Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
BMC Plant Biol ; 24(1): 863, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39272009

ABSTRACT

BACKGROUND: Developmental leaf senescence (DLS) is an irreversible process followed by cell death. Dark-induced leaf senescence (DILS) is a reversible process that allows adaptations to changing environmental conditions. As a result of exposure to adverse environmental changes, plants have developed mechanisms that enable them to survive. One of these is the redirection of metabolism into the senescence pathway. The plant seeks to optimise resource allocation. Our research aims to demonstrate how epigenetic machinery regulates leaf senescence, including its irreversibility. RESULTS: In silico analyses allowed the complex identification and characterisation of 117 genes involved in epigenetic processes in barley. These genes include those responsible for DNA methylation, post-translational histone modifications, and ATP-dependent chromatin remodelling complexes. We then performed RNAseq analysis after DILS and DLS to evaluate their expression in senescence-dependent leaf metabolism. Principal component analysis revealed that evaluated gene expression in developmental senescence was similar to controls, while induced senescence displayed a distinct profile. Western blot experiments revealed that senescence engages senescence-specific histone modification. During DILS and DLS, the methylation of histone proteins H3K4me3 and H3K9me2 increased. H3K9ac acetylation levels significantly decreased during DILS and remained unchanged during DLS. CONCLUSIONS: The study identified different epigenetic regulations of senescence types in barley leaves. These findings are valuable for exploring epigenetic regulation of senescence-related molecular mechanisms, particularly in response to premature, induced leaf senescence. Based on the results, we suggest the presence of an epigenetically regulated molecular switch between cell survival and cell death in DILS, highlighting an epigenetically driven cell survival metabolic response.


Subject(s)
Epigenesis, Genetic , Hordeum , Plant Leaves , Plant Senescence , Hordeum/genetics , Hordeum/physiology , Plant Leaves/genetics , Plant Leaves/physiology , Plant Senescence/genetics , Gene Expression Regulation, Plant , DNA Methylation , Histones/metabolism
2.
Aging (Albany NY) ; 16(12): 10489-10511, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38888515

ABSTRACT

Kidney renal clear cell carcinoma (KIRC) is a cancer that is closely associated with epigenetic alterations, and histone modifiers (HMs) are closely related to epigenetic regulation. Therefore, this study aimed to comprehensively explore the function and prognostic value of HMs-based signature in KIRC. HMs were first obtained from top journal. Then, the mRNA expression profiles and clinical information in KIRC samples were downloaded from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) datasets. Cox regression analysis and least absolute shrinkage and selection operator (Lasso) analysis were implemented to find prognosis-related HMs and construct a risk model related to the prognosis in KIRC. Kaplan-Meier analysis was used to determine prognostic differences between high- and low-risk groups. Immune infiltration and drug sensitivity analysis were also performed between high- and low-risk groups. Eventually, 8 HMs were successfully identified for the construction of a risk model in KIRC. The results of the correlation analysis between risk signature and the prognosis showed HMs-based signature has good prognostic value in KIRC. Results of immune analysis of risk models showed there were significant differences in the level of immune cell infiltration and expression of immune checkpoints between high- and low-risk groups. The results of the drug sensitivity analysis showed that the high-risk group was more sensitive to several chemotherapeutic agents such as Sunitinib, Tipifarnib, Nilotinib and Bosutinib than the low-risk group. In conclusion, we successfully constructed HMs-based prognostic signature that can predict the prognosis of KIRC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/drug therapy , Prognosis , Gene Expression Regulation, Neoplastic , Epigenesis, Genetic , Gene Expression Profiling , Histones/metabolism , Histones/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Transcriptome
3.
Cancers (Basel) ; 14(17)2022 Aug 24.
Article in English | MEDLINE | ID: mdl-36077629

ABSTRACT

The use of inhibitors of epigenetic modifiers in the treatment of acute myeloid leukemia (AML) has become increasingly appealing due to the highly epigenetic nature of the disease. We evaluated a library of 164 epigenetic compounds in a cohort of 9 heterogeneous AML patients using an ex vivo drug screen. AML blasts were isolated from bone marrow biopsies according to established protocols and treatment response to the epigenetic library was evaluated. We find that 11 histone deacetylase (HDAC) inhibitors, which act upon mechanisms of cell cycle arrest and apoptotic pathways through inhibition of zinc-dependent classes of HDACs, showed efficacy in all patient-derived samples. Other compounds, including bromodomain and extraterminal domain (BET) protein inhibitors, showed efficacy in most samples. Specifically, HDAC inhibitors are already clinically available and can be repurposed for use in AML. Results in this cohort of AML patient-derived samples reveal several epigenetic compounds with high anti-blast activity in all samples, despite the molecular diversity of the disease. These results further enforce the notion that AML is a predominantly epigenetic disease and that similar epigenetic mechanisms may underlie disease development and progression in all patients, despite differences in genetic mutations.

4.
Adv Exp Med Biol ; 1390: 277-293, 2022.
Article in English | MEDLINE | ID: mdl-36107325

ABSTRACT

The androgen receptor (AR) is a ligand-activated transcription factor belonging to the nuclear receptor (NR) superfamily. As with other members of the NR family, transcriptional activity of the AR is regulated by interactions with coregulatory proteins, which either enhance (coactivators) or repress (corepressors) its transcriptional activity. AR associated coregulators are functionally diverse, but a large fraction are epigenetic histone and chromatin modifiers. Epigenetic coregulators are recruited to gene regulatory regions as part of multi-protein complexes, often acting in a dynamic and inter-dependent manner to remodel chromatin, thereby allowing or inhibiting the access of AR-associated transcriptional machinery to target genes; functional consequences being regulation of transcriptional output. Epigenetic modifiers, including those that function as AR coregulators, are frequently mutated or aberrantly expressed in prostate cancer and are implicated in disease progression. Some of these modifiers are being investigated as therapeutic targets in several cancer types and could potentially be used to modulate aberrant AR activity in prostate cancer. In this chapter we will summarise the functional role of epigenetic coregulators in AR signalling, their dysregulation during prostate cancer progression and the current status of drugs targeting these enzymes.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Chromatin , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Epigenesis, Genetic , Histones/genetics , Histones/metabolism , Humans , Ligands , Male , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcription Factors/metabolism
5.
Arch Microbiol ; 204(7): 411, 2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35729284

ABSTRACT

The epigenome of an organism is as important as the genome for the normal development and functioning of an individual. The human epigenome can be affected by various environmental factors including nutrients, microbiota and probiotics through epigenetic modifiers and mediates various health-promoting effects. The present study was aimed to explore the temporal changes in DNA and histone modifiers (DNMT1, TET2, p300, HDAC1, KMT2A, KDM5B, EzH2 and JMJD3) in intestinal epithelial cells (Caco-2) by probiotic lactobacilli (Limosilactobacillus fermentum MTCC 5898 and Lacticaseibacillus rhamnosus MTCC 5897) in comparison to opportunistic commensal pathogen Escherichia coli (ATCC 14849). Cells were treated separately with probiotic strains and E. coli for different durations and temporal changes in gene expression among DNA and histone modifiers were measured. Time-dependent studies showed that L. fermentum enhanced the transcription of epigenetic modifiers at 12 h of treatment (P < 0.05) contrary to E. coli which reduced the expression of these genes during the same duration of treatment. On the other hand, probiotic L. rhamnosus was not able to induce any significant changes in gene expression of these modifiers. Furthermore, during the exclusion of E. coli by L. fermentum, the probiotic was found to resist the changes made by E. coli in the transcription of some of the epigenetic modifiers. Thus, it is concluded that the probiotics modulated the mRNA expression of DNA and histone modifiers contrarily to E. coli in a strain-specific manner.


Subject(s)
Lactobacillus , Probiotics , Caco-2 Cells , Epigenesis, Genetic , Epithelial Cells , Escherichia coli/genetics , Histones , Humans , Lactobacillus/genetics , Probiotics/pharmacology , RNA, Messenger
6.
Biomolecules ; 13(1)2022 12 28.
Article in English | MEDLINE | ID: mdl-36671446

ABSTRACT

Over the past two decades, novel hallmarks of cancer have been described, including the altered epigenetic landscape of malignant diseases. In addition to the methylation and hyd-roxymethylation of DNA, numerous novel forms of histone modifications and nucleosome remodeling have been discovered, giving rise to a wide variety of targeted therapeutic interventions. DNA hypomethylating drugs, histone deacetylase inhibitors and agents targeting histone methylation machinery are of distinguished clinical significance. The major focus of this review is placed on targeted epigenetic interventions in the most common pediatric malignancies, including acute leukemias, brain and kidney tumors, neuroblastoma and soft tissue sarcomas. Upcoming novel challenges include specificity and potential undesirable side effects. Different epigenetic patterns of pediatric and adult cancers should be noted. Biological significance of epigenetic alterations highly depends on the tissue microenvironment and widespread interactions. An individualized treatment approach requires detailed genetic, epigenetic and metabolomic evaluation of cancer. Advances in molecular technologies and clinical translation may contribute to the development of novel pediatric anticancer treatment strategies, aiming for improved survival and better patient quality of life.


Subject(s)
Neoplasms , Neuroblastoma , Sarcoma , Humans , Child , DNA Methylation/genetics , Epigenesis, Genetic , Quality of Life , Neoplasms/drug therapy , Neoplasms/genetics , Neuroblastoma/genetics , Sarcoma/genetics , Tumor Microenvironment
7.
Biochim Biophys Acta Gene Regul Mech ; 1865(1): 194780, 2022 01.
Article in English | MEDLINE | ID: mdl-34968769

ABSTRACT

In bone biology, epigenetics plays a key role in mesenchymal stem cells' (MSCs) commitment towards osteoblasts. It involves gene regulatory mechanisms governed by chromatin modulators. Predominant epigenetic mechanisms for efficient osteogenic differentiation include DNA methylation, histone modifications, and non-coding RNAs. Among these mechanisms, histone modifications critically contribute to altering chromatin configuration. Histone based epigenetic mechanisms are an essential mediator of gene expression during osteoblast differentiation as it directs the bivalency of the genome. Investigating the importance of histone modifications in osteogenesis may lead to the development of epigenetic-based remedies for genetic disorders of bone. Hence, in this review, we have highlighted the importance of epigenetic modifications such as post-translational modifications of histones, including methylation, acetylation, phosphorylation, ubiquitination, and their role in the activation or suppression of gene expression during osteoblast differentiation. Further, we have emphasized the future advancements in the field of epigenetics towards orthopaedical therapeutics.


Subject(s)
Histones , Osteogenesis , DNA Methylation , Epigenesis, Genetic , Histones/metabolism , Osteoblasts/metabolism , Osteogenesis/genetics
8.
Int J Mol Sci ; 22(16)2021 Aug 17.
Article in English | MEDLINE | ID: mdl-34445553

ABSTRACT

Since their discovery, R-loops have been associated with both physiological and pathological functions that are conserved across species. R-loops are a source of replication stress and genome instability, as seen in neurodegenerative disorders and cancer. In response, cells have evolved pathways to prevent R-loop accumulation as well as to resolve them. A growing body of evidence correlates R-loop accumulation with changes in the epigenetic landscape. However, the role of chromatin modification and remodeling in R-loops homeostasis remains unclear. This review covers various mechanisms precluding R-loop accumulation and highlights the role of chromatin modifiers and remodelers in facilitating timely R-loop resolution. We also discuss the enigmatic role of RNA:DNA hybrids in facilitating DNA repair, epigenetic landscape and the potential role of replication fork preservation pathways, active fork stability and stalled fork protection pathways, in avoiding replication-transcription conflicts. Finally, we discuss the potential role of several Chro-Mates (chromatin modifiers and remodelers) in the likely differentiation between persistent/detrimental R-loops and transient/benign R-loops that assist in various physiological processes relevant for therapeutic interventions.


Subject(s)
Chromatin Assembly and Disassembly , DNA Damage , DNA Repair , DNA Replication , Genomic Instability , R-Loop Structures , Humans
9.
Cancers (Basel) ; 13(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669182

ABSTRACT

Genetic and epigenetic factors contribute to the development of cancer. Epigenetic dysregulation is common in gynaecological cancers and includes altered methylation at CpG islands in gene promoter regions, global demethylation that leads to genome instability and histone modifications. Histones are a major determinant of chromosomal conformation and stability, and unlike DNA methylation, which is generally associated with gene silencing, are amenable to post-translational modifications that induce facultative chromatin regions, or condensed transcriptionally silent regions that decondense resulting in global alteration of gene expression. In comparison, other components, crucial to the manipulation of chromatin dynamics, such as histone modifying enzymes, are not as well-studied. Inhibitors targeting DNA modifying enzymes, particularly histone modifying enzymes represent a potential cancer treatment. Due to the ability of epigenetic therapies to target multiple pathways simultaneously, tumours with complex mutational landscapes affected by multiple driver mutations may be most amenable to this type of inhibitor. Interrogation of the actionable landscape of different gynaecological cancer types has revealed that some patients have biomarkers which indicate potential sensitivity to epigenetic inhibitors. In this review we describe the role of epigenetics in gynaecological cancers and highlight how it may exploited for treatment.

10.
Mol Biol Rep ; 47(9): 7273-7276, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32754863

ABSTRACT

NLRP3 pathway plays a vital role in the pathogenesis of different human cancers but still the regulation of NLRP3 pathway largely unknown. Therefore, we examined the levels of NLRP3 and its downstream components (caspase-1 and IL-1ß) and its relationship with histone modifiers in renal cancer pathogenesis. Total 30 cases of clear cell renal cell carcinoma (ccRCC), were studied for NLRP3, caspase-1 and IL-1ß expression using real-time PCR, which showed the augmented levels of all the three components of NLRP3 inflammasome pathway in ccRCC. Next, role of the FAD dependent monoamine oxidases (LSD2) and jumonji C (JmjC)-domain-containing, iron-dependent dioxygenases (KDM5A) histone demethylases were evaluated in regulation of NLRP3 inflammasome pathway in-vitro using RCC cell line. It was observed that silencing of KDM5A didn't alter the levels of neither of the NLRP3 component but inhibition of LSD2 showed significant effect on NLRP3 expression while no change in caspase-1 and IL-1ß levels. This study suggests that rather LSD2 not KDM5A lysine demethylase family might be involved in the regulation of NLRP3 inflammasome in cancer cells which could be useful for deciphering the future therapeutic targets for the disease.


Subject(s)
Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Inflammasomes/metabolism , Kidney Neoplasms/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasm Proteins/metabolism , Carcinoma, Renal Cell/pathology , Female , Histone Demethylases , Humans , Male , Middle Aged , Pilot Projects
11.
Int J Mol Sci ; 21(9)2020 May 04.
Article in English | MEDLINE | ID: mdl-32375326

ABSTRACT

Vascular calcification (VC) is characterized by calcium deposition inside arteries and is closely associated with the morbidity and mortality of atherosclerosis, chronic kidney disease, diabetes, and other cardiovascular diseases (CVDs). VC is now widely known to be an active process occurring in vascular smooth muscle cells (VSMCs) involving multiple mechanisms and factors. These mechanisms share features with the process of bone formation, since the phenotype switching from the contractile to the osteochondrogenic phenotype also occurs in VSMCs during VC. In addition, VC can be regulated by epigenetic factors, including DNA methylation, histone modification, and noncoding RNAs. Although VC is commonly observed in patients with chronic kidney disease and CVD, specific drugs for VC have not been developed. Thus, discovering novel therapeutic targets may be necessary. In this review, we summarize the current experimental evidence regarding the role of epigenetic regulators including histone deacetylases and propose the therapeutic implication of these regulators in the treatment of VC.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Vascular Calcification/genetics , Vascular Calcification/metabolism , Acetylation , Animals , Biomarkers , DNA Methylation , Gene Expression Regulation , Humans , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteogenesis/genetics , Protein Processing, Post-Translational , Signal Transduction
12.
Glia ; 68(8): 1584-1595, 2020 08.
Article in English | MEDLINE | ID: mdl-32034929

ABSTRACT

Schwann cells (SCs) are the main glial cells present in the peripheral nervous system (PNS). Their primary functions are to insulate peripheral axons to protect them from the environment and to enable fast conduction of electric signals along big caliber axons by enwrapping them in a thick myelin sheath rich in lipids. In addition, SCs have the peculiar ability to foster axonal regrowth after a lesion by demyelinating and converting into repair cells that secrete neurotrophic factors and guide axons back to their former target to finally remyelinate regenerated axons. The different steps of SC development and their role in the maintenance of PNS integrity and regeneration after lesion are controlled by various factors among which transcription factors and chromatin-remodeling enzymes hold major functions. In this review, we discussed how histone modifications and histone-modifying enzymes control SC development, maintenance of PNS integrity and response to injury. The functions of histone modifiers as part of chromatin-remodeling complexes are discussed in another review published in the same issue of Glia.


Subject(s)
Histones/metabolism , Myelin Sheath/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Animals , Axons/metabolism , Axons/pathology , Humans , Myelin Sheath/pathology , Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology
13.
Life Sci ; 238: 116977, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31639400

ABSTRACT

AIMS: In the cell, both transcriptional and translational processes are tightly regulated. Cancer is a multifactorial disease characterized by aberrant protein expression. Since epigenetic control mechanisms are also frequently disrupted during carcinogenesis, they have been the center of attention in cancer research within the past decades. EEF1 complex members, which are required for the elongation process in eukaryotes, have recently been implicated in carcinogenesis. This study aims to investigate genetic alterations within EEF1A1, EEF1A2, EEF1B2, EEF1D, EEF1E1 and EEF1G genes and their potential effects on epigenetic regulation mechanisms. MATERIALS AND METHODS: In this study, we analyzed DNA sequencing and mRNA expression data available on The Cancer Genome Atlas (TCGA) across different cancer types to detect genetic alterations in EEF1 genes and investigated their potential impact on selected epigenetic modulators. KEY FINDINGS: We found that EEF1 complex proteins were deregulated in several types of cancer. Lower EEF1A1, EEF1B2, EEF1D and EEF1G levels were correlated with poor survival in glioma, while lower EEF1B2, EEF1D and EEF1E1 levels were correlated with better survival in hepatocellular carcinoma. We detected genetic alterations within EEF1 genes in up to 35% of the patients and showed that these alterations resulted in down-regulation of histone modifying enzymes KMT2C, KMT2D, KMT2E, KAT6A and EP300. SIGNIFICANCE: Here in this study, we showed that EEF1 deregulations might result in differential epigenomic landscapes, which affect the overall transcriptional profile, contributing to carcinogenesis. Identification of these molecular distinctions might be useful in developing targeted drug therapies and personalized medicine.


Subject(s)
Biomarkers, Tumor/genetics , Carcinogenesis/pathology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/pathology , Peptide Elongation Factor 1/genetics , Carcinogenesis/metabolism , Gene Regulatory Networks , Guanine Nucleotide Exchange Factors/genetics , Humans , Peptide Elongation Factors/genetics , Tumor Suppressor Proteins/genetics
14.
Front Plant Sci ; 10: 1031, 2019.
Article in English | MEDLINE | ID: mdl-31552061

ABSTRACT

Soybean is an important economic crop for human diet, animal feeds and biodiesel due to high protein and oil content. Its productivity is significantly hampered by salt stress, which impairs plant growth and development by affecting gene expression, in part, through epigenetic modification of chromatin status. However, little is known about epigenetic regulation of stress response in soybean roots. Here, we used RNA-seq and ChIP-seq technologies to study the dynamics of genome-wide transcription and histone methylation patterns in soybean roots under salt stress. Eight thousand seven hundred ninety eight soybean genes changed their expression under salt stress treatment. Whole-genome ChIP-seq study of an epigenetic repressive mark, histone H3 lysine 27 trimethylation (H3K27me3), revealed the changes in H3K27me3 deposition during the response to salt stress. Unexpectedly, we found that most of the inactivation of genes under salt stress is strongly correlated with the de novo establishment of H3K27me3 in various parts of the promoter or coding regions where there is no H3K27me3 in control plants. In addition, the soybean histone modifiers were identified which may contribute to de novo histone methylation and gene silencing under salt stress. Thus, dynamic chromatin regulation, switch between active and inactive modes, occur at target loci in order to respond to salt stress in soybean. Our analysis demonstrates histone methylation modifications are correlated with the activation or inactivation of salt-inducible genes in soybean roots.

15.
Int J Mol Sci ; 20(17)2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31443358

ABSTRACT

DNA damage response (DDR) in eukaryotic cells is initiated in the chromatin context. DNA damage and repair depend on or have influence on the chromatin dynamics associated with genome stability. Epigenetic modifiers, such as chromatin remodelers, histone modifiers, DNA (de-)methylation enzymes, and noncoding RNAs regulate DDR signaling and DNA repair by affecting chromatin dynamics. In recent years, significant progress has been made in the understanding of plant DDR and DNA repair. SUPPRESSOR OF GAMMA RESPONSE1, RETINOBLASTOMA RELATED1 (RBR1)/E2FA, and NAC103 have been proven to be key players in the mediation of DDR signaling in plants, while plant-specific chromatin remodelers, such as DECREASED DNA METHYLATION1, contribute to chromatin dynamics for DNA repair. There is accumulating evidence that plant epigenetic modifiers are involved in DDR and DNA repair. In this review, I examine how DDR and DNA repair machineries are concertedly regulated in Arabidopsis thaliana by a variety of epigenetic modifiers directing chromatin remodeling and epigenetic modification. This review will aid in updating our knowledge on DDR and DNA repair in plants.


Subject(s)
Chromatin Assembly and Disassembly , DNA Damage , Epigenesis, Genetic , Gene Expression Regulation, Plant , Plants/genetics , Acetylation , Chromatin/genetics , Chromatin/metabolism , DNA Methylation , DNA Repair , Histones/metabolism , Plants/metabolism , Signal Transduction
16.
Immunol Rev ; 288(1): 75-84, 2019 03.
Article in English | MEDLINE | ID: mdl-30874352

ABSTRACT

The humoral immune response requires coordination of molecular programs to mediate differentiation into unique B cell subsets that help clear the infection and form immune memory. Epigenetic modifications are crucial for ensuring that the appropriate genes are transcribed or repressed during B cell differentiation. Recent studies have illuminated the changes in DNA methylation and histone post-translational modifications that accompany the formation of germinal center and antibody-secreting cells during an immune response. In particular, the B cell subset-specific expression and function of DNA methyltransferases and histone-modifying complexes that mediate epigenome changes have begun to be unravelled. This review will discuss the recent advances in this field, as well as highlight critical questions about the relationship between epigenetic regulation and B cell fate and function that are yet to be answered.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Epigenesis, Genetic/immunology , Germinal Center/immunology , Animals , Antibody Formation , Cell Differentiation/genetics , DNA Methylation , Gene Expression Regulation , Humans
17.
Oncologist ; 24(3): 319-326, 2019 03.
Article in English | MEDLINE | ID: mdl-30018131

ABSTRACT

BACKGROUND: The incidence of colorectal cancer (CRC) in younger patients is rising, mostly due to tumors in the descending colon and rectum. Therefore, we aimed to explore the molecular differences of left-sided CRC between younger (≤45 years) and older patients (≥65). SUBJECTS, MATERIALS, AND METHODS: In total, 1,126 CRC tumor samples from the splenic flexure to (and including) the rectum were examined by next-generation sequencing (NGS), immunohistochemistry, and in situ hybridization. Microsatellite instability (MSI) and tumor mutational burden (TMB) were assessed by NGS. RESULTS: Younger patients (n = 350), when compared with older patients (n = 776), showed higher mutation rates in genes associated with cancer-predisposing syndromes (e.g., Lynch syndrome), such as MSH6 (4.8% vs. 1.2%, p = .005), MSH2 (2.7% vs. 0.0%, p = .004), POLE (1.6% vs. 0.0%, p = .008), NF1 (5.9% vs. 0.5%, p < .001), SMAD4 (14.3% vs. 8.3%, p = .024), and BRCA2 (3.7% vs. 0.5%, p = .002). Genes involved in histone modification were also significantly more mutated: KDM5C (1.9% vs. 0%, p = .036), KMT2A (1.1% vs. 0%, p = .033), KMT2C (1.6% vs. 0%, p = .031), KMT2D (3.8% vs. 0.7%, p = .005), and SETD2 (3.2% vs. 0.9%, p = .039). Finally, TMB-high (9.7% vs. 2.8%, p < .001) and MSI-high (MSI-H; 8.1% vs. 1.9%, p = .009) were more frequent in younger patients. CONCLUSION: Our findings highlight the importance of genetic counseling and screening in younger CRC patients. MSI-H and TMB-high tumors could benefit from immune-checkpoint inhibitors, now approved for the treatment of MSI-H/deficient mismatch repair metastatic CRC patients. Finally, histone modifiers could serve as a new promising therapeutic target. With confirmatory studies, these results may influence our approach to younger adults with CRC. IMPLICATIONS FOR PRACTICE: The increasing rate of colorectal cancers (CRC), primarily distal tumors, among young adults poses a global health issue. This study investigates the molecular differences between younger (≤45 years old) and older (≥65) adults with left-sided CRCs. Younger patients more frequently harbor mutations in genes associated with cancer-predisposing syndromes. Higher rates of microsatellite instability-high and tumor mutational burden-high tumors occur in younger patients, who could benefit from immune-checkpoint inhibitors. Finally, histone modifiers are more frequently mutated in younger patients and could serve as a new promising therapeutic target. This study provides new insights into mutations that may guide development of novel tailored therapy in younger CRC patients.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Adult , Age Factors , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Female , Humans , Incidence , Male , Middle Aged , Young Adult
18.
Front Plant Sci ; 8: 639, 2017.
Article in English | MEDLINE | ID: mdl-28496451

ABSTRACT

Modifications to histones, including acetylation and methylation processes, play crucial roles in the regulation of gene expression in plant development as well as in stress responses. However, limited information on the enzymes catalyzing histone acetylation and methylation in non-model plants is currently available. In this study, several histone modifier (HM) types, including six histone acetyltransferases (HATs), 11 histone deacetylases (HDACs), 48 histone methyltransferases (HMTs), and 22 histone demethylases (HDMs), are identified in litchi (Litchi chinensis Sonn. cv. Feizixiao) based on similarities in their sequences to homologs in Arabidopsis (A. thaliana), tomato (Solanum lycopersicum), and rice (Oryza sativa). Phylogenetic analyses reveal that HM enzymes can be grouped into four HAT, two HDAC, two HMT, and two HDM subfamilies, respectively, while further expression profile analyses demonstrate that 17 HMs were significantly altered during fruit abscission in two field treatments. Analyses reveal that these genes exhibit four distinct patterns of expression in response to fruit abscission, while an in vitro assay was used to confirm the HDAC activity of LcHDA2, LcHDA6, and LcSRT2. Our findings are the first in-depth analysis of HMs in the litchi genome, and imply that some are likely to play important roles in fruit abscission in this commercially important plant.

19.
Prog Biophys Mol Biol ; 118(1-2): 8-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25841987

ABSTRACT

Epigenetic reprogramming that occurs during critical periods of development can increase the susceptibility to many diseases in adulthood. Programming of the epigenome during development occurs via the activity of a variety of epigenetic modifiers, including "readers, writers and erasers" of histone methyl marks. Posttranslational modification of these programmers can alter their activity, resulting in global or gene-specific changes in histone methylation and gene transcription. This review summarizes what is currently known about phosphorylation of histone methyltransferases ("writers"), demethylases ("erasers") and effector proteins ("readers) that program the epigenome, and the impact of this posttranslational modification on their activity. Understanding how the activity of these epigenetic programmers is perturbed by environmental exposures via changes in phosphorylation is key to understanding mechanisms of developmental reprogramming and the epigenetic basis of health and disease.


Subject(s)
Disease/genetics , Epigenesis, Genetic , Growth and Development/genetics , Health , Animals , Carcinogens, Environmental/toxicity , Epigenesis, Genetic/drug effects , Growth and Development/drug effects , Humans , Phosphorylation/drug effects
20.
Reprod Toxicol ; 54: 136-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25554384

ABSTRACT

Environmental and lifestyle factors are considered significant components of the increasing breast cancer risk in the last 50 years. Specifically, exposure to environmental endocrine disrupting compounds is correlated with cancer susceptibility in a variety of tissues. In both human and rodent models, the exposure to ubiquitous environmental estrogens during early life has been shown to disrupt normal mammary development and cause permanent adverse effects. Recent studies indicate that environmental estrogens not only have the ability to disrupt estrogen receptor (ER) signaling, but can also reprogram the epigenome by altering DNA and histone methylation through rapid, nongenomic ER actions. We have observed xenoestrogen-mediated activation of several nongenomic signaling pathways and have identified a target for epigenetic reprogramming in MCF-7 breast cancer cells. These observations, in addition to data from the literature, support the hypothesis that activation of rapid signaling by environmental estrogens can lead to epigenetic reprogramming and contribute to the progression of breast cancer.


Subject(s)
Breast Neoplasms/chemically induced , Carcinogens, Environmental/toxicity , Cell Transformation, Neoplastic/chemically induced , Cellular Reprogramming/drug effects , Endocrine Disruptors/toxicity , Environmental Exposure/adverse effects , Epigenesis, Genetic/drug effects , Estrogens/toxicity , Signal Transduction/drug effects , Age Factors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Risk Assessment , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL