Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Adv Cancer Res ; 161: 223-320, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39032951

RESUMEN

Prostate cancer is one of the most common malignancies among men worldwide. Besides genetic alterations, epigenetic modulations including DNA methylation, histone modifications and miRNA mediated alteration of gene expression are the key driving forces for the prostate tumor development and cancer progression. Aberrant expression and/or the activity of the epigenetic modifiers/enzymes, results in aberrant expression of genes involved in DNA repair, cell cycle regulation, cell adhesion, apoptosis, autophagy, tumor suppression and hormone response and thereby disease progression. Altered epigenome is associated with prostate cancer recurrence, progression, aggressiveness and transition from androgen-dependent to androgen-independent phenotype. These epigenetic modifications are reversible and various compounds/drugs targeting the epigenetic enzymes have been developed that are effective in cancer treatment. This chapter focuses on the epigenetic alterations in prostate cancer initiation and progression, listing different epigenetic biomarkers for diagnosis and prognosis of the disease and their potential as therapeutic targets. This chapter also summarizes different epigenetic drugs approved for prostate cancer therapy and the drugs available for clinical trials.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Metilación de ADN/genética , Andrógenos/metabolismo , Animales
2.
Int J Mol Sci ; 25(13)2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-39000010

RESUMEN

Histone lysine demethylases (KDMs) play an essential role in biological processes such as transcription regulation, RNA maturation, transposable element control, and genome damage sensing and repair. In most cases, their action requires catalytic activities, but non-catalytic functions have also been shown in some KDMs. Indeed, some strictly KDM-related proteins and some KDM isoforms do not act as histone demethylase but show other enzymatic activities or relevant non-enzymatic functions in different cell types. Moreover, many studies have reported on functions potentially supported by catalytically dead mutant KDMs. This is probably due to the versatility of the catalytical core, which can adapt to assume different molecular functions, and to the complex multi-domain structure of these proteins which encompasses functional modules for targeting histone modifications, promoting protein-protein interactions, or recognizing nucleic acid structural motifs. This rich modularity and the availability of multiple isoforms in the various classes produced variants with enzymatic functions aside from histone demethylation or variants with non-catalytical functions during the evolution. In this review we will catalog the proteins with null or questionable demethylase activity and predicted or validated inactive isoforms, summarizing what is known about their alternative functions. We will then go through some experimental evidence for the non-catalytical functions of active KDMs.


Asunto(s)
Histona Demetilasas , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Histona Demetilasas/química , Humanos , Animales , Histonas/metabolismo
3.
Antioxidants (Basel) ; 13(4)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38671918

RESUMEN

Imbalanced osteogenic cell-mediated bone gain and osteoclastic remodeling accelerates the development of osteoporosis, which is the leading risk factor of disability in the elderly. Harmonizing the metabolic actions of bone-making cells and bone resorbing cells to the mineralized matrix network is required to maintain bone mass homeostasis. The tricarboxylic acid (TCA) cycle in mitochondria is a crucial process for cellular energy production and redox homeostasis. The canonical actions of TCA cycle enzymes and intermediates are indispensable in oxidative phosphorylation and adenosine triphosphate (ATP) biosynthesis for osteogenic differentiation and osteoclast formation. Knockout mouse models identify these enzymes' roles in bone mass and microarchitecture. In the noncanonical processes, the metabolites as a co-factor or a substrate involve epigenetic modification, including histone acetyltransferases, DNA demethylases, RNA m6A demethylases, and histone demethylases, which affect genomic stability or chromatin accessibility for cell metabolism and bone formation and resorption. The genetic manipulation of these epigenetic regulators or TCA cycle intermediate supplementation compromises age, estrogen deficiency, or inflammation-induced bone mass loss and microstructure deterioration. This review sheds light on the metabolic functions of the TCA cycle in terms of bone integrity and highlights the crosstalk of the TCA cycle and redox and epigenetic pathways in skeletal tissue metabolism and the intermediates as treatment options for delaying osteoporosis.

4.
Curr Opin Chem Biol ; 79: 102428, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38330792

RESUMEN

The hypoxia-inducible factors are α,ß-heterodimeric transcription factors that mediate the chronic response to hypoxia in humans and other animals. Protein hydroxylases belonging to two different structural subfamilies of the Fe(II) and 2-oxoglutarate (2OG)-dependent oxygenase superfamily modify HIFα. HIFα prolyl-hydroxylation, as catalysed by the PHDs, regulates HIFα levels and, consequently, α,ß-HIF levels. HIFα asparaginyl-hydroxylation, as catalysed by factor inhibiting HIF (FIH), regulates the transcriptional activity of α,ß-HIF. The activities of the PHDs and FIH are regulated by O2 availability, enabling them to act as hypoxia sensors. We provide an overview of the biochemistry of the HIF hydroxylases, discussing evidence that their kinetic and structural properties may be tuned to their roles in the HIF system. Avenues for future research and therapeutic modulation are discussed.


Asunto(s)
Oxigenasas de Función Mixta , Factores de Transcripción , Animales , Humanos , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/metabolismo , Factores de Transcripción/metabolismo , Hipoxia/metabolismo , Hidroxilación
5.
Cancer Metastasis Rev ; 43(2): 795-821, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38227150

RESUMEN

Modulation of histone methylation status is regarded as an important mechanism of epigenetic regulation and has substantial clinical potential for the therapy of diseases, including cancer and other disorders. The present study aimed to provide a comprehensive introduction to the enzymology of histone demethylases, as well as their cancerous roles, molecular mechanisms, therapeutic possibilities, and challenges for targeting them, in order to advance drug design for clinical therapy and highlight new insight into the mechanisms of these enzymes in cancer. A series of clinical trials have been performed to explore potential roles of histone demethylases in several cancer types. Numerous targeted inhibitors associated with immunotherapy, chemotherapy, radiotherapy, and targeted therapy have been used to exert anticancer functions. Future studies should evaluate the dynamic transformation of histone demethylases leading to carcinogenesis and explore individual therapy.


Asunto(s)
Histona Demetilasas , Neoplasias , Humanos , Neoplasias/genética , Neoplasias/enzimología , Neoplasias/patología , Neoplasias/tratamiento farmacológico , Histona Demetilasas/metabolismo , Histona Demetilasas/antagonistas & inhibidores , Animales , Epigénesis Genética , Histonas/metabolismo , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología
6.
Eur J Med Chem ; 264: 115999, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38043489

RESUMEN

Histone demethylases play a critical role in gene transcription regulation and have been implicated in cancer. Numerous reports have highlighted the overexpression of histone demethylases, such as LSD1 and JmjC, in various malignant tumor tissues, identifying them as effective therapeutic targets for cancer treatment. Despite many histone demethylase inhibitors entering clinical trials, their clinical efficacy has been limited. Therefore, combination therapies based on histone demethylase inhibitors, along with other modulators like dual-acting inhibitors, have gained significant attention and made notable progress in recent years. In this review, we provide an overview of recent advances in drug discovery targeting histone demethylases, focusing specifically on drug combination therapy and histone demethylases-targeting dual inhibitors. We discuss the rational design, pharmacodynamics, pharmacokinetics, and clinical status of these approaches. Additionally, we summarize the co-crystal structures of LSD1 inhibitors and their target proteins as well as describe the corresponding binding interactions. Finally, we also provided the challenges and future directions for utilizing histone demethylases in cancer therapy, such as PROTACs and molecular glue etc.


Asunto(s)
Histona Demetilasas , Neoplasias , Humanos , Histona Demetilasas/metabolismo , Neoplasias/tratamiento farmacológico , Descubrimiento de Drogas
7.
Curr Opin Struct Biol ; 83: 102707, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37832177

RESUMEN

Histone methylation, one of the most common histone modifications, has fundamental roles in regulating chromatin-based processes. Jumonji histone lysine demethylases (JMJC KDMs) influence regulation of gene transcription through both their demethylation and chromatin scaffolding functions. It has recently been demonstrated that dysregulation of JMJC KDMs contributes to pathogenesis and progression of several diseases, including cancer. These observations have led to an increased interest in modulation of enzymes that regulate lysine methylation. Here, we highlight recent progress in understanding catalysis of JMJC KDMs. Specifically, we focus on recent research advances on elucidation of JMJC KDM substrate recognition and interactomes. We also highlight recently reported JMJC KDM inhibitors and describe their therapeutic potentials and challenges. Finally, we discuss alternative strategies to target these enzymes, which rely on targeting JMJC KDMs accessory domains as well as utilization of the targeted protein degradation strategy.


Asunto(s)
Histona Demetilasas , Histonas , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Catálisis , Cromatina
8.
Biochim Biophys Acta Gene Regul Mech ; 1866(4): 194986, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37722486

RESUMEN

Differential expression of genes involved in certain processes is a collaborative outcome of crosstalk between signalling molecules and epigenetic modifiers. In response to environmental stimulus, interplay between transcription factors and epigenetic modifiers together dictates the regulation of genes. MLLs and KDM5A are functionally antagonistic proteins, as one acts as a writer and the other erases the active chromatin mark, i.e., H3K4me3. KDM5A influences the process of EMT by binding to both epithelial and mesenchymal gene promoters. Through this work, we show that when bound to E-cadherin promoter, KDM5A acts as a classical repressor by demethylating H3K4me3, but on mesenchymal markers, it acts as a transcriptional activator by inhibiting the activity of HDACs and increasing H3K18ac. Further, through our chromatin immunoprecipitation experiments, we observed a co-occupancy of KDM5A with MLLs, we tested whether KDM5A might physically interact with MLLs and WDR5, and here we provide experimental evidence that KDM5A indeed interacts with MLLs and WDR5.


Asunto(s)
Transición Epitelial-Mesenquimal , Regulación de la Expresión Génica , Transición Epitelial-Mesenquimal/genética , Cromatina , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Adv Sci (Weinh) ; 10(28): e2301367, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37565374

RESUMEN

Lysine demethylase KDM7A removes histone modifications H3K9me1/2 and H3K27me1/2. KDM7A plays critical roles in gene expression and contribute to biological processes including tumorigenesis, metabolism, and embryonic development. However, the functions of KDM7A in mammalian nervous system are still poorly explored. In this study, functional roles of KDM7A are comprehensively investigated in neuronal cells by applying CUT&Tag-seq, RNA-seq and mice models. Knockdown of Kdm7a in N2A cells result in the alteration of histone modifications near transcription start sites (TSSs) and the expression changes of a large number of genes. In particular, the expression of immediate early genes (IEGs), a series of genes maintaining the function of the nervous system and associating with neurological disorders, are significantly decreased upon Kdm7a knockdown. Furthermore, in vivo knockdown of Kdm7a in dentate gyrus (DG) neuron of mice hippocampus, via Adeno-associated virus (AAV)-based stereotaxic microinjection, led to a significant decrease of the expression of c-Fos, a marker of neuron activity. Behavior assays in mice further revealed that Kdm7a knockdown in hippocampus repress neuron activity, which leading to impairment of emotion and memory. Collectively, the study reveals that KDM7A affects neuron functions by regulating IEGs, which may provide new clues for understanding epigenetic mechanisms in neurological disorders.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji , Enfermedades del Sistema Nervioso , Ratones , Animales , Histona Demetilasas con Dominio de Jumonji/genética , Lisina/genética , Genes Inmediatos-Precoces/genética , Neuronas/metabolismo , Mamíferos/metabolismo
10.
Clin. transl. oncol. (Print) ; 25(6): 1594-1605, jun. 2023. ilus
Artículo en Inglés | IBECS | ID: ibc-221192

RESUMEN

Hepatocellular carcinoma (HCC) is the prevalent form of liver cancer in adults and the fourth most common cause of cancer-related death worldwide. HCC predominantly arises in the context of cirrhosis as a result of chronic liver disease, injury and inflammation. Full-blown HCC has poor prognosis because it is highly aggressive and resistant to therapy. Consequently, interventions that can prevent or restrain HCC emergence from pre-cancerous diseased liver are a desirable strategy. Histone methylation is a dynamic, reversible epigenetic modification involving the addition or removal of methyl groups from lysine, arginine or glutamine residues. Aberrant activity of histone methylation writers, erases and readers has been implicated in several cancer types, including HCC. In this review, we provide an overview of research on the role of histone methylation in pre-cancerous and cancerous HCC published over the last 5 years. In particular, we present the evidence linking environmental factors such as diet, viral infections and carcinogenic agents with dysregulation of histone methylation during liver cancer progression with the aim to highlight future therapeutic possibilities (AU)


Asunto(s)
Humanos , Adulto , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Histonas/metabolismo , Lesiones Precancerosas , Metilación
11.
Chemistry ; 29(51): e202301305, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37258457

RESUMEN

KDM6A (UTX) and KDM6B (JMJD3) are human non-heme Fe(II) and 2-oxoglutarate (2OG) dependent JmjC oxygenases that catalyze the demethylation of trimethylated lysine 27 in the N-terminal tail of histone H3, a post-translational modification that regulates transcription. A Combined Quantum Mechanics/ Molecular Mechanics (QM/MM) and Molecular Dynamics (MD) study on the catalytic mechanism of KDM6A/B reveals that the transition state for the rate-limiting hydrogen atom transfer (HAT) reaction in KDM6A catalysis is stabilized by polar (Asn217) and aromatic (Trp369)/non-polar (Pro274) residues in contrast to KDM4, KDM6B and KDM7 demethylases where charged residues (Glu, Arg, Asp) are involved. KDM6A employs both σ- and π-electron transfer pathways for HAT, whereas KDM6B employs the σ-electron pathway. Differences in hydrogen bonding of the Fe-chelating Glu252(KDM6B) contribute to the lower energy barriers in KDM6B vs. KDM6A. The study reveals a dependence of the activation barrier of the rebound hydroxylation on the Fe-O-C angle in the transition state of KDM6A. Anti-correlation of the Zn-binding domain with the active site residues is a key factor distinguishing KDM6A/B from KDM7/4s. The results reveal the importance of communication between the Fe center, second coordination sphere, and long-range interactions in catalysis by KDMs and, by implication, other 2OG oxygenases.


Asunto(s)
Histona Demetilasas , Histonas , Humanos , Histona Demetilasas/metabolismo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Oxigenasas/metabolismo , Catálisis , Compuestos Ferrosos/metabolismo
12.
Epigenomes ; 7(2)2023 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-37218871

RESUMEN

Epigenetic modifications are heritable, reversible changes in histones or the DNA that control gene functions, being exogenous to the genomic sequence itself. Human diseases, particularly cancer, are frequently connected to epigenetic dysregulations. One of them is histone methylation, which is a dynamically reversible and synchronously regulated process that orchestrates the three-dimensional epigenome, nuclear processes of transcription, DNA repair, cell cycle, and epigenetic functions, by adding or removing methylation groups to histones. Over the past few years, reversible histone methylation has become recognized as a crucial regulatory mechanism for the epigenome. With the development of numerous medications that target epigenetic regulators, epigenome-targeted therapy has been used in the treatment of malignancies and has shown meaningful therapeutic potential in preclinical and clinical trials. The present review focuses on the recent advances in our knowledge on the role of histone demethylases in tumor development and modulation, in emphasizing molecular mechanisms that control cancer cell progression. Finally, we emphasize current developments in the advent of new molecular inhibitors that target histone demethylases to regulate cancer progression.

13.
Biochim Biophys Acta Rev Cancer ; 1878(3): 188865, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36841366

RESUMEN

Histone methylation, one of the most prominent epigenetic modifications, plays a vital role in gene transcription, and aberrant histone methylation levels cause tumorigenesis. Histone methylation is a reversible enzyme-dependent reaction, and histone methyltransferases and demethylases are involved in this reaction. This review addresses the biological and clinical relevance of these histone methylation-modifying enzymes for skin cancer. In particular, the roles of histone lysine methyltransferases, histone arginine methyltransferase, lysine-specific demethylases, and JmjC demethylases in skin cancer are discussed in detail. In addition, we summarize the efficacy of several epigenetic inhibitors targeting histone methylation-modifying enzymes in cutaneous cancers, such as basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and melanoma. In conclusion, we propose histone methylation-modifying enzymes as novel targets for next-generation pharmaceuticals in the treatment of skin cancers and further provide a rationale for the development of epigenetic drugs (epidrugs) that target specific histone methylases/demethylases in cutaneous tumors.


Asunto(s)
Histonas , Neoplasias Cutáneas , Humanos , Histonas/genética , Metilación , Lisina/metabolismo , Histona Demetilasas/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , N-Metiltransferasa de Histona-Lisina/genética , Histona Metiltransferasas
14.
Front Pharmacol ; 14: 1120911, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36817147

RESUMEN

Histone lysine-specific demethylase 1 (LSD1/KDM1A) was first identified in 2004 as an epigenetic enzyme able to demethylate specific lysine residues of histone H3, namely H3K4me1/2 and H3K9me1/2, using FAD as the cofactor. It is ubiquitously overexpressed in many types of cancers (breast, gastric, prostate, hepatocellular, and esophageal cancer, acute myeloid leukemia, and others) leading to block of differentiation and increase of proliferation, migration and invasiveness at cellular level. LSD1 inhibitors can be grouped in covalent and non-covalent agents. Each group includes some hybrid compounds, able to inhibit LSD1 in addition to other target(s) at the same time (dual or multitargeting compounds). To date, 9 LSD1 inhibitors have entered clinical trials, for hematological and/or solid cancers. Seven of them (tranylcypromine, iadademstat (ORY-1001), bomedemstat (IMG-7289), GSK-2879552, INCB059872, JBI-802, and Phenelzine) covalently bind the FAD cofactor, and two are non-covalent LSD1 inhibitors [pulrodemstat (CC-90011) and seclidemstat (SP-2577)]. Another TCP-based LSD1/MAO-B dual inhibitor, vafidemstat (ORY-2001), is in clinical trial for Alzheimer's diseases and personality disorders. The present review summarizes the structure and functions of LSD1, its pathological implications in cancer and non-cancer diseases, and the identification of LSD1 covalent and non-covalent inhibitors with different chemical scaffolds, including those involved in clinical trials, highlighting their potential as potent and selective anticancer agents.

15.
Clin Transl Oncol ; 25(6): 1594-1605, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36650321

RESUMEN

Hepatocellular carcinoma (HCC) is the prevalent form of liver cancer in adults and the fourth most common cause of cancer-related death worldwide. HCC predominantly arises in the context of cirrhosis as a result of chronic liver disease, injury and inflammation. Full-blown HCC has poor prognosis because it is highly aggressive and resistant to therapy. Consequently, interventions that can prevent or restrain HCC emergence from pre-cancerous diseased liver are a desirable strategy. Histone methylation is a dynamic, reversible epigenetic modification involving the addition or removal of methyl groups from lysine, arginine or glutamine residues. Aberrant activity of histone methylation writers, erases and readers has been implicated in several cancer types, including HCC. In this review, we provide an overview of research on the role of histone methylation in pre-cancerous and cancerous HCC published over the last 5 years. In particular, we present the evidence linking environmental factors such as diet, viral infections and carcinogenic agents with dysregulation of histone methylation during liver cancer progression with the aim to highlight future therapeutic possibilities.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Adulto , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Metilación , Histonas/metabolismo
16.
Comb Chem High Throughput Screen ; 26(1): 224-235, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35585823

RESUMEN

BACKGROUND: cGAS-STING signaling has been primarily discovered as an important DNA sensing machinery, bridging innate immunity and adaptive immunity. Beyond its antiviral response, recent evidence expanded its complicated role in cancer therapy. METHODS: UALCAN, The TCGA Wander, GEPIA, SMART, TIMER, Kaplan-Meier plotter, TCGA Data, and cBioPortal were utilized in the investigation. RESULTS: We evaluated the expression of four key molecules (MB21D1, TMEM173, TBK1, and IRF3) in the cGAS-STING pathway and found that the TMEM173 gene was significantly downregulated in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). Not only immunostimulatory cells but also regulatory T cells were triggered by the DNA sensing pathway. With gene enrichment analysis, we revealed that cell cycle and mechanotransduction/cytoskeleton signals were most closely connected with cGAS-STING signal alterations in non-small-cell lung cancer (NSCLC). cGAS-STING signaling was robustly correlated with methylation changes, especially histone H3K4 lysine demethylase KDM5s. Transient activation of cGAS-STING was found to exert tumor surveillance effect, and inhibition of STING signaling co-opt elevated KDM5 demethylases might inadvertently worsen clinical outcomes. CONCLUSION: cGAS-STING signaling and KDM5 demethylases have the potential to be used as targets for evaluating an effective immune response in the tumor microenvironment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Metilación , Mecanotransducción Celular , Neoplasias Pulmonares/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , ADN , Microambiente Tumoral
17.
J Cell Biochem ; 123(11): 1780-1792, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35933705

RESUMEN

Glycyrrhiza inflata Bat. is a type of abiotic-stress-resistant plant with extremely high medicinal value. Histone demethylases (HDMTs) have been known to play crucial roles in the regulation of abiotic stress response. However, the molecular functions of HDMTs has not been studied in G. inflata. Here we identified 34 GiHDMT genes in G. inflata, which could be divided into the 6 groups through phylogenetic analysis. We further found that the gene structure and conserved protein motifs exhibit high conservation in each group of GiHDMT genes. Various abiotic-stress-related elements are detected in GiHDMT promoters, especially for the light-responsive element and abscisic acid-responsive element. Collinearity analysis indicated that segmental duplication contributed to the expansion of the GiHDMT family in licorice. Subcellular localization analysis revealed that green fluorescent protein-tagged GiHDMT2 and GiHDMT18 were predominantly localized in the nucleus, whereas GiHDMT1 were found in both the cytoplasm and nucleus. Real-time quantitative polymerase chain reaction showed that GiHDMTs presented differential expression patterns across different tissues. Moreover, changes in transcription level of GiHDMTs under abiotic stress indicate the potential role of GiHDMTs in the stress response in licorice. Finally, we found the histone methylation levels probably mediated by GiHDMT genes are changed with the treatment of NaCl and Na2 CO3 . Our study will lay the foundation for future research on the regulatory roles of GiHDMT genes in the environmental stress.


Asunto(s)
Glycyrrhiza , Glycyrrhiza/química , Glycyrrhiza/metabolismo , Regulación de la Expresión Génica de las Plantas , Filogenia , Proteínas de Plantas/genética , Histona Demetilasas/genética , Estrés Fisiológico/genética
18.
Int J Mol Sci ; 23(14)2022 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-35887017

RESUMEN

The Jumonji-C (JmjC) family of lysine demethylases (KDMs) (JMJC-KDMs) plays an essential role in controlling gene expression and chromatin structure. In most cases, their function has been attributed to the demethylase activity. However, accumulating evidence demonstrates that these proteins play roles distinct from histone demethylation. This raises the possibility that they might share domains that contribute to their functional outcome. Here, we show that the JMJC-KDMs contain low-complexity domains and intrinsically disordered regions (IDR), which in some cases reached 70% of the protein. Our data revealed that plant homeodomain finger protein (PHF2), KDM2A, and KDM4B cluster by phase separation. Moreover, our molecular analysis implies that PHF2 IDR contributes to transcription regulation. These data suggest that clustering via phase separation is a common feature that JMJC-KDMs utilize to facilitate their functional responses. Our study uncovers a novel potential function for the JMJC-KDM family that sheds light on the mechanisms to achieve the competent concentration of molecules in time and space within the cell nucleus.


Asunto(s)
Histona Demetilasas , Histona Demetilasas con Dominio de Jumonji , Núcleo Celular/metabolismo , Desmetilación , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Lisina/metabolismo
19.
Clin Genet ; 102(3): 169-181, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35713103

RESUMEN

Epigenetic modifications of DNA and histone tails are essential for gene expression regulation. They play an essential role in neurodevelopment as nervous system development is a complex process requiring a dynamic pattern of gene expression. Histone methylation is one of the vital epigenetic regulators and mostly occurs on lysine residues of histones H3 and H4. Histone methylation is catalyzed by two sets of enzymes: histone lysine methyltransferases (KMTs) and histone lysine demethylases (KDMs). KMT2 enzymes form a distinct multi-subunit complex known as COMPASS to enhance their catalytic activity and diversify their biologic functions. Several neurodevelopmental syndromes result from defects in histone methylation which can be caused by deficiencies in histone methyltransferases and demethylases, loss of the histone methyltransferase activator TASP1, or derangements in COMPASS formation. In this review article, the molecular mechanism of histone methylation is discussed followed by summarizing clinical syndromes caused by monogenic defects in histone methylation.


Asunto(s)
Histonas , Lisina , Histona Demetilasas/genética , Histona Metiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/metabolismo , Humanos , Lisina/química , Lisina/genética , Lisina/metabolismo , Metilación , Síndrome
20.
Methods Mol Biol ; 2529: 63-88, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733010

RESUMEN

Dynamic histone methylation regulates gene activation and repression. It is involved in proliferation, differentiation, lineage specification, and development. Histone demethylase assays are invaluable in studying histone demethylation substrate recognition, kinetics, regulation, and inhibition by small molecules, many of which are potential therapeutics. Here we describe general procedures to purify recombinant enzymes from different expression hosts, and to prepare a broad range of substrates, as well as to set up a variety of in vitro histone demethylase assays. These assays provide useful tools for discoveries from enzymes to drugs.


Asunto(s)
Histona Demetilasas , Histonas , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Metilación , Procesamiento Proteico-Postraduccional , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA