Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Alcohol ; 117: 27-41, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508286

ABSTRACT

While DNA serves as the fundamental genetic blueprint for an organism, it is not a static entity. Gene expression, the process by which genetic information is utilized to create functional products like proteins, can be modulated by a diverse range of environmental factors. Epigenetic mechanisms, including DNA methylation, histone modification, and microRNAs, play a pivotal role in mediating the intricate interplay between the environment and gene expression. Intriguingly, alterations in the epigenome have the potential to be inherited across generations. Alcohol use disorder (AUD) poses significant health issues worldwide. Alcohol has the capability to induce changes in the epigenome, which can be inherited by offspring, thus impacting them even in the absence of direct alcohol exposure. This review delves into the impact of alcohol on the epigenome, examining how its effects vary based on factors such as the age of exposure (adolescence or adulthood), the duration of exposure (chronic or acute), and the specific sample collected (brain, blood, or sperm). The literature underscores that alcohol exposure can elicit diverse effects on the epigenome during different life stages. Furthermore, compelling evidence from human and animal studies demonstrates that alcohol induces alterations in epigenome content, affecting both the brain and blood. Notably, rodent studies suggest that these epigenetic changes can result in lasting phenotype alterations that extend across at least two generations. In conclusion, the comprehensive literature analysis supports the notion that alcohol exposure induces lasting epigenetic alterations, influencing the behavior and health of future generations. This knowledge emphasizes the significance of addressing the potential transgenerational effects of alcohol and highlights the importance of preventive measures to minimize the adverse impact on offspring.


Subject(s)
Alcoholism , Epigenome , Animals , Female , Humans , Male , Alcohol Drinking/genetics , Alcohol Drinking/adverse effects , Alcoholism/genetics , Brain/drug effects , Brain/metabolism , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Epigenome/drug effects , Ethanol/pharmacology , MicroRNAs/genetics
2.
Cancer Res ; 83(2): 167-169, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36651076

ABSTRACT

While the goal of most anticancer treatments is to kill cancer cells, some therapies halt cancer progression by inducing cancer cell differentiation. For example, retinoic acid induces neuroblastoma cell differentiation in vitro and is used as maintenance therapy for children with high-risk neuroblastoma. A new study by Jiang and colleagues has revealed the mitochondrial uncoupler niclosamide ethanolamine (NEN) induces neuroblastoma cell differentiation in vitro and slows neuroblastoma tumor growth in vivo. Mitochondrial uncoupler molecules alter cell metabolism by forcing cells to "burn" more nutrients, resulting in a switch from anabolic to catabolic metabolism. NEN-induced neuroblastoma cell differentiation was associated with disruption of Warburg metabolism, epigenetic remodeling, and downregulation of key oncogenic drivers of neuroblastoma development, including MYCN. NEN is currently used as an antiparasitic worm treatment and is safe to use in children but has poor pharmacokinetic properties. However, derivatives of NEN and structurally distinct uncouplers that have improved pharmacokinetic properties are in development. Results of this study ignite the idea that mitochondrial uncouplers could be used as differentiating agents and expand the pharmacotherapy toolkit to treat cancer, including neuroblastoma. See related article by Jiang et al., p. 181.


Subject(s)
Antineoplastic Agents , Neuroblastoma , Humans , Epigenome/drug effects , Antineoplastic Agents/pharmacology , Tretinoin/pharmacology , Cell Differentiation/drug effects , Niclosamide/pharmacology , Ethanolamine/pharmacology , Ethanolamine/therapeutic use , Neuroblastoma/pathology
3.
Cell Cycle ; 22(8): 870-905, 2023 04.
Article in English | MEDLINE | ID: mdl-36648057

ABSTRACT

Disruption of the complex network that regulates redox homeostasis often underlies resistant phenotypes, which hinder effective and long-lasting cancer eradication. In addition, the RNA methylome-dependent control of gene expression also critically affects traits of cellular resistance to anti-cancer agents. However, few investigations aimed at establishing whether the epitranscriptome-directed adaptations underlying acquired and/or innate resistance traits in cancer could be implemented through the involvement of redox-dependent or -responsive signaling pathways. This is unexpected mainly because: i) the effectiveness of many anti-cancer approaches relies on their capacity to promote oxidative stress (OS); ii) altered redox milieu and reprogramming of mitochondrial function have been acknowledged as critical mediators of the RNA methylome-mediated response to OS. Here we summarize the current state of understanding on this topic, as well as we offer new perspectives that might lead to original approaches and strategies to delay or prevent the problem of refractory cancer and tumor recurrence.


Subject(s)
Antineoplastic Agents , Neoplasms , Oxidative Stress , RNA Processing, Post-Transcriptional , RNA , RNA/genetics , RNA/metabolism , Methylation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Oxidative Stress/drug effects , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Epigenesis, Genetic/drug effects , Transcriptome/drug effects , Oxidation-Reduction/drug effects , Epigenome/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Recurrence , Humans , Animals , RNA Processing, Post-Transcriptional/drug effects , Homeostasis/drug effects , Reactive Oxygen Species/metabolism
5.
J Clin Endocrinol Metab ; 107(1): 150-166, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34487152

ABSTRACT

CONTEXT: Chronic glucocorticoid (GC) overexposure, resulting from endogenous Cushing's syndrome (CS) or exogenous GC therapy, causes several adverse outcomes, including persistent central fat accumulation associated with a low-grade inflammation. However, no previous multiomics studies in visceral adipose tissue (VAT) from patients exposed to high levels of unsuppressed GC during active CS or after remission are available yet. OBJECTIVE: To determine the persistent VAT transcriptomic alterations and epigenetic fingerprints induced by chronic hypercortisolism. METHODS: We employed a translational approach combining high-throughput data on endogenous CS patients and a reversible CS mouse model. We performed RNA sequencing and chromatin immunoprecipitation sequencing on histone modifications (H3K4me3, H3K27ac, and H3K27me3) to identify persistent transcriptional and epigenetic signatures in VAT produced during active CS and maintained after remission. RESULTS: VAT dysfunction was associated with low-grade proinflammatory status, macrophage infiltration, and extracellular matrix remodeling. Most notably, chronic hypercortisolism caused a persistent circadian rhythm disruption in VAT through core clock genes modulation. Importantly, changes in the levels of 2 histone modifications associated to gene transcriptional activation (H3K4me3 and H3K27ac) correlated with the observed differences in gene expression during active CS and after CS remission. CONCLUSION: We identified for the first time the persistent transcriptional and epigenetic signatures induced by hypercortisolism in VAT, providing a novel integrated view of molecular components driving the long-term VAT impairment associated with CS.


Subject(s)
Adrenal Gland Neoplasms/complications , Cushing Syndrome/metabolism , Glucocorticoids/adverse effects , Intra-Abdominal Fat/immunology , Obesity, Abdominal/genetics , Administration, Oral , Adrenal Gland Neoplasms/diagnosis , Adrenal Gland Neoplasms/immunology , Adrenal Gland Neoplasms/urine , Adult , Animals , Biopsy , Chromatin Immunoprecipitation Sequencing , Corticosterone/administration & dosage , Corticosterone/adverse effects , Cross-Sectional Studies , Cushing Syndrome/immunology , Cushing Syndrome/pathology , Disease Models, Animal , Epigenome/drug effects , Epigenome/immunology , Female , Glucocorticoids/administration & dosage , Glucocorticoids/metabolism , Humans , Hydrocortisone/metabolism , Hydrocortisone/urine , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Male , Mice , Middle Aged , Obesity, Abdominal/immunology , Obesity, Abdominal/pathology , RNA-Seq , Transcriptome/drug effects , Transcriptome/immunology
6.
Cell Rep ; 37(3): 109836, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686347

ABSTRACT

Clinical evidence suggests that rapid and sustained antidepressant action can be attained with a single exposure to psychedelics. However, the biological substrates and key mediators of psychedelics' enduring action remain unknown. Here, we show that a single administration of the psychedelic DOI produces fast-acting effects on frontal cortex dendritic spine structure and acceleration of fear extinction via the 5-HT2A receptor. Additionally, a single dose of DOI leads to changes in chromatin organization, particularly at enhancer regions of genes involved in synaptic assembly that stretch for days after the psychedelic exposure. These DOI-induced alterations in the neuronal epigenome overlap with genetic loci associated with schizophrenia, depression, and attention deficit hyperactivity disorder. Together, these data support that epigenomic-driven changes in synaptic plasticity sustain psychedelics' long-lasting antidepressant action but also warn about potential substrate overlap with genetic risks for certain psychiatric conditions.


Subject(s)
Amphetamines/pharmacology , Dendritic Spines/drug effects , Epigenesis, Genetic/drug effects , Epigenome/drug effects , Frontal Lobe/drug effects , Hallucinogens/pharmacology , Neuronal Plasticity/drug effects , Receptor, Serotonin, 5-HT2A/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology , Synapses/drug effects , Animals , Behavior, Animal/drug effects , Dendritic Spines/metabolism , Epigenomics , Extinction, Psychological/drug effects , Fear/drug effects , Frontal Lobe/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/metabolism , Synapses/metabolism , Time Factors
7.
J Toxicol Environ Health A ; 84(24): 1004-1019, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34459362

ABSTRACT

Cigarette smoking is a major cause of lung cancer. Although tobacco smoking-induced genotoxicity has been well established, there is apparent lack of abundance functional epigenetic effects reported On cigarette smoke-induced lung carcinogenesis. The aim of this study was to determine effects of intratracheal administration of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) utilizing target gene expression DNA methylation patterns in lung tissues of mice following twice weekly for 8 weeks treatment. An unbiased approach where genomic regions was undertaken to assess early methylation changes within mouse pulmonary tissues. A methylated-CpG island recovery assay (MIRA) was performed to map the DNA methylome in lung tissues, with the position of methylated DNA determined using a Genome Analyzer (MIRA-SEQ). Alterations in epigenetic-regulated target genes were confirmed with quantitative reverse transcription-PCR, which revealed 35 differentially hypermethylated genes including Cdkn1C, Hsf4, Hnf1a, Cdx1, and Hoxa5 and 30 differentially hypomethylated genes including Ddx4, Piwi1, Mdm2, and Pce1 in NNK-exposed lung tissue compared with controls. The main pathway of these genes for mediating biological information was analyzed using the Kyoto Encyclopedia of Genes and Genomes database. Among them, Rssf1 and Mdm2 were closely associated with NNK-induced lung carcinogenesis. Taken together, our data provide valuable resources for detecting cigarette smoke-induced lung carcinogenesis.


Subject(s)
Carcinogenesis/chemically induced , Carcinogens/toxicity , Epigenesis, Genetic/drug effects , Lung/drug effects , Nitrosamines/toxicity , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogens/analysis , DNA Methylation/drug effects , Epigenome/drug effects , Gene Expression/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Nitrosamines/analysis , Tobacco Smoking/adverse effects
8.
Genes (Basel) ; 12(7)2021 07 19.
Article in English | MEDLINE | ID: mdl-34356111

ABSTRACT

Prenatal alcohol exposure is one of the most significant causes of developmental disability in the Western world. Maternal alcohol consumption during pregnancy leads to an increased risk of neurological deficits and developmental abnormalities in the fetus. Over the past decade, several human and animal studies have demonstrated that alcohol causes alterations in epigenetic marks, including DNA methylation, histone modifications, and non-coding RNAs. There is an increasing amount of evidence that early pregnancy is a sensitive period for environmental-induced epigenetic changes. It is a dynamic period of epigenetic reprogramming, cell divisions, and DNA replication and, therefore, a particularly interesting period to study the molecular changes caused by alcohol exposure as well as the etiology of alcohol-induced developmental disorders. This article will review the current knowledge about the in vivo and in vitro effects of alcohol exposure on the epigenome, gene regulation, and the phenotype during the first weeks of pregnancy.


Subject(s)
Alcohol Drinking/adverse effects , Embryonic Development/drug effects , Epigenome/genetics , Animals , DNA Methylation/drug effects , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Epigenome/drug effects , Female , Fetus/drug effects , Fetus/physiology , Histone Code , Histones/metabolism , Humans , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/genetics
9.
Biol Reprod ; 105(3): 720-734, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34258621

ABSTRACT

Each year, millions of infants and children are anesthetized for medical and surgical procedures. Yet, a substantial body of preclinical evidence suggests that anesthetics are neurotoxins that cause rapid and widespread apoptotic cell death in the brains of infant rodents and nonhuman primates. These animals have persistent impairments in cognition and behavior many weeks or months after anesthesia exposure, leading us to hypothesize that anesthetics do more than simply kill brain cells. Indeed, anesthetics cause chronic neuropathology in neurons that survive the insult, which then interferes with major aspects of brain development, synaptic plasticity, and neuronal function. Understanding the phenomenon of anesthesia-induced developmental neurotoxicity is of critical public health importance because clinical studies now report that anesthesia in human infancy is associated with cognitive and behavioral deficits. In our search for mechanistic explanations for why a young and pliable brain cannot fully recover from a relatively brief period of anesthesia, we have accumulated evidence that neonatal anesthesia can dysregulate epigenetic tags that influence gene transcription such as histone acetylation and DNA methylation. In this review, we briefly summarize the phenomenon of anesthesia-induced developmental neurotoxicity. We then discuss chronic neuropathology caused by neonatal anesthesia, including disturbances in cognition, socio-affective behavior, neuronal morphology, and synaptic plasticity. Finally, we present evidence of anesthesia-induced genetic and epigenetic dysregulation within the developing brain that may be transmitted intergenerationally to anesthesia-naïve offspring.


Subject(s)
Anesthesia/adverse effects , Animals, Newborn/genetics , Epigenome/drug effects , Primates/genetics , Animals , Humans , Infant, Newborn , Mice , Rats
10.
Antiviral Res ; 192: 105103, 2021 08.
Article in English | MEDLINE | ID: mdl-34082058

ABSTRACT

The ability to establish, and reactivate from, latent infections is central to the biology and pathogenesis of HSV-1. It also poses a strong challenge to antiviral therapy, as latent HSV-1 genomes do not replicate or express any protein to be targeted. Although the processes regulating the establishment and maintenance of, and reactivation from, latency are not fully elucidated, the current general consensus is that epigenetics play a major role. A unifying model postulates that whereas HSV-1 avoids or counteracts chromatin silencing in lytic infections, it becomes silenced during latency, silencing which is somewhat disrupted during reactivation. Many years of work by different groups using a variety of approaches have also shown that the lytic HSV-1 chromatin is distinct and has unique biophysical properties not shared with most cellular chromatin. Nonetheless, the lytic and latent viral chromatins are typically enriched in post translational modifications or histone variants characteristic of active or repressed transcription, respectively. Moreover, a variety of small molecule epigenetic modulators inhibit viral replication and reactivation from latency. Despite these successes in culture and animal models, it is not obvious how epigenetic modulation would be used in antiviral therapy if the same epigenetic mechanisms governed viral and cellular gene expression. Recent work has highlighted several important differences between the viral and cellular chromatins, which appear to be of consequence to their respective epigenetic regulations. In this review, we will discuss the distinctiveness of the viral chromatin, and explore whether it is regulated by mechanisms unique enough to be exploited in antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Chromatin/genetics , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Viral/drug effects , Herpesvirus 1, Human/drug effects , Animals , Antiviral Agents/therapeutic use , Chromatin/drug effects , Epigenome/drug effects , Herpes Simplex/drug therapy , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Virus Latency/drug effects , Virus Latency/genetics
11.
Recent Pat Anticancer Drug Discov ; 16(3): 377-392, 2021.
Article in English | MEDLINE | ID: mdl-33888051

ABSTRACT

BACKGROUND: Bee venom is a promising agent for cancer treatment due to its selective cytotoxic potential for cancer cells through apoptotic pathways. However, there is no evidence for changes in the epigenome and mitochondrial DNA copy numbers after bee venom application. The purpose of this study was to determine the impact of bee venom on cytosine modifications and mitochondrial DNA copy number variation. METHODS: A broad range of methods was applied to elucidate the impact of bee venom on neoplastic cells. These included MTT assay for detection of cytotoxicity, immunostaining of cytosine modifications and mitochondria, assessment of cellular morphology by flow cytometry, and quantification of mitochondrial DNA copy numbers using QPCR. RESULTS: Bee venom-induced cell death was selective for cancer cells, where it triggered a response characterized by alteration of cytosine modification. In contrast, normal cells were more resistant to DNA modifications. Furthermore, application of the venom resulted in variation of mitochondrial membrane permeability and mitochondrial DNA copy numbers, together with alterations in cell morphology, manifesting as reduced affected cell size. CONCLUSION: The study findings suggest that bee venom can be used as a selective DNA (de)methylating agent in cancer. Various agents (such as decitabine and 5-azacytidine) have been synthesized and developed for cancer treatment, and a range of syntheses and preparation and application methods have been described for these patented drugs. However, to the best of our knowledge, no previous research has investigated the use of bee venom or any component thereof for epigenetic therapy in cancer cells.


Subject(s)
Bee Venoms/pharmacology , DNA, Mitochondrial/drug effects , Epigenome/drug effects , Mitochondria/drug effects , Animals , Apitherapy , Cell Line, Tumor , Cell Shape , Cell Size , DNA Copy Number Variations/drug effects , Epigenesis, Genetic/drug effects , Epigenome/genetics , Hep G2 Cells , Humans , Mice , Mitochondria/genetics , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , NIH 3T3 Cells , Permeability/drug effects
12.
Mol Carcinog ; 60(6): 391-402, 2021 06.
Article in English | MEDLINE | ID: mdl-33848375

ABSTRACT

Epigenetics/epigenomics has been shown to be involved in carcinogenesis. However, how the epigenome would be altered in the transgenic adenocarcinoma of the mouse prostate (TRAMP) cancer model and the effect of cancer chemopreventive phytochemical phenethyl isothiocyanate (PEITC) on the epigenome in TRAMP mice are not known. PEITC has been reported to reduce the risk of many cancers including prostate cancer (PCa). In this study, male TRAMP mice were fed a control diet or diet containing 0.05% PEITC from 8 weeks to 16 weeks. The tumor incidence was reduced in the PEITC diet (0/6) as compared with the control diet (6/7). RNA-sequencing (RNA-seq) analyses on nontumor and tumor prostatic tissues revealed several pathways like cell cycle/Cdc42 signaling, inflammation, and cancer-related signaling, were activated in prostate tissues of TRAMP mice but were reversed or attenuated in TRAMP mice fed with PEITC diet. DNA CpG methyl-seq analyses showed that global methylation patterns of prostate samples from TRAMP mice were hugely different from those of wild-type mice. Dietary PEITC partially reversed the global methylation changes during prostatic carcinogenesis. Integration of RNA-seq and DNA methyl-seq analyses identified a list of genes, including Adgrb1 and Ebf4, with an inverse regulatory relationship between their RNA expression and CpG methylation. In summary, our current study demonstrates that alteration of the global epigenome in TRAMP prostate tumor and PEITC administration suppresses PCa carcinogenesis, impacts global CpG epigenome and transcriptome, and attenuates carcinogenic pathways like cell cycle arrest and inflammation. These results may provide insights and epigenetic markers/targets for PCa prevention and treatment in human PCa patients.


Subject(s)
Anticarcinogenic Agents/pharmacology , DNA Methylation/drug effects , Isothiocyanates/pharmacology , Prostatic Neoplasms/prevention & control , Animals , Epigenome/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/genetics , Prostatic Neoplasms/genetics
13.
Mol Neurobiol ; 58(8): 3759-3768, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33826069

ABSTRACT

N6-methyladenosine (m6A) is the most prevalent internal modification found in mRNAs and lncRNA and plays a vital role in posttranscriptional regulation in mammals. m6A is abundant in the nervous system, where it modulates neuronal development and hippocampus-dependent learning and memory. However, the roles of RNAs m6A modification and its related enzymes in cocaine reward are still not fully understood. In this study, we found that the fat mass and obesity-associated gene (FTO) demethylase, but not methyltransferase-like 3 (METTL3) and 14 (METTL14), was downregulated in the hippocampus following cocaine-induced conditioned place preference (CPP), and the level of m6A is notably higher in the hippocampus of cocaine CPP training mice. Using methylated m6A RNA immunoprecipitation sequencing (MeRIP-m6A-seq), we identified a total of 6516 m6A peaks within 4460 mRNAs, and 3083 m6A peaks within 850 lncRNAs were significantly dysregulated. Intriguingly, the altered m6A peaks within mRNAs and lncRNAs were enriched in synapse maturation and localization processes. Our study uncovers a critical role for an m6A epitranscriptomic dysregulation and downregulation of FTO expression in the hippocampus following cocaine-induced CPP.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/biosynthesis , Cocaine/administration & dosage , Conditioning, Psychological/physiology , Hippocampus/metabolism , Membrane Glycoproteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Sequence Analysis, RNA/methods , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/antagonists & inhibitors , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Animals , Conditioning, Psychological/drug effects , Epigenome/drug effects , Epigenome/physiology , Hippocampus/drug effects , Male , Membrane Glycoproteins/genetics , Methylation/drug effects , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics
14.
Int J Mol Sci ; 22(4)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672782

ABSTRACT

Parental exposure to bisphenol A (BPA) has been linked to a greater incidence of congenital diseases. We have demonstrated that BPA induces in zebrafish males an increase in the acetylation of sperm histones that is transmitted to the blastomeres of the unexposed progeny. This work is aimed to determine whether histone hyperacetylation promoted by paternal exposure to BPA is the molecular mechanism underlying the cardiogenesis impairment in the descendants. Zebrafish males were exposed to 100 and 2000 µg/L BPA during early spermatogenesis and mated with non-exposed females. We analyzed in the progeny the expression of genes involved in cardiogenesis and the epigenetic profile. Once the histone hyperacetylation was confirmed, treatment with epigallocatechin gallate (EGCG), an inhibitor of histone acetyltransferases, was assayed on F1 embryos. Embryos from males exposed to 2000 µg/L BPA overexpressed the transcription factor hand2 and the receptor esr2b, showing their own promoters-as well as that of kat6a-an enrichment in H3K9ac. In embryos treated with EGCG, both gene expression and histone acetylation (global and specific) returned to basal levels, and the phenotype was recovered. As shown by the results, the histone hyperacetylated landscape promoted by BPA in the sperm alters the chromatin structure of the progeny, leading to the overexpression of the histone acetyltransferase and genes involved in cardiogenesis.


Subject(s)
Benzhydryl Compounds/toxicity , Cardiotoxicity/genetics , Epigenome/genetics , Paternal Inheritance/genetics , Phenols/toxicity , Spermatozoa/metabolism , Acetylation , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Embryo, Nonmammalian/metabolism , Epigenesis, Genetic/drug effects , Epigenome/drug effects , Histones/metabolism , Male , Spermatozoa/drug effects , Transcriptome/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Curr Cancer Drug Targets ; 21(4): 306-325, 2021.
Article in English | MEDLINE | ID: mdl-33535953

ABSTRACT

Epigenetic modulation of gene expression is essential for tissue-specific development and maintenance in mammalian cells. Disruption of epigenetic processes, and the subsequent alteration of gene functions, can result in inappropriate activation or inhibition of various cellular signaling pathways, leading to cancer. Recent advancements in the understanding of the role of epigenetics in cancer initiation and progression have uncovered functions for DNA methylation, histone modifications, nucleosome positioning, and non-coding RNAs. Epigenetic therapies have shown some promise for hematological malignancies, and a wide range of epigenetic-based drugs are undergoing clinical trials. However, in a dynamic survival strategy, cancer cells exploit their heterogeneous population which frequently results in the rapid acquisition of therapy resistance. Here, we describe novel approaches in drug discovery targeting the epigenome, highlighting recent advances the selective degradation of target proteins using Proteolysis Targeting Chimera (PROTAC) to address drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Molecular Targeted Therapy , Neoplasms , Proteolysis , Drug Discovery/methods , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Epigenome/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Neoplasms/drug therapy , Neoplasms/genetics
16.
Epigenomics ; 13(4): 285-297, 2021 02.
Article in English | MEDLINE | ID: mdl-33401928

ABSTRACT

Aims: Paternal age is increasing in developed countries. Understanding of aging-related epigenetic changes in sperm is needed as well as factors that modify such changes. Materials & methods: Young pubertal and mature rats were exposed perinatally to vehicle or environmental xenobiotic 2,2',4,4'-tetrabromodiphenyl ether. Epididymal sperm was reduced representation bisulfite sequenced. Differentially methylated regions (DMRs) were identified via MethPipe. Results: In control animals, 5319 age-dependent DMRs were identified. Age-related DMRs were enriched for embryonic development. In exposed rats, DNA methylation was higher in young and lower in mature animals then in controls. Conclusions: Sperm methylome undergoes significant age-dependent changes, which may represent a causal link between paternal age and offspring phenotype. Environmental xenobiotics can interfere with the natural process of epigenetic aging.


Subject(s)
Aging/physiology , DNA Methylation/drug effects , Flame Retardants/adverse effects , Spermatozoa/drug effects , Animals , Embryonic Development/drug effects , Epigenesis, Genetic/drug effects , Epigenome/drug effects , Epigenomics/methods , Female , Male , Parturition/drug effects , Paternal Age , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar
17.
J Drug Target ; 29(1): 69-77, 2021 01.
Article in English | MEDLINE | ID: mdl-32672115

ABSTRACT

Post-replicative DNA methylation is essential for diverse biological processes in both eukaryotes and prokaryotes. Mycobacterium tuberculosis (M. tuberculosis), the causative agent of tuberculosis, remains one of the most formidable threats worldwide. Although DNA methylation of M. tuberculosis has been documented, little information is available for clinical drug-resistant M. tuberculosis. Single-molecule real-time (SMRT) sequencing was used to profile the core methylome of three clinical isolates, namely multidrug-resistant (MDR), extensively drug-resistant (XDR) and extremely drug-resistant (XXDR) strains. 3812, 6808 and 6041 DNA methylated sites were identified in MDR-MTB, XDR-MTB and XXDR-MTB genome, respectively. There are two types of methylated motifs, namely N6-methyladenine (m6A) and N4-methylcytosine (m4C). A novel widespread 6 mA methylation motif 5'-CACGCAG-3' was found in XDR-MTB and XXDR-MTB. The methylated genes are involved in multiple cellular processes, especially metabolic enzymes engaged in glucose metabolism, fatty acid and TCA cycle. Many methylated genes are involved in mycobacterial virulence, antibiotic resistance and tolerance. This provided a comprehensive list of methylated genes in drug-resistant clinical isolates and the basis for further functional elucidation.


Subject(s)
Antitubercular Agents/pharmacology , Epigenome/genetics , Extensively Drug-Resistant Tuberculosis/genetics , Mycobacterium tuberculosis/genetics , Sequence Analysis, DNA/methods , Single Molecule Imaging/methods , Antitubercular Agents/therapeutic use , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Epigenome/drug effects , Extensively Drug-Resistant Tuberculosis/drug therapy , Humans , Mycobacterium tuberculosis/drug effects , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/genetics
18.
J Agric Food Chem ; 68(52): 15449-15459, 2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33320666

ABSTRACT

DNA methylation is an epigenetic event that plays critical roles in the pathogenesis, progression, and treatment of human diseases. In this study, we investigated the epigenetic mechanisms for Astragalus polysaccharide (APS)-improved osteoporosis in a rat model. The results showed that APS significantly changed the DNA methylome in colonic epithelia with great efficiency. Gene set enrichment analysis (GSEA) based on differentially methylated sites (DMSs) revealed that APS caused promoter DNA methylation changes of genes associated with calcium homeostasis, osteoclast/osteoblast balance, Wnt signaling, and hormone-related processes. Further analysis showed high consistency of APS-induced gene methylomic changes in colonic epithelia and its effects on diabetes, virus infection, and wound healing, which had been reported already. Moreover, we suggested new functions and the involved mechanisms of APS in heart disease, neurological disorder, reproductive problem, and olfactory dysfunction. In this study, we offered epigenetic mechanisms for APS-improved osteoporosis. More importantly, we proposed and proved a reliable method to explore the beneficial effects of bioactive polysaccharides by studying DNA methylation changes at nonfocal sites. We firmly believed the promising prospects of this method for its great efficiency, rapidness, and economy in exploring possible beneficial or therapeutic effects of functional macromolecules with one single experiment.


Subject(s)
Astragalus Plant/chemistry , Drugs, Chinese Herbal/administration & dosage , Osteoporosis/drug therapy , Polysaccharides/administration & dosage , Animals , Calcium/metabolism , DNA Methylation/drug effects , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Epigenesis, Genetic/drug effects , Epigenome/drug effects , Female , Humans , Osteoporosis/genetics , Osteoporosis/metabolism , Pilot Projects , Polysaccharides/chemistry , Rats , Rats, Sprague-Dawley
19.
PLoS Med ; 17(11): e1003229, 2020 11.
Article in English | MEDLINE | ID: mdl-33151971

ABSTRACT

BACKGROUND: Higher maternal plasma glucose (PG) concentrations, even below gestational diabetes mellitus (GDM) thresholds, are associated with adverse offspring outcomes, with DNA methylation proposed as a mediating mechanism. Here, we examined the relationships between maternal dysglycaemia at 24 to 28 weeks' gestation and DNA methylation in neonates and whether a dietary and physical activity intervention in pregnant women with obesity modified the methylation signatures associated with maternal dysglycaemia. METHODS AND FINDINGS: We investigated 557 women, recruited between 2009 and 2014 from the UK Pregnancies Better Eating and Activity Trial (UPBEAT), a randomised controlled trial (RCT), of a lifestyle intervention (low glycaemic index (GI) diet plus physical activity) in pregnant women with obesity (294 contol, 263 intervention). Between 27 and 28 weeks of pregnancy, participants had an oral glucose (75 g) tolerance test (OGTT), and GDM diagnosis was based on diagnostic criteria recommended by the International Association of Diabetes and Pregnancy Study Groups (IADPSG), with 159 women having a diagnosis of GDM. Cord blood DNA samples from the infants were interrogated for genome-wide DNA methylation levels using the Infinium Human MethylationEPIC BeadChip array. Robust regression was carried out, adjusting for maternal age, smoking, parity, ethnicity, neonate sex, and predicted cell-type composition. Maternal GDM, fasting glucose, 1-h, and 2-h glucose concentrations following an OGTT were associated with 242, 1, 592, and 17 differentially methylated cytosine-phosphate-guanine (dmCpG) sites (false discovery rate (FDR) ≤ 0.05), respectively, in the infant's cord blood DNA. The most significantly GDM-associated CpG was cg03566881 located within the leucine-rich repeat-containing G-protein coupled receptor 6 (LGR6) (FDR = 0.0002). Moreover, we show that the GDM and 1-h glucose-associated methylation signatures in the cord blood of the infant appeared to be attenuated by the dietary and physical activity intervention during pregnancy; in the intervention arm, there were no GDM and two 1-h glucose-associated dmCpGs, whereas in the standard care arm, there were 41 GDM and 160 1-h glucose-associated dmCpGs. A total of 87% of the GDM and 77% of the 1-h glucose-associated dmCpGs had smaller effect sizes in the intervention compared to the standard care arm; the adjusted r2 for the association of LGR6 cg03566881 with GDM was 0.317 (95% confidence interval (CI) 0.012, 0.022) in the standard care and 0.240 (95% CI 0.001, 0.015) in the intervention arm. Limitations included measurement of DNA methylation in cord blood, where the functional significance of such changes are unclear, and because of the strong collinearity between treatment modality and severity of hyperglycaemia, we cannot exclude that treatment-related differences are potential confounders. CONCLUSIONS: Maternal dysglycaemia was associated with significant changes in the epigenome of the infants. Moreover, we found that the epigenetic impact of a dysglycaemic prenatal maternal environment appeared to be modified by a lifestyle intervention in pregnancy. Further research will be needed to investigate possible medical implications of the findings. TRIAL REGISTRATION: ISRCTN89971375.


Subject(s)
Diabetes, Gestational/epidemiology , Diet , Epigenome , Life Style , Adult , Diet/adverse effects , Epigenome/drug effects , Epigenome/physiology , Exercise/physiology , Female , Gestational Age , Humans , Infant , Infant, Newborn , Obesity/epidemiology , Obesity/therapy , Pregnancy
20.
Adv Exp Med Biol ; 1258: 55-75, 2020.
Article in English | MEDLINE | ID: mdl-32767234

ABSTRACT

Epigenetic deregulation is an emerging hallmark of cancer that enables tumor cells to escape surveillance by tumor suppressors and ultimately progress. The structure of the epigenome consists of covalent modifications of chromatin components, including acetylation by histone acetyltransferases (HATs) and deacetylation by histone deacetylases (HDACs). Targeting these enzymes with inhibitors to restore epigenetic homeostasis has been explored for many cancers. Osteosarcoma, an aggressive bone malignancy that primarily affects children and young adults, is notable for widespread genetic and epigenetic instability. This may explain why therapy directed at unique molecular pathways has failed to substantially improve outcomes in osteosarcoma over the past four decades. In this review, we discuss the potential of targeting the cancer epigenome, with a focus on histone deacetylase inhibitors (HDACi) for osteosarcoma. We additionally highlight the safety and tolerance of HDACi, combination chemotherapy with HDACi, and the ongoing challenges in the development of these agents.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Epigenome/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Acetylation/drug effects , Bone Neoplasms/enzymology , Histone Deacetylases/metabolism , Humans , Osteosarcoma/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...