Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circulation ; 149(19): 1501-1515, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38223978

RESUMEN

BACKGROUND: During the neonatal stage, the cardiomyocyte undergoes a constellation of molecular, cytoarchitectural, and functional changes known collectively as cardiomyocyte maturation to increase myocardial contractility and cardiac output. Despite the importance of cardiomyocyte maturation, the molecular mechanisms governing this critical process remain largely unexplored. METHODS: We leveraged an in vivo mosaic knockout system to characterize the role of Carm1, the founding member of protein arginine methyltransferase, in cardiomyocyte maturation. Using a battery of assays, including immunohistochemistry, immuno-electron microscopy imaging, and action potential recording, we assessed the effect of loss of Carm1 function on cardiomyocyte cell growth, myofibril expansion, T-tubule formation, and electrophysiological maturation. Genome-wide transcriptome profiling, H3R17me2a chromatin immunoprecipitation followed by sequencing, and assay for transposase-accessible chromatin with high-throughput sequencing were used to investigate the mechanisms by which CARM1 (coactivator-associated arginine methyltransferase 1) regulates cardiomyocyte maturation. Finally, we interrogated the human syntenic region to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks for single-nucleotide polymorphisms associated with human heart diseases. RESULTS: We report that mosaic ablation of Carm1 disrupts multiple aspects of cardiomyocyte maturation cell autonomously, leading to reduced cardiomyocyte size and sarcomere thickness, severe loss and disorganization of T tubules, and compromised electrophysiological maturation. Genomics study demonstrates that CARM1 directly activates genes that underlie cardiomyocyte cytoarchitectural and electrophysiological maturation. Moreover, our study reveals significant enrichment of human heart disease-associated single-nucleotide polymorphisms in the human genomic region syntenic to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks. CONCLUSIONS: This study establishes a critical and multifaceted role for CARM1 in regulating cardiomyocyte maturation and demonstrates that deregulation of CARM1-dependent cardiomyocyte maturation gene expression may contribute to human heart diseases.


Asunto(s)
Epigénesis Genética , Miocitos Cardíacos , Proteína-Arginina N-Metiltransferasas , Animales , Humanos , Ratones , Diferenciación Celular/genética , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo
2.
Nat Immunol ; 25(2): 268-281, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38195702

RESUMEN

Melanoma cells, deriving from neuroectodermal melanocytes, may exploit the nervous system's immune privilege for growth. Here we show that nerve growth factor (NGF) has both melanoma cell intrinsic and extrinsic immunosuppressive functions. Autocrine NGF engages tropomyosin receptor kinase A (TrkA) on melanoma cells to desensitize interferon γ signaling, leading to T and natural killer cell exclusion. In effector T cells that upregulate surface TrkA expression upon T cell receptor activation, paracrine NGF dampens T cell receptor signaling and effector function. Inhibiting NGF, either through genetic modification or with the tropomyosin receptor kinase inhibitor larotrectinib, renders melanomas susceptible to immune checkpoint blockade therapy and fosters long-term immunity by activating memory T cells with low affinity. These results identify the NGF-TrkA axis as an important suppressor of anti-tumor immunity and suggest larotrectinib might be repurposed for immune sensitization. Moreover, by enlisting low-affinity T cells, anti-NGF reduces acquired resistance to immune checkpoint blockade and prevents melanoma recurrence.


Asunto(s)
Melanoma , Receptor de Factor de Crecimiento Nervioso , Humanos , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Tropomiosina , Melanoma/terapia , Receptor trkA/genética , Receptor trkA/metabolismo , Citoprotección , Inhibidores de Puntos de Control Inmunológico , Células T de Memoria , Terapia de Inmunosupresión , Inmunoterapia , Receptores de Antígenos de Linfocitos T
3.
Adv Sci (Weinh) ; 11(2): e2303489, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37964763

RESUMEN

The essential branched-chain amino acids (BCAAs) leucine, isoleucine, and valine play critical roles in protein synthesis and energy metabolism. Despite their widespread use as nutritional supplements, BCAAs' full effects on mammalian physiology remain uncertain due to the complexities of BCAA metabolic regulation. Here a novel mechanism linking intrinsic alterations in BCAA metabolism is identified to cellular senescence and the senescence-associated secretory phenotype (SASP), both of which contribute to organismal aging and inflammation-related diseases. Altered BCAA metabolism driving the SASP is mediated by robust activation of the BCAA transporters Solute Carrier Family 6 Members 14 and 15 as well as downregulation of the catabolic enzyme BCAA transaminase 1 during onset of cellular senescence, leading to highly elevated intracellular BCAA levels in senescent cells. This, in turn, activates the mammalian target of rapamycin complex 1 (mTORC1) to establish the full SASP program. Transgenic Drosophila models further indicate that orthologous BCAA regulators are involved in the induction of cellular senescence and age-related phenotypes in flies, suggesting evolutionary conservation of this metabolic pathway during aging. Finally, experimentally blocking BCAA accumulation attenuates the inflammatory response in a mouse senescence model, highlighting the therapeutic potential of modulating BCAA metabolism for the treatment of age-related and inflammatory diseases.


Asunto(s)
Aminoácidos de Cadena Ramificada , Fenotipo Secretor Asociado a la Senescencia , Animales , Ratones , Aminoácidos de Cadena Ramificada/metabolismo , Leucina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Metabolismo Energético , Mamíferos/metabolismo
4.
Mol Plant ; 16(11): 1847-1865, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37822080

RESUMEN

Histone H2A monoubiquitination is associated with transcriptional repression and needs to be removed by deubiquitinases to facilitate gene transcription in eukaryotes. However, the deubiquitinase responsible for genome-wide H2A deubiquitination in plants has yet to be identified. In this study, we found that the previously identified PWWP-EPCR-ARID-TRB (PEAT) complex components interact with both the ubiquitin-specific protease UBP5 and the redundant histone acetyltransferases HAM1 and HAM2 (HAM1/2) to form a larger version of PEAT complex in Arabidopsis thaliana. UBP5 functions as an H2A deubiquitinase in a nucleosome substrate-dependent manner in vitro and mediates H2A deubiquitination at the whole-genome level in vivo. HAM1/2 are shared subunits of the PEAT complex and the conserved NuA4 histone acetyltransferase complex, and are responsible for histone H4K5 acetylation. Within the PEAT complex, the PWWP components (PWWP1, PWWP2, and PWWP3) directly interact with UBP5 and are necessary for UBP5-mediated H2A deubiquitination, while the EPCR components (EPCR1 and EPCR2) directly interact with HAM1/2 and are required for HAM1/2-mediated H4K5 acetylation. Collectively, our study not only identifies dual roles of the PEAT complex in H2A deubiquitination and H4K5 acetylation but also illustrates how these processes collaborate at the whole-genome level to regulate the transcription and development in plants.


Asunto(s)
Arabidopsis , Histonas , Histonas/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Receptor de Proteína C Endotelial , Acetilación , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Enzimas Desubicuitinizantes , Suelo
5.
Nature ; 621(7978): 423-430, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37674078

RESUMEN

Translational reprogramming allows organisms to adapt to changing conditions. Upstream start codons (uAUGs), which are prevalently present in mRNAs, have crucial roles in regulating translation by providing alternative translation start sites1-4. However, what determines this selective initiation of translation between conditions remains unclear. Here, by integrating transcriptome-wide translational and structural analyses during pattern-triggered immunity in Arabidopsis, we found that transcripts with immune-induced translation are enriched with upstream open reading frames (uORFs). Without infection, these uORFs are selectively translated owing to hairpins immediately downstream of uAUGs, presumably by slowing and engaging the scanning preinitiation complex. Modelling using deep learning provides unbiased support for these recognizable double-stranded RNA structures downstream of uAUGs (which we term uAUG-ds) being responsible for the selective translation of uAUGs, and allows the prediction and rational design of translating uAUG-ds. We found that uAUG-ds-mediated regulation can be generalized to human cells. Moreover, uAUG-ds-mediated start-codon selection is dynamically regulated. After immune challenge in plants, induced RNA helicases that are homologous to Ded1p in yeast and DDX3X in humans resolve these structures, allowing ribosomes to bypass uAUGs to translate downstream defence proteins. This study shows that mRNA structures dynamically regulate start-codon selection. The prevalence of this RNA structural feature and the conservation of RNA helicases across kingdoms suggest that mRNA structural remodelling is a general feature of translational reprogramming.


Asunto(s)
Codón Iniciador , Conformación de Ácido Nucleico , ARN Bicatenario , ARN Mensajero , Humanos , Arabidopsis/genética , Arabidopsis/inmunología , Codón Iniciador/genética , Reconocimiento de Inmunidad Innata , Sistemas de Lectura Abierta/genética , Biosíntesis de Proteínas/genética , Biosíntesis de Proteínas/inmunología , Ribosomas/metabolismo , ARN Bicatenario/química , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Mensajero/genética , Transcriptoma , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Aprendizaje Profundo
6.
Cell Res ; 33(7): 516-532, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37169907

RESUMEN

Cellular senescence is a stress-induced, stable cell cycle arrest phenotype which generates a pro-inflammatory microenvironment, leading to chronic inflammation and age-associated diseases. Determining the fundamental molecular pathways driving senescence instead of apoptosis could enable the identification of senolytic agents to restore tissue homeostasis. Here, we identify thrombomodulin (THBD) signaling as a key molecular determinant of the senescent cell fate. Although normally restricted to endothelial cells, THBD is rapidly upregulated and maintained throughout all phases of the senescence program in aged mammalian tissues and in senescent cell models. Mechanistically, THBD activates a proteolytic feed-forward signaling pathway by stabilizing a multi-protein complex in early endosomes, thus forming a molecular basis for the irreversibility of the senescence program and ensuring senescent cell viability. Therapeutically, THBD signaling depletion or inhibition using vorapaxar, an FDA-approved drug, effectively ablates senescent cells and restores tissue homeostasis in liver fibrosis models. Collectively, these results uncover proteolytic THBD signaling as a conserved pro-survival pathway essential for senescent cell viability, thus providing a pharmacologically exploitable senolytic target for senescence-associated diseases.


Asunto(s)
Células Endoteliales , Trombomodulina , Animales , Senescencia Celular , Cirrosis Hepática/tratamiento farmacológico , Transducción de Señal , Apoptosis , Mamíferos
7.
Nat Cell Biol ; 24(2): 230-241, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35145222

RESUMEN

Many cancers have an unusual dependence on glutamine. However, most previous studies have focused on the contribution of glutamine to metabolic building blocks and the energy supply. Here, we report that cancer cells with aberrant expression of glutamate decarboxylase 1 (GAD1) rewire glutamine metabolism for the synthesis of γ-aminobutyric acid (GABA)-a prominent neurotransmitter-in non-nervous tissues. An analysis of clinical samples reveals that increased GABA levels predict poor prognosis. Mechanistically, we identify a cancer-intrinsic pathway through which GABA activates the GABAB receptor to inhibit GSK-3ß activity, leading to enhanced ß-catenin signalling. This GABA-mediated ß-catenin activation both stimulates tumour cell proliferation and suppresses CD8+ T cell intratumoural infiltration, such that targeting GAD1 or GABABR in mouse models overcomes resistance to anti-PD-1 immune checkpoint blockade therapy. Our findings uncover a signalling role for tumour-derived GABA beyond its classic function as a neurotransmitter that can be targeted pharmacologically to reverse immunosuppression.


Asunto(s)
Proliferación Celular , Neoplasias/metabolismo , Escape del Tumor , Microambiente Tumoral/inmunología , beta Catenina/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Células A549 , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Receptores de GABA-B/metabolismo , Carga Tumoral , Escape del Tumor/efectos de los fármacos , Vía de Señalización Wnt , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
8.
Front Plant Sci ; 12: 771850, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069626

RESUMEN

Soybean pubescence plays an important role in insect resistance, drought tolerance, and other stresses. Hence, a deep understanding of the molecular mechanism underlying pubescence is a prerequisite to a deeper understanding of insect resistance and drought tolerance. In the present study, quantitative trait loci (QTL) mapping of pubescence traits was performed using a high-density inter-specific linkage map of one recombinant inbred line (RIL) population, designated NJRINP. It was observed that pubescence length (PL) was negatively correlated with pubescence density (PD). A total of 10 and 9 QTLs distributed on six and five chromosomes were identified with phenotypic variance (PV) of 3.0-9.9% and 0.8-15.8% for PL and PD, respectively, out of which, eight and five were novel. Most decreased PL (8 of 10) and increased PD (8 of 9) alleles were from the wild soybean PI 342618B. Based on gene annotation, Protein ANalysis THrough Evolutionary Relationships and literature search, 21 and 12 candidate genes were identified related to PL and PD, respectively. In addition, Glyma.12G187200 from major QTLs qPL-12-1 and qPD-12-2, was identified as Ps (sparse pubescence) before, having an expression level of fivefold greater in NN 86-4 than in PI 342618B, hence it might be the candidate gene that is conferring both PL and PD. Based on gene expression and cluster analysis, three and four genes were considered as the important candidate genes of PL and PD, respectively. Besides, leaves with short and dense (SD) pubescence, which are similar to the wild soybean pubescence morphology, had the highest resistance to common cutworm (CCW) in soybean. In conclusion, the findings in the present study provide a better understanding of genetic basis and candidate genes information of PL and PD and the relationship with resistance to CCW in soybean.

9.
Plant Cell ; 32(7): 2178-2195, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32358072

RESUMEN

Chromatin remodeling and histone modifications are important for development and floral transition in plants. However, it is largely unknown whether and how these two epigenetic regulators coordinately regulate the important biological processes. Here, we identified three types of Imitation Switch (ISWI) chromatin-remodeling complexes in Arabidopsis (Arabidopsis thaliana). We found that AT-RICH INTERACTING DOMAIN5 (ARID5), a subunit of a plant-specific ISWI complex, can regulate development and floral transition. The ARID-PHD dual domain cassette of ARID5 recognizes both the H3K4me3 histone mark and AT-rich DNA. We determined the ternary complex structure of the ARID5 ARID-PHD cassette with an H3K4me3 peptide and an AT-containing DNA. The H3K4me3 peptide is combinatorially recognized by the PHD and ARID domains, while the DNA is specifically recognized by the ARID domain. Both PHD and ARID domains are necessary for the association of ARID5 with chromatin. The results suggest that the dual recognition of AT-rich DNA and H3K4me3 by the ARID5 ARID-PHD cassette may facilitate the association of the ISWI complex with specific chromatin regions to regulate development and floral transition.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/fisiología , Proteínas de Unión al ADN/genética , Flores/fisiología , Histonas/metabolismo , Proteínas de Arabidopsis/metabolismo , Ensamble y Desensamble de Cromatina , Cristalografía por Rayos X , ADN de Plantas/genética , ADN de Plantas/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica de las Plantas , Histonas/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Plantas Modificadas Genéticamente , Dominios Proteicos
10.
J Integr Plant Biol ; 62(11): 1703-1716, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32396248

RESUMEN

Imitation Switch (ISWI) chromatin remodelers are known to function in diverse multi-subunit complexes in yeast and animals. However, the constitution and function of ISWI complexes in Arabidopsis thaliana remain unclear. In this study, we identified forkhead-associated domain 2 (FHA2) as a plant-specific subunit of an ISWI chromatin-remodeling complex in Arabidopsis. By in vivo and in vitro analyses, we demonstrated that FHA2 directly binds to RLT1 and RLT2, two redundant subunits of the ISWI complex in Arabidopsis. The stamen filament is shorter in the fha2 and rlt1/2 mutants than in the wild type, whereas their pistil lengths are comparable. The shorter filament, which is due to reduced cell size, results in insufficient pollination and reduced fertility. The rlt1/2 mutant shows an early-flowering phenotype, whereas the phenotype is not shared by the fha2 mutant. Consistent with the functional specificity of FHA2, our RNA-seq analysis indicated that the fha2 mutant affects a subset of RLT1/2-regulated genes that does not include genes involved in the regulation of flowering time. This study demonstrates that FHA2 functions as a previously uncharacterized subunit of the Arabidopsis ISWI complex and is exclusively involved in regulating stamen development and plant fertility.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Flores/metabolismo , Proteínas Nucleares/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Proteínas Nucleares/genética , Nucleosomas/metabolismo , Infertilidad Vegetal/genética , Infertilidad Vegetal/fisiología
11.
EMBO J ; 39(7): e102008, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32115743

RESUMEN

Deposition of H2A.Z in chromatin is known to be mediated by a conserved SWR1 chromatin-remodeling complex in eukaryotes. However, little is known about whether and how the SWR1 complex cooperates with other chromatin regulators. Using immunoprecipitation followed by mass spectrometry, we found all known components of the Arabidopsis thaliana SWR1 complex and additionally identified the following three classes of previously uncharacterized plant-specific SWR1 components: MBD9, a methyl-CpG-binding domain-containing protein; CHR11 and CHR17 (CHR11/17), ISWI chromatin remodelers responsible for nucleosome sliding; and TRA1a and TRA1b, accessory subunits of the conserved NuA4 histone acetyltransferase complex. MBD9 directly interacts with CHR11/17 and the SWR1 catalytic subunit PIE1, and is responsible for the association of CHR11/17 with the SWR1 complex. MBD9, TRA1a, and TRA1b function as canonical components of the SWR1 complex to mediate H2A.Z deposition. CHR11/17 are not only responsible for nucleosome sliding but also involved in H2A.Z deposition. These results indicate that the association of the SWR1 complex with CHR11/17 may facilitate the coupling of H2A.Z deposition with nucleosome sliding, thereby co-regulating gene expression, development, and flowering time.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Adenosina Trifosfatasas/metabolismo , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Histona Acetiltransferasas/metabolismo , Nucleosomas/metabolismo , Mapas de Interacción de Proteínas , Factores de Transcripción/metabolismo
12.
EMBO J ; 37(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30104406

RESUMEN

In eukaryotes, heterochromatin regions are typically subjected to transcriptional silencing. DNA methylation has an important role in such silencing and has been studied extensively. However, little is known about how methylated heterochromatin regions are subjected to silencing. We conducted a genetic screen and identified an epcr (enhancer of polycomb-related) mutant that releases heterochromatin silencing in Arabidopsis thaliana We demonstrated that EPCR1 functions redundantly with its paralog EPCR2 and interacts with PWWP domain-containing proteins (PWWPs), AT-rich interaction domain-containing proteins (ARIDs), and telomere repeat binding proteins (TRBs), thus forming multiple functionally redundant protein complexes named PEAT (PWWPs-EPCRs-ARIDs-TRBs). The PEAT complexes mediate histone deacetylation and heterochromatin condensation and thereby facilitate heterochromatin silencing. In heterochromatin regions, the production of small interfering RNAs (siRNAs) and DNA methylation is repressed by the PEAT complexes. The study reveals how histone deacetylation, heterochromatin condensation, siRNA production, and DNA methylation interplay with each other and thereby maintain heterochromatin silencing.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Regulación de la Expresión Génica de las Plantas/fisiología , Silenciador del Gen/fisiología , Heterocromatina/metabolismo , Histonas/metabolismo , Complejos Multiproteicos/metabolismo , Acetilación , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Heterocromatina/genética , Histonas/genética , Complejos Multiproteicos/genética
13.
Nat Commun ; 7: 11715, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27273316

RESUMEN

Transposable elements and other repetitive DNA sequences are usually subject to DNA methylation and transcriptional silencing. However, anti-silencing mechanisms that promote transcription in these regions are not well understood. Here, we describe an anti-silencing factor, Bromodomain and ATPase domain-containing protein 1 (BRAT1), which we identified by a genetic screen in Arabidopsis thaliana. BRAT1 interacts with an ATPase domain-containing protein, BRP1 (BRAT1 Partner 1), and both prevent transcriptional silencing at methylated genomic regions. Although BRAT1 mediates DNA demethylation at a small set of loci targeted by the 5-methylcytosine DNA glycosylase ROS1, the involvement of BRAT1 in anti-silencing is largely independent of DNA demethylation. We also demonstrate that the bromodomain of BRAT1 binds to acetylated histone, which may facilitate the prevention of transcriptional silencing. Thus, BRAT1 represents a potential link between histone acetylation and transcriptional anti-silencing at methylated genomic regions, which may be conserved in eukaryotes.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Silenciador del Gen , Histonas/metabolismo , Acetilación , Proteínas de Arabidopsis/química , Desmetilación del ADN , Metilación de ADN , Sitios Genéticos , Modelos Biológicos , Mutación/genética , Unión Proteica , Dominios Proteicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Planta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA