood. To identify mechanisms involved in cancer development, we analyzed gene expression data from duodena of mice exposed to Cr(VI) in drinking water. This analysis included (i) identification of upstream regulatory molecules that are likely responsible for the observed gene expression changes, (ii) identification of annotated gene expression data from public repositories that correlate with gene expression changes in duodena of Cr(VI)-exposed mice, and (iii) identification of hallmark and oncogenic signature gene sets relevant to these data. We identified the inactivated CFTR gene among the top scoring upstream regulators, and found positive correlations between the expression data from duodena of Cr(VI)-exposed mice and other datasets in public repositories associated with the inactivation of the CFTR gene. In addition, we found enrichment of signatures for oncogenic signaling, sustained cell proliferation, impaired apoptosis and tissue remodeling. Results of our computational study support the tumor-suppressor role of the CFTR gene. Furthermore, our results support human relevance of the Cr(VI)-mediated carcinogenesis observed in the small intestines of exposed mice and suggest possible groups that may be more vulnerable to the adverse outcomes associated with the inactivation of CFTR by hexavalent chromium or other agents. Lastly, our findings predict, for the first time, the role of CFTR inactivation in chemical carcinogenesis and expand the range of plausible mechanisms that may be operative in Cr(VI)-mediated carcinogenesis of intestinal and possibly other tissues.


Asunto(s)
Transformación Celular Neoplásica/inducido químicamente , Cromo/toxicidad , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Neoplasias Duodenales/inducido químicamente , Duodeno/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Contaminantes Químicos del Agua/toxicidad , Administración Oral , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cromo/administración & dosificación , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Bases de Datos Genéticas , Agua Potable , Neoplasias Duodenales/genética , Neoplasias Duodenales/metabolismo , Neoplasias Duodenales/patología , Duodeno/metabolismo , Duodeno/patología , Perfilación de la Expresión Génica , Ratones , Medición de Riesgo , Biología de Sistemas , Transcriptoma , Proteínas Supresoras de Tumor/metabolismo , Contaminantes Químicos del Agua/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA