Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
aBIOTECH ; 5(1): 1-16, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38576437

RESUMEN

Di- and tri-methylation of lysine 36 on histone H3 (H3K36me2/3) is catalysed by histone methyltransferase Set2, which plays an essential role in transcriptional regulation. Although there is a single H3K36 methyltransferase in yeast and higher eukaryotes, two H3K36 methyltransferases, Ash1 and Set2, were present in many filamentous fungi. However, their roles in H3K36 methylation and transcriptional regulation remained unclear. Combined with methods of RNA-seq and ChIP-seq, we revealed that both Ash1 and Set2 are redundantly required for the full H3K36me2/3 activity in Magnaporthe oryzae, which causes the devastating worldwide rice blast disease. Ash1 and Set2 distinguish genomic H3K36me2/3-marked regions and are differentially associated with repressed and activated transcription, respectively. Furthermore, Ash1-catalysed H3K36me2 was co-localized with H3K27me3 at the chromatin, and Ash1 was required for the enrichment and transcriptional silencing of H3K27me3-occupied genes. With the different roles of Ash1 and Set2, in H3K36me2/3 enrichment and transcriptional regulation on the stress-responsive genes, they differentially respond to various stresses in M. oryzae. Overall, we reveal a novel mechanism by which two H3K36 methyltransferases catalyze H3K36me2/3 that differentially associate with transcriptional activities and contribute to enrichment of facultative heterochromatin in eukaryotes. Supplementary Information: The online version contains supplementary material available at 10.1007/s42994-023-00127-3.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38422619

RESUMEN

Targeting the transient receptor potential vanilloid 2 channels (TRPV2) in order to alleviate or reverse the course of several diseases including multiple cancers, cardiovascular, immunological, or neurological disorders have been a matter of focus for several years now. SET2, a selective TRPV2 inhibitor, represents an innovative molecule which came into recognition in 2019 and seems to be a promising therapeutic modality in cancer and cardiac diseases. Drug discovery and bioanalysis in clinical environment demands simple, excellent, highly reliable, fast, sensitive, and selective analytical approaches which enable unambiguous identification and quantification of demanded molecule. Here, a targeted ultra-high-performance liquid chromatography - tandem mass spectrometry with electrospray ionization was developed for the quantification of SET2 in plasma samples. The developed method enabled analysis of approx. 15 samples within one hour. Simplicity of the whole analytical procedure can be emphasized by a very simple sample pretreatment based only on the protein precipitation with organic acid (here, 2 M tricholoroacetic acid). The validation procedure was characterized by promising validation parameters and excellent sensitivity what was documented by the limit of detection value at pg.mL-1 concentration level. Analytical validation reported intra- and interday accuracy < 15 % for all quality control samples concentration levels. Similarly, excellent level of intra- (0.1 - 4.8 %) and interday (0.5 - 3.3 %) precision for the tested quality control samples was obtained. The applicability of the developed method was proven by quantifying SET2 concentration levels in plasma samples obtained from Wistar rats that were administered this drug intraperitoneally at a dose of 25 mg/kg. We expect that our new analytical method represents a very attractive tool that could be easily implemented in pharmacokinetics studies and/or therapeutic drug monitoring. Moreover, its applicability was confirmed by the new practicability evaluation metric tool.


Asunto(s)
Descubrimiento de Drogas , Espectrometría de Masas en Tándem , Ratas , Animales , Espectrometría de Masas en Tándem/métodos , Cromatografía Líquida de Alta Presión/métodos , Ratas Wistar , Calibración , Reproducibilidad de los Resultados
3.
Cell Rep ; 42(10): 113186, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37796660

RESUMEN

Loss of transcription-coupled histone H3 lysine 36 trimethylation (H3K36me3) contributes to shorter lifespans in eukaryotes. However, the molecular mechanism of the decline of H3K36me3 during aging remains poorly understood. Here, we report that the degradation of the methyltransferase Set2 is the cause of decreased H3K36me3 levels during chronological aging in budding yeast. We show that Set2 protein degradation during cellular senescence and chronological aging is mainly mediated by the ubiquitin-conjugating E2 enzyme Ubc3 and the E3 ligase Bre1. Lack of Bre1 or abolishment of the ubiquitination stabilizes Set2 protein, sustains H3K36me3 levels at the aging-related gene loci, and upregulates their gene expression, thus leading to extended chronological lifespan. We further illustrate that Gcn5-mediated Set2 acetylation is a prerequisite for Bre1-catalyzed Set2 polyubiquitination and proteolysis during aging. We propose that two sequential post-translational modifications regulate Set2 homeostasis, suggesting a potential strategy to target the Gcn5-Bre1-Set2 axis for intervention of longevity.


Asunto(s)
Envejecimiento , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Histonas/metabolismo , Metilación , Metiltransferasas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Envejecimiento/genética
4.
J Cardiovasc Dev Dis ; 10(7)2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37504562

RESUMEN

Methyltransferases regulate transcriptome dynamics during development and aging, as well as in disease. Various methyltransferases have been linked to heart disease, through disrupted expression and activity, and genetic variants associated with congenital heart disease. However, in vivo functional data for many of the methyltransferases in the context of the heart are limited. Here, we used the Drosophila model system to investigate different histone 3 lysine 36 (H3K36) methyltransferases for their role in heart development. The data show that Drosophila Ash1 is the functional homolog of human ASH1L in the heart. Both Ash1 and Set2 H3K36 methyltransferases are required for heart structure and function during development. Furthermore, Ash1-mediated H3K36 methylation (H3K36me2) is essential for healthy heart function, which depends on both Ash1-complex components, Caf1-55 and MRG15, together. These findings provide in vivo functional data for Ash1 and its complex, and Set2, in the context of H3K36 methylation in the heart, and support a role for their mammalian homologs, ASH1L with RBBP4 and MORF4L1, and SETD2, during heart development and disease.

5.
Genetics ; 223(4)2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36810679

RESUMEN

Post-translational modifications on histones are well known to regulate chromatin structure and function, but much less information is available on modifications of the centromeric histone H3 variant and their effect at the kinetochore. Here, we report two modifications on the centromeric histone H3 variant CENP-A/Cse4 in the yeast Saccharomyces cerevisiae, methylation at arginine 143 (R143me) and lysine 131 (K131me), that affect centromere stability and kinetochore function. Both R143me and K131me lie in the core region of the centromeric nucleosome, near the entry/exit sites of the DNA from the nucleosome. Unexpectedly, mutation of Cse4-R143 (cse4-R143A) exacerbated the kinetochore defect of mutations in components of the NDC80 complex of the outer kinetochore (spc25-1) and the MIND complex (dsn1-7). The analysis of suppressor mutations of the spc25-1 cse4-R143A growth defect highlighted residues in Spc24, Ndc80, and Spc25 that localize to the tetramerization domain of the NDC80 complex and the Spc24-Spc25 stalk, suggesting that the mutations enhance interactions among NDC80 complex components and thus stabilize the complex. Furthermore, the Set2 histone methyltransferase inhibited kinetochore function in spc25-1 cse4-R143A cells, possibly by methylating Cse4-K131. Taken together, our data suggest that Cse4-R143 methylation and Cse4-K131 methylation affect the stability of the centromeric nucleosome, which is detrimental in the context of defective NDC80 tetramerization and can be compensated for by strengthening interactions among NDC80 complex components.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Cinetocoros/metabolismo , Proteína A Centromérica/genética , Proteína A Centromérica/metabolismo , Lisina/genética , Histonas/metabolismo , Metilación , Nucleosomas/genética , Arginina/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Nucleares/genética
6.
Autophagy ; 19(1): 224-240, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35503435

RESUMEN

Dietary restriction (DR) is known to promote autophagy to exert its longevity effect. While SAMS-1 (S-adenosyl methionine synthetase-1) has been shown to be a key mediator of the DR response, little is known about the roles of S-adenosyl methionine (SAM) and SAM-dependent methyltransferase in autophagy and DR-induced longevity. In this study, we show that DR and SAMS-1 repress the activity of SET-2, a histone H3K4 methyltransferase, by limiting the availability of SAM. Consequently, the reduced H3K4me3 levels promote the expression and activity of two transcription factors, HLH-30/TFEB and PHA-4/FOXA, which both regulate the transcription of autophagy-related genes. We then find that HLH-30/TFEB and PHA-4/FOXA act collaboratively on their common target genes to mediate the transcriptional response of autophagy-related genes and consequently the lifespan of the animals. Our study thus shows that the SAMS-1-SET-2 axis serves as a nutrient-sensing module to epigenetically coordinate the activation of HLH-30/TFEB and PHA-4/FOXA transcription factors to control macroautophagy/autophagy and longevity in response to DR.Abbreviations: ChIP: chromatin immunoprecipitation; ChIP-seq: chromatin immuno precipitation-sequencing; COMPASS: complex of proteins associated with Set1; DR: dietary restriction; GO: gene ontology; SAM: S-adenosyl methionine; SAMS-1: S-adenosyl methionine synthetase-1; TSS: transcription start site; WT: wild-type.


Asunto(s)
Proteínas de Caenorhabditis elegans , Longevidad , Animales , Longevidad/fisiología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Histonas/metabolismo , Metilación , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/metabolismo , Autofagia/genética , Factores de Transcripción/metabolismo , Metionina , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
7.
Virulence ; 13(1): 1358-1378, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35943142

RESUMEN

Aspergillus flavus infects various crops with aflatoxins, and leads to aspergillosis opportunistically. Though H3K36 methylation plays an important role in fungal toxin metabolism and virulence, no data about the biological function of H3K36 methylation in A. flavus virulence has been reported. Our study showed that the Set2 histone methyltransferase family, AshA and SetB, involves in morphogenesis and mycotoxin anabolism by regulating related transcriptional factors, and they are important for fungal virulence to crops and animals. Western-blotting and double deletion analysis revealed that AshA mainly regulates H3K36me2, whereas SetB is mainly responsible for H3K36me3 in the nucleus. By construction of domain deletion A. flavus strain and point mutation strains by homologous recombination, the study revealed that SET domain is indispensable in mycotoxin anabolism and virulence of A. flavus, and N455 and V457 in it are the key amino acid residues. ChIP analysis inferred that the methyltransferase family controls fungal reproduction and regulates the production of AFB1 by directly regulating the production of the transcriptional factor genes, including wetA, steA, aflR and amylase, through H3K36 trimethylation in their chromatin fragments, based on which this study proposed that, by H3K36 trimethylation, this methyltransferase family controls AFB1 anabolism through transcriptional level and substrate utilization level. This study illuminates the epigenetic mechanism of the Set2 family in regulating fungal virulence and mycotoxin production, and provides new targets for controlling the virulence of the fungus A. flavus.AUTHOR SUMMARYThe methylation of H3K36 plays an important role in the fungal secondary metabolism and virulence, but no data about the regulatory mechanism of H3K36 methylation in the virulence of A. flavus have been reported. Our study revealed that, in the histone methyltransferase Set2 family, AshA mainly catalyzes H3K36me2, and involves in the methylation of H3K36me1, and SetB mainly catalyzes H3K36me3 and H3K36me1. Through domain deletion and point mutation analysis, this study also revealed that the SET domain was critical for the normal biological function of the Set2 family and that N455 and V457 in the domain were critical for AshA. By ChIP-seq and ChIP-qPCR analysis, H3K36 was found to be trimethylation modified in the promotors and ORF positions of wetA, steA, aflR and the amylase gene (AFLA_084340), and further qRT-PCR results showed that these methylation modifications regulate the expression levels of these genes. According to the results of ChIP-seq analysis, we proposed that, by H3K36 trimethylation, this methyltransferase family controls the metabolism of mycotoxin through transcriptional level and substrate utilization level. All the results from this study showed that Set2 family is essential for fungal secondary metabolism and virulence, which lays a theoretical groundwork in the early prevention and treatment of A. flavus pollution, and also provides an effective strategy to fight against other pathogenic fungi.


Asunto(s)
Aspergillus flavus , Micotoxinas , Amilasas/metabolismo , Animales , Aspergillus flavus/genética , Histona Metiltransferasas/metabolismo , Histonas/genética , Histonas/metabolismo , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Micotoxinas/genética , Micotoxinas/metabolismo , Metabolismo Secundario , Virulencia
8.
Cell Mol Life Sci ; 79(6): 346, 2022 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-35661267

RESUMEN

Histone modifying enzymes play critical roles in many key cellular processes and are appealing proteins for targeting by small molecules in disease. However, while the functions of histone modifying enzymes are often linked to epigenetic regulation of the genome, an emerging theme is that these enzymes often also act by non-catalytic and/or non-epigenetic mechanisms. SETD2 (Set2 in yeast) is best known for associating with the transcription machinery and methylating histone H3 on lysine 36 (H3K36) during transcription. This well-characterized molecular function of SETD2 plays a role in fine-tuning transcription, maintaining chromatin integrity, and mRNA processing. Here we give an overview of the various molecular functions and mechanisms of regulation of H3K36 methylation by Set2/SETD2. These fundamental insights are important to understand SETD2's role in disease, most notably in cancer in which SETD2 is frequently inactivated. SETD2 also methylates non-histone substrates such as α-tubulin which may promote genome stability and contribute to the tumor-suppressor function of SETD2. Thus, to understand its role in disease, it is important to understand and dissect the multiple roles of SETD2 within the cell. In this review we discuss how histone methylation by Set2/SETD2 has led the way in connecting histone modifications in active regions of the genome to chromatin functions and how SETD2 is leading the way to showing that we also have to look beyond histones to truly understand the physiological role of an 'epigenetic' writer enzyme in normal cells and in disease.


Asunto(s)
Epigénesis Genética , Histonas , Cromatina/genética , Cromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Metilación , Procesamiento Proteico-Postraduccional/genética , Saccharomyces cerevisiae/metabolismo
9.
Epigenetics Chromatin ; 15(1): 17, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35581654

RESUMEN

The methylation of histone H3 at lysine 36 (H3K36me) is essential for maintaining genomic stability. Indeed, this methylation mark is essential for proper transcription, recombination, and DNA damage response. Loss- and gain-of-function mutations in H3K36 methyltransferases are closely linked to human developmental disorders and various cancers. Structural analyses suggest that nucleosomal components such as the linker DNA and a hydrophobic patch constituted by histone H2A and H3 are likely determinants of H3K36 methylation in addition to the histone H3 tail, which encompasses H3K36 and the catalytic SET domain. Interaction of H3K36 methyltransferases with the nucleosome collaborates with regulation of their auto-inhibitory changes fine-tunes the precision of H3K36me in mediating dimethylation by NSD2 and NSD3 as well as trimethylation by Set2/SETD2. The identification of specific structural features and various cis-acting factors that bind to different forms of H3K36me, particularly the di-(H3K36me2) and tri-(H3K36me3) methylated forms of H3K36, have highlighted the intricacy of H3K36me functional significance. Here, we consolidate these findings and offer structural insight to the regulation of H3K36me2 to H3K36me3 conversion. We also discuss the mechanisms that underlie the cooperation between H3K36me and other chromatin modifications (in particular, H3K27me3, H3 acetylation, DNA methylation and N6-methyladenosine in RNAs) in the physiological regulation of the epigenomic functions of chromatin.


Asunto(s)
Histonas , Procesamiento Proteico-Postraduccional , Cromatina , Histonas/metabolismo , Humanos , Metilación , Nucleosomas
10.
Int J Biochem Cell Biol ; 144: 106155, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34990836

RESUMEN

Epigenetic dysregulation is an important contributor to carcinogenesis. This is not surprising, as chromatin-genomic DNA organized around structural histone scaffolding-serves as the template on which occurs essential nuclear processes, such as transcription, DNA replication and DNA repair. Histone H3 lysine 36 (H3K36) methyltransferases, such as the SET-domain 2 protein (SETD2), have emerged as critical tumor suppressors. Previous work on mammalian SETD2 and its counterpart in model organisms, Set2, has highlighted the role of this protein in governing genomic stability through transcriptional elongation and splicing, as well as in DNA damage response processes and cell cycle progression. A compendium of SETD2 mutations have been documented, garnered from sequenced cancer patient genome data, and these findings underscore the cancer-driving properties of SETD2 loss-of-function. In this review, we consolidate the molecular mechanisms regulated by SETD2/Set2 and discuss evidence of its dysregulation in tumorigenesis. Insight into the genetic interactions that exist between SETD2 and various canonical intracellular signaling pathways has not only empowered pharmacological intervention by taking advantage of synthetic lethality but underscores SETD2 as a druggable target for precision cancer therapy.


Asunto(s)
Histonas , Neoplasias , Animales , Carcinogénesis/genética , Cromatina , Inestabilidad Genómica , Histonas/genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , Mamíferos/genética , Mamíferos/metabolismo , Metilación , Neoplasias/genética
11.
Development ; 149(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34878097

RESUMEN

Gamete formation from germline stem cells (GSCs) is essential for sexual reproduction. However, the regulation of GSC differentiation is incompletely understood. Set2, which deposits H3K36me3 modifications, is required for GSC differentiation during Drosophila oogenesis. We discovered that the H3K36me3 reader Male-specific lethal 3 (Msl3) and histone acetyltransferase complex Ada2a-containing (ATAC) cooperate with Set2 to regulate GSC differentiation in female Drosophila. Msl3, acting independently of the rest of the male-specific lethal complex, promotes transcription of genes, including a germline-enriched ribosomal protein S19 paralog RpS19b. RpS19b upregulation is required for translation of RNA-binding Fox protein 1 (Rbfox1), a known meiotic cell cycle entry factor. Thus, Msl3 regulates GSC differentiation by modulating translation of a key factor that promotes transition to an oocyte fate.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Oogénesis , Oogonios/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Meiosis , Proteínas Nucleares/genética , Oogonios/citología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Factores de Transcripción/genética
12.
Am J Infect Control ; 49(11): 1423-1426, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34689884

RESUMEN

This case study is part of a series centered on the Centers for Disease Control and Prevention's National Healthcare Safety Network's (NHSN) health care-associated infection (HAI) surveillance definitions. This is the first analytic case study published in AJIC since the CDC/ NHSN updated its HAI risk adjustment models and rebaselined the standardized infection ratios (SIRs) in 2015. This case describes a scenario that Infection Preventionists (IPs) have encountered during their analysis of surgical site infection (SSI) surveillance data. The case study is intended to illustrate how specific models can impact the SIR results by highlighting differences in the criteria for NHSN's older and newer risk models: the original versions and the updated models introduced in 2015. Understanding these differences provides insight into how SSI SIR calculations differ between the older and newer NHSN baseline models. NHSN plans to produce another set of HAI risk adjustment models in the future, using newer HAI incidence and risk factor data. While the timetable for these changes remains to be determined, the statistical methods used to produce future models and SIR calculations will continue the precedents that NHSN has established. An online survey link is provided where participants may confidentially answer questions related to the case study and receive immediate feedback in the form of correct answers, explanations, rationales, and summary of teaching points. Details of the case study, answers, and explanations have been reviewed and approved by NHSN staff. We hope that participants take advantage of this educational offering and thereby gain a greater understanding of the NHSN's HAI data analysis. There are 2 baselines available for SSI standardized infection ration (SIRs) in the National Healthcare Safety Network (NHSN); one based on the 2006-2008 national aggregate data and another based on the 2015 data. Each of the 2 baselines has a different set of inclusion criteria for the SSI data, which impact the calculation of the SIR. In this case study, we focused on the impact of the inclusion of PATOS in the calculation of the 2006-2008 baseline SSI SIR and the exclusion of PATOS from the calculation of the 2015 baseline SSI SIR. In the 2006-2008 baseline SSI SIRs, PATOS events and the procedures to which they are linked are included in the calculation of the SSI SIR whereas in the 2015 baseline SSI SIRs, PATOS events and the procedures to which they are linked are excluded from the calculation of the SSI SIR. Meaning, if we control for all other inclusion criteria other than PATOS data for both baselines, we will notice differences in the number of observed events as well as the number of predicted infections for the 2 baselines. For details of the 2015 baseline and risk adjustment calculation, please review the NHSN Guide to the SIR referenced below. For details of the 2006-2008 baseline4 and risk adjustment, please see the SHEA paper "Improving Risk-Adjusted Measures of Surgical Site Infection for the National Healthcare Safety Network" by author Yi Mu.


Asunto(s)
Infección Hospitalaria , Infección de la Herida Quirúrgica , Infección Hospitalaria/epidemiología , Instituciones de Salud , Humanos , Factores de Riesgo , Infección de la Herida Quirúrgica/epidemiología
13.
FEBS Open Bio ; 11(8): 2225-2235, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34115924

RESUMEN

Methyltransferase Set2-mediated methylation of histone H3 lysine 36 (H3K36), which involves the addition of up to three methyl groups at this site, has been demonstrated to function in many chromatin-coupled events. The methylation of H3K36 is known to recruit different chromatin effector proteins, affecting transcription, mRNA splicing and DNA repair. In this study, we engineered two yeast set2 mutants that lack H3K36 mono/dimethylation (H3K36me1/2) and trimethylation (H3K36me3), respectively, and characterized their roles in the production of antisense transcripts under nutrient-rich conditions. Using our new bioinformatics identification pipeline analysis, we are able to identify a larger number of antisense transcripts in set2∆ cells than has been published previously. We further show that H3K36me1/2 or H3K36me3 redundantly repressed the production of antisense transcripts. Moreover, gene ontology (GO) analysis implies that H3K36me3-mediated antisense transcription might play a role in DNA replication and DNA damage repair, which is independent of regulation of the corresponding sense gene expression. Overall, our results validate a coregulatory mechanism of different H3K36 methylation states, particularly in the repression of antisense transcription.

14.
Mol Cell ; 81(13): 2793-2807.e8, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33979575

RESUMEN

DNA replication initiates at genomic locations known as origins of replication, which, in S. cerevisiae, share a common DNA consensus motif. Despite being virtually nucleosome-free, origins of replication are greatly influenced by the surrounding chromatin state. Here, we show that histone H3 lysine 37 mono-methylation (H3K37me1) is catalyzed by Set1p and Set2p and that it regulates replication origin licensing. H3K37me1 is uniformly distributed throughout most of the genome, but it is scarce at replication origins, where it increases according to the timing of their firing. We find that H3K37me1 hinders Mcm2 interaction with chromatin, maintaining low levels of MCM outside of conventional replication origins. Lack of H3K37me1 results in defective DNA replication from canonical origins while promoting replication events at inefficient and non-canonical sites. Collectively, our results indicate that H3K37me1 ensures correct execution of the DNA replication program by protecting the genome from inappropriate origin licensing and spurious DNA replication.


Asunto(s)
Replicación del ADN , ADN de Hongos/biosíntesis , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Metiltransferasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , ADN de Hongos/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Metilación , Metiltransferasas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
15.
Cell Rep ; 31(10): 107751, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32521276

RESUMEN

Set2 co-transcriptionally methylates lysine 36 of histone H3 (H3K36), producing mono-, di-, and trimethylation (H3K36me1/2/3). These modifications recruit or repel chromatin effector proteins important for transcriptional fidelity, mRNA splicing, and DNA repair. However, it was not known whether the different methylation states of H3K36 have distinct biological functions. Here, we use engineered forms of Set2 that produce different lysine methylation states to identify unique and shared functions for H3K36 modifications. Although H3K36me1/2 and H3K36me3 are functionally redundant in many SET2 deletion phenotypes, we found that H3K36me3 has a unique function related to Bur1 kinase activity and FACT (facilitates chromatin transcription) complex function. Further, during nutrient stress, either H3K36me1/2 or H3K36me3 represses high levels of histone acetylation and cryptic transcription that arises from within genes. Our findings uncover the potential for the regulation of diverse chromatin functions by different H3K36 methylation states.


Asunto(s)
Histonas/metabolismo , Procesamiento Proteico-Postraduccional/genética , Transcripción Genética/genética , Humanos , Metilación
16.
Cell Rep ; 27(13): 3760-3769.e4, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31242410

RESUMEN

In the eukaryotic cell, spliceosomes assemble onto pre-mRNA cotranscriptionally. Spliceosome assembly takes place in the context of the chromatin environment, suggesting that the state of the chromatin may affect splicing. The molecular details and mechanisms through which chromatin affects splicing, however, are still unclear. Here, we show a role for the histone methyltransferase Set2 and its histone modification, H3K36 methylation, in pre-mRNA splicing through high-throughput sequencing. Moreover, the effect of H3K36 methylation on pre-mRNA splicing is mediated through the chromodomain protein Eaf3. We find that Eaf3 is recruited to intron-containing genes and that Eaf3 interacts with the splicing factor Prp45. Eaf3 acts with Prp45 and Prp19 after formation of the precatalytic B complex around the time of splicing activation, thus revealing the step in splicing that is regulated by H3K36 methylation. These studies support a model whereby H3K36 facilitates recruitment of an "adapter protein" to support efficient, constitutive splicing.


Asunto(s)
Acetiltransferasas/metabolismo , Histonas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Empalmosomas/metabolismo , Transcripción Genética , Acetiltransferasas/genética , Histonas/genética , Metilación , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Empalmosomas/genética
17.
FEBS J ; 286(13): 2490-2504, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30941832

RESUMEN

CXXC finger binding protein 1 (CFP-1) is an evolutionarily conserved protein that binds to non-methylated CpG-rich promoters in mammals and Caenorhabditis elegans. This conserved epigenetic regulator is part of the COMPASS complex that contains the H3K4me3 methyltransferase SET1 in mammals and SET-2 in C. elegans. Previous studies have indicated the importance of CFP1 in embryonic stem cell differentiation and cell fate specification. However, neither the function nor the mechanism of action of CFP1 is well understood at the organismal level. Here, we have used cfp-1(tm6369) and set-2(bn129) C. elegans mutants to investigate the function of CFP-1 in gene induction and development. We have characterised C. elegansCOMPASS mutants cfp-1(tm6369) and set-2(bn129) and found that both cfp-1 and set-2 play an important role in the regulation of fertility and development of the organism. Furthermore, we found that both cfp-1 and set-2 are required for H3K4 trimethylation and play a repressive role in the expression of heat shock and salt-inducible genes. Interestingly, we found that cfp-1 but not set-2 genetically interacts with histone deacetylase (HDAC1/2) complexes to regulate fertility, suggesting a function of CFP-1 outside of the COMPASS complex. Additionally, we found that cfp-1 and set-2 independently regulate fertility and development of the organism. Our results suggest that CFP-1 genetically interacts with HDAC1/2 complexes to regulate fertility, independent of its function within the COMPASS complex. We propose that CFP-1 could cooperate with the COMPASS complex and/or HDAC1/2 in a context-dependent manner.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Epistasis Genética , Fertilidad , Respuesta al Choque Térmico , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Mutación
18.
Zhonghua Xue Ye Xue Za Zhi ; 40(1): 29-34, 2019 Jan 14.
Artículo en Chino | MEDLINE | ID: mdl-30704225

RESUMEN

Objective: To observe the effect of meisoindigo on apoptosis and proliferation of JAK2/V617F heterozygous mutation cell line-SET2 cell line to further explore the role of JAK-STAT pathway in this effect. Methods: Cell apoptosis after treated with different concentration of meisoindigo (0, 5, and 10 µmol/L) was evaluated by flow cytometry at different time points (24, 48, 72 h). Cell proliferation with CCK8 test was evaluated at different time points (24, 48, 72, 96 h) after administered with different concentration of meisoindigo (0, 5, 10, and 20 µmol/L). After treatment with different concentration of meisoindigo (0, 5, 10, and 20 µmol/L), SET2 cells were collected after 12 h, and then cultured in incomplete methylcellulose-based medium for clone formation. JAK-STAT signaling pathway and apoptosis related protein by Western blot test were evaluated 12 h after administered with different concentration of meisoindigo (0, 5, 10, and 20 µmol/L). Results: At different time points after treated with meisoindigo, the apoptosis rate of SET2 cell lines increased (P<0.01) with the inhibited proliferation (P<0.01), and the decreased clone formation rate of SET2 cell lines [0 µmol/L meisoindigo: (4.48±1.19)%, 20 µmol/L meisoindigo: (2.55±0.36)%; Dunnett P=0.020] in the presence of augmented concentrations of meisoindigo. At 12 hours after administration with meisoindigo, the reduced expressions of JAK2, P-JAK2, P-STAT1, P-STAT3, P-STAT3, STAT5, the decreased anti-apoptosis proteins BCL-2, BCL-XL and the increased pro-apoptosis protein BID, BIM were observed in the presence of increased concentrations of meisoindigo. Conclusion: Meisoindigo played an important role during the apoptosis and the inhibition of proliferation in ph-negative myeloproliferative neoplasm cell-SET2 cell lines, which might be related to the inhibition of JAK-STAT signaling pathway with up-regulation of pro-apoptosis protein and down-regulation of anti-apoptosis protein.


Asunto(s)
Apoptosis , Línea Celular , Proliferación Celular , Indoles , Factor de Transcripción STAT3
19.
J Cell Sci ; 132(6)2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30674555

RESUMEN

Replication stress is a common feature of cancer cells, and thus a potentially important therapeutic target. Here, we show that cyclin-dependent kinase (CDK)-induced replication stress, resulting from Wee1 inactivation, is synthetic lethal with mutations disrupting dNTP homeostasis in fission yeast. Wee1 inactivation leads to increased dNTP demand and replication stress through CDK-induced firing of dormant replication origins. Subsequent dNTP depletion leads to inefficient DNA replication, DNA damage and to genome instability. Cells respond to this replication stress by increasing dNTP supply through histone methyltransferase Set2-dependent MBF-induced expression of Cdc22, the catalytic subunit of ribonucleotide reductase (RNR). Disrupting dNTP synthesis following Wee1 inactivation, through abrogating Set2-dependent H3K36 tri-methylation or DNA integrity checkpoint inactivation results in critically low dNTP levels, replication collapse and cell death, which can be rescued by increasing dNTP levels. These findings support a 'dNTP supply and demand' model in which maintaining dNTP homeostasis is essential to prevent replication catastrophe in response to CDK-induced replication stress.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Nucleótidos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Puntos de Control del Ciclo Celular , Daño del ADN , Replicación del ADN , Código de Histonas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Homeostasis , Metilación , Schizosaccharomyces/metabolismo , Mutaciones Letales Sintéticas , Factores de Transcripción/metabolismo
20.
Chinese Journal of Hematology ; (12): 29-34, 2019.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-810391

RESUMEN

Objective@#To observe the effect of meisoindigo on apoptosis and proliferation of JAK2/V617F heterozygous mutation cell line-SET2 cell line to further explore the role of JAK-STAT pathway in this effect.@*Methods@#Cell apoptosis after treated with different concentration of meisoindigo (0, 5, and 10 μmol/L) was evaluated by flow cytometry at different time points (24, 48, 72 h). Cell proliferation with CCK8 test was evaluated at different time points (24, 48, 72, 96 h) after administered with different concentration of meisoindigo (0, 5, 10, and 20 μmol/L). After treatment with different concentration of meisoindigo (0, 5, 10, and 20 μmol/L), SET2 cells were collected after 12 h, and then cultured in incomplete methylcellulose-based medium for clone formation. JAK-STAT signaling pathway and apoptosis related protein by Western blot test were evaluated 12 h after administered with different concentration of meisoindigo (0, 5, 10, and 20 μmol/L).@*Results@#At different time points after treated with meisoindigo, the apoptosis rate of SET2 cell lines increased (P<0.01) with the inhibited proliferation (P<0.01), and the decreased clone formation rate of SET2 cell lines [0 μmol/L meisoindigo: (4.48±1.19)%, 20 μmol/L meisoindigo: (2.55±0.36)%; Dunnett P=0.020] in the presence of augmented concentrations of meisoindigo. At 12 hours after administration with meisoindigo, the reduced expressions of JAK2, P-JAK2, P-STAT1, P-STAT3, P-STAT3, STAT5, the decreased anti-apoptosis proteins BCL-2, BCL-XL and the increased pro-apoptosis protein BID, BIM were observed in the presence of increased concentrations of meisoindigo.@*Conclusion@#Meisoindigo played an important role during the apoptosis and the inhibition of proliferation in ph-negative myeloproliferative neoplasm cell-SET2 cell lines, which might be related to the inhibition of JAK-STAT signaling pathway with up-regulation of pro-apoptosis protein and down-regulation of anti-apoptosis protein.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA