Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (79)

Search Parameters:
Keywords = brain epigenomics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
47 pages, 2976 KiB  
Review
Epigenetic Alterations in Glioblastoma Multiforme as Novel Therapeutic Targets: A Scoping Review
by Marco Meleiro and Rui Henrique
Int. J. Mol. Sci. 2025, 26(12), 5634; https://doi.org/10.3390/ijms26125634 - 12 Jun 2025
Viewed by 1306
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor with a dismal prognosis despite advances in multimodal treatment. Conventional therapies fail to achieve durable responses due to GBM’s molecular heterogeneity and capacity to evade therapeutic pressures. Epigenetic alterations have emerged as critical [...] Read more.
Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor with a dismal prognosis despite advances in multimodal treatment. Conventional therapies fail to achieve durable responses due to GBM’s molecular heterogeneity and capacity to evade therapeutic pressures. Epigenetic alterations have emerged as critical contributors to GBM pathobiology, including aberrant DNA methylation, histone modifications, and non-coding RNA (ncRNA) dysregulation. These mechanisms drive oncogenesis, therapy resistance, and immune evasion. This scoping review evaluates the current state of knowledge on epigenetic modifications in GBM, synthesizing findings from original articles and preclinical and clinical trials published over the last decade. Particular attention is given to MGMT promoter hypermethylation status as a biomarker for temozolomide (TMZ) sensitivity, histone deacetylation and methylation as modulators of chromatin structure, and microRNAs as regulators of pathways such as apoptosis and angiogenesis. Therapeutically, epigenetic drugs, like DNA methyltransferase inhibitors (DNMTis) and histone deacetylase inhibitors (HDACis), appear as promising approaches in preclinical models and early trials. Emerging RNA-based therapies targeting dysregulated ncRNAs represent a novel approach to reprogram the tumor epigenome. Combination therapies, pairing epigenetic agents with immune checkpoint inhibitors or chemotherapy, are explored for their potential to enhance treatment response. Despite these advancements, challenges such as tumor heterogeneity, the blood–brain barrier (BBB), and off-target effects remain significant. Future directions emphasize integrative omics approaches to identify patient-specific targets and refine therapies. This article thus highlights the potential of epigenetics in reshaping GBM treatment paradigms. Full article
(This article belongs to the Special Issue Glioblastoma: Molecular Pathogenesis and Treatment)
Show Figures

Graphical abstract

25 pages, 1508 KiB  
Review
Modulating Cognition-Linked Histone Acetyltransferases (HATs) as a Therapeutic Strategy for Neurodegenerative Diseases: Recent Advances and Future Trends
by Huong Anh Mai, Christina M. Thomas, Gu Gu Nge and Felice Elefant
Cells 2025, 14(12), 873; https://doi.org/10.3390/cells14120873 - 10 Jun 2025
Viewed by 840
Abstract
Recent investigations into the neuroepigenome of the brain are providing unparalleled understanding into the impact of post-translational modifications (PTMs) of histones in regulating dynamic gene expression patterns required for adult brain cognitive function and plasticity. Histone acetylation is one of the most well-characterized [...] Read more.
Recent investigations into the neuroepigenome of the brain are providing unparalleled understanding into the impact of post-translational modifications (PTMs) of histones in regulating dynamic gene expression patterns required for adult brain cognitive function and plasticity. Histone acetylation is one of the most well-characterized PTMs shown to be required for neuronal function and cognition. Histone acetylation initiates neural circuitry plasticity via chromatin control, enabling neurons to respond to external environmental stimuli and adapt their transcriptional responses accordingly. While interplay between histone acetylation and deacetylation is critical for these functions, dysregulation during the aging process can lead to significant alterations in the neuroepigenetic landscape. These alterations contribute to impaired cognitive functions, neuronal cell death, and brain atrophy, all hallmarks of age-related neurodegenerative disease. Significantly, while age-related generation of DNA mutations remains irreversible, most neuroepigenetic PTMs are reversible. Thus, manipulation of the neural epigenome is proving to be an effective therapeutic strategy for neuroprotection in multiple types of age-related neurodegenerative disorders (NDs) that include Alzheimer’s disease (AD), Parkinson’s disease (PD), Amyotrophic lateral sclerosis (ALS) and Huntington’s disease (HD). Here, we highlight recent progress in research focusing on specific HAT-based neuroepigenetic mechanisms that underlie cognition and pathogenesis that is hallmarked in age-related NDs. We further discuss how these findings have potential to be translated into HAT-mediated cognitive-enhancing therapeutics to treat these debilitating disorders. Full article
(This article belongs to the Special Issue Biological Mechanisms in the Treatment of Neuropsychiatric Diseases)
Show Figures

Figure 1

18 pages, 1666 KiB  
Review
Molecular Insights into Neurological Regression with a Focus on Rett Syndrome—A Narrative Review
by Jatinder Singh and Paramala Santosh
Int. J. Mol. Sci. 2025, 26(11), 5361; https://doi.org/10.3390/ijms26115361 - 3 Jun 2025
Viewed by 658
Abstract
Rett syndrome (RTT) is a multisystem neurological disorder. Pathogenic changes in the MECP2 gene that codes for methyl-CpG-binding protein 2 (MeCP2) in RTT lead to a loss of previously established motor and cognitive skills. Unravelling the mechanisms of neurological regression in RTT is [...] Read more.
Rett syndrome (RTT) is a multisystem neurological disorder. Pathogenic changes in the MECP2 gene that codes for methyl-CpG-binding protein 2 (MeCP2) in RTT lead to a loss of previously established motor and cognitive skills. Unravelling the mechanisms of neurological regression in RTT is complex, due to multiple components of the neural epigenome being affected. Most evidence has primarily focused on deciphering the complexity of transcriptional machinery at the molecular level. Little attention has been paid to how epigenetic changes across the neural epigenome in RTT lead to neurological regression. In this narrative review, we examine how pathogenic changes in MECP2 can disrupt the balance of the RTT neural epigenome and lead to neurological regression. Environmental and genetic factors can disturb the balance of the neural epigenome in RTT, modifying the onset of neurological regression. Methylation changes across the RTT neural epigenome and the consequent genotoxic stress cause neurons to regress into a senescent state. These changes influence the brain as it matures and lead to the emergence of specific symptoms at different developmental periods. Future work could focus on epidrugs or epi-editing approaches that may theoretically help to restore the epigenetic imbalance and thereby minimise the impact of genotoxic stress on the RTT neural epigenome. Full article
Show Figures

Figure 1

53 pages, 1175 KiB  
Review
Revisiting the Pathogenesis of X-Linked Adrenoleukodystrophy
by Pierre Bougnères and Catherine Le Stunff
Genes 2025, 16(5), 590; https://doi.org/10.3390/genes16050590 - 17 May 2025
Cited by 1 | Viewed by 1505
Abstract
Background: X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal [...] Read more.
Background: X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal cord axonopathy starting in middle-aged adults. The accumulation of undegraded VLCFA in glial cell membranes and myelin has long been thought to be the central mechanism of X-ALD. Methods: This review discusses studies in mouse and drosophila models that have modified our views of X-ALD pathogenesis. Results: In the Abcd1 knockout (KO) mouse that mimics the spinal cord disease, the late manifestations of axonopathy are rapidly reversed by ABCD1 gene transfer into spinal cord oligodendrocytes (OLs). In a peroxin-5 KO mouse model, the selective impairment of peroxisomal biogenesis in OLs achieves an almost perfect phenocopy of cerebral ALD. A drosophila knockout model revealed that VLCFA accumulation in glial myelinating cells causes the production of a toxic lipid able to poison axons and activate inflammatory cells. Other mouse models showed the critical role of OLs in providing energy substrates to axons. In addition, studies on microglial changing substates have improved our understanding of neuroinflammation. Conclusions: Animal models supporting a primary role of OLs and axonal pathology and a secondary role of microglia allow us to revisit of X-ALD mechanisms. Beyond ABCD1 mutations, pathogenesis depends on unidentified contributors, such as genetic background, cell-specific epigenomics, potential environmental triggers, and stochasticity of crosstalk between multiple cell types among billions of glial cells and neurons. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

20 pages, 2343 KiB  
Article
Robust Single-Cell RNA-Seq Analysis Using Hyperdimensional Computing: Enhanced Clustering and Classification Methods
by Hossein Mohammadi, Maziyar Baranpouyan, Krishnaprasad Thirunarayan and Lingwei Chen
AI 2025, 6(5), 94; https://doi.org/10.3390/ai6050094 - 1 May 2025
Viewed by 957
Abstract
Background. Single-cell RNA sequencing (scRNA-seq) has transformed genomics by enabling the study of cellular heterogeneity. However, its high dimensionality, noise, and sparsity pose significant challenges for data analysis. Methods. We investigate the use of Hyperdimensional Computing (HDC), a brain-inspired computational framework recognized for [...] Read more.
Background. Single-cell RNA sequencing (scRNA-seq) has transformed genomics by enabling the study of cellular heterogeneity. However, its high dimensionality, noise, and sparsity pose significant challenges for data analysis. Methods. We investigate the use of Hyperdimensional Computing (HDC), a brain-inspired computational framework recognized for its noise robustness and hardware efficiency, to tackle the challenges in scRNA-seq data analysis. We apply HDC to both supervised classification and unsupervised clustering tasks. Results. Our experiments demonstrate that HDC consistently outperforms established methods such as XGBoost, Seurat reference mapping, and scANVI in terms of noise tolerance and scalability. HDC achieves superior accuracy in classification tasks and maintains robust clustering performance across varying noise levels. Conclusions. These results highlight HDC as a promising framework for accurate and efficient single-cell data analysis. Its potential extends to other high-dimensional biological datasets including proteomics, epigenomics, and transcriptomics, with implications for advancing bioinformatics and personalized medicine. Full article
Show Figures

Figure 1

26 pages, 1921 KiB  
Article
Advancing Personalized Medicine in Alzheimer’s Disease: Liquid Biopsy Epigenomics Unveil APOE ε4-Linked Methylation Signatures
by Mónica Macías, Juan José Alba-Linares, Blanca Acha, Idoia Blanco-Luquin, Agustín F. Fernández, Johana Álvarez-Jiménez, Amaya Urdánoz-Casado, Miren Roldan, Maitane Robles, Eneko Cabezon-Arteta, Daniel Alcolea, Javier Sánchez Ruiz de Gordoa, Jon Corroza, Carolina Cabello, María Elena Erro, Ivonne Jericó, Mario F. Fraga and Maite Mendioroz
Int. J. Mol. Sci. 2025, 26(7), 3419; https://doi.org/10.3390/ijms26073419 - 5 Apr 2025
Cited by 2 | Viewed by 1027
Abstract
Recent studies show that patients with Alzheimer’s disease (AD) harbor specific methylation marks in the brain that, if accessible, could be used as epigenetic biomarkers. Liquid biopsy enables the study of circulating cell-free DNA (cfDNA) fragments originated from dead cells, including neurons affected [...] Read more.
Recent studies show that patients with Alzheimer’s disease (AD) harbor specific methylation marks in the brain that, if accessible, could be used as epigenetic biomarkers. Liquid biopsy enables the study of circulating cell-free DNA (cfDNA) fragments originated from dead cells, including neurons affected by neurodegenerative processes. Here, we isolated and epigenetically characterized plasma cfDNA from 35 patients with AD and 35 cognitively healthy controls by using the Infinium® MethylationEPIC BeadChip array. Bioinformatics analysis was performed to identify differential methylation positions (DMPs) and regions (DMRs), including APOE ε4 genotype stratified analysis. Plasma pTau181 (Simoa) and cerebrospinal fluid (CSF) core biomarkers (Fujirebio) were also measured and correlated with differential methylation marks. Validation was performed with bisulfite pyrosequencing and bisulfite cloning sequencing. Epigenome-wide cfDNA analysis identified 102 DMPs associated with AD status. Most DMPs correlated with clinical cognitive and functional tests including 60% for Mini-Mental State Examination (MMSE) and 80% for Global Deterioration Scale (GDS), and with AD blood and CSF biomarkers. In silico functional analysis connected 30 DMPs to neurological processes, identifying key regulators such as SPTBN4 and APOE genes. Several DMRs were annotated to genes previously reported to harbor epigenetic brain changes in AD (HKR1, ZNF154, HOXA5, TRIM40, ATG16L2, ADAMST2) and were linked to APOE ε4 genotypes. Notably, a DMR in the HKR1 gene, previously shown to be hypermethylated in the AD hippocampus, was validated in cfDNA from an orthogonal perspective. These results support the feasibility of studying cfDNA to identify potential epigenetic biomarkers in AD. Thus, liquid biopsy could improve non-invasive AD diagnosis and aid personalized medicine by detecting epigenetic brain markers in blood. Full article
Show Figures

Figure 1

17 pages, 3846 KiB  
Article
The Enhancer–Promoter-Mediated Wnt8a Transcription During Neurite Regrowth of Injured Cortical Neurons
by Shr-Han Weng, Wen-Ling Liao and Linyi Chen
Cells 2025, 14(5), 319; https://doi.org/10.3390/cells14050319 - 20 Feb 2025
Viewed by 624
Abstract
Brain injuries can result from accidents, warfare, sports injuries, or brain diseases. Identifying regeneration-associated genes (RAGs) during epigenome remodeling upon brain injury could have a significant impact on reducing neuronal death and subsequent neurodegeneration for patients with brain injury. We previously identified several [...] Read more.
Brain injuries can result from accidents, warfare, sports injuries, or brain diseases. Identifying regeneration-associated genes (RAGs) during epigenome remodeling upon brain injury could have a significant impact on reducing neuronal death and subsequent neurodegeneration for patients with brain injury. We previously identified several WNT genes as RAGs involved in the neurite regrowth of injured cortical neurons. Among them, the expression of the Wnt8a gene increased most significantly during neurite regrowth, indicating its potential to promote neuronal regeneration. In this study, we investigated the regulatory mechanism of Wnt8a transcription. An algorithm was developed to predict the novel enhancer regions of candidate genes. By combining active enhancer marks, histone H3 lysine 27 acetylation (H3K27ac), and histone H3 lysine 4 mono-methylation (H3K4me1), we identified a candidate enhancer region for Wnt8a located 1.7 Mb upstream and 0.1 Mb downstream of the Wnt8a gene. This region was organized into enhancers (Ens) 1–15. Enhancer RNA expression from the predicted En1–15 regions, DNA topological dynamics, and the activity of predicted enhancers were analyzed to validate the candidate active enhancers. Our findings showed that the En8, 9, 10, 14, and 15 regions expressed higher eRNAs during neurite regrowth. Notably, the En8-2 and En14-2 subregions showed significantly up-regulated H3K4me1 modification during neurite regrowth. Using chromatin conformation capture assays and enhancer–reporter assays, we delineated that the molecular regulation of Wnt8a transcription during neurite regrowth occurs through looped En8-promoter interplay. Full article
(This article belongs to the Section Cells of the Nervous System)
Show Figures

Figure 1

14 pages, 1197 KiB  
Review
Maternal Gut Microbiome-Mediated Epigenetic Modifications in Cognitive Development and Impairments: A New Frontier for Therapeutic Innovation
by Shabnam Nohesara, Hamid Mostafavi Abdolmaleky, Faith Dickerson, Adrián A. Pinto-Tomás, Dilip V. Jeste and Sam Thiagalingam
Nutrients 2024, 16(24), 4355; https://doi.org/10.3390/nu16244355 - 17 Dec 2024
Cited by 2 | Viewed by 1626
Abstract
Cognitive impairment in various mental illnesses, particularly neuropsychiatric disorders, has adverse functional and clinical consequences. While genetic mutations and epigenetic dysregulations of several genes during embryonic and adult periods are linked to cognitive impairment in mental disorders, the composition and diversity of resident [...] Read more.
Cognitive impairment in various mental illnesses, particularly neuropsychiatric disorders, has adverse functional and clinical consequences. While genetic mutations and epigenetic dysregulations of several genes during embryonic and adult periods are linked to cognitive impairment in mental disorders, the composition and diversity of resident bacteria in the gastrointestinal tract—shaped by environmental factors—also influence the brain epigenome, affecting behavior and cognitive functions. Accordingly, many recent studies have provided evidence that human gut microbiota may offer a potential avenue for improving cognitive deficits. In this review, we provide an overview of the relationship between cognitive impairment, alterations in the gut microbiome, and epigenetic alterations during embryonic and adult periods. We examine how various factors beyond genetics—such as lifestyle, age, and maternal diet—impact the composition, diversity, and epigenetic functionality of the gut microbiome, consequently influencing cognitive performance. Additionally, we explore the potential of maternal gut microbiome signatures and epigenetic biomarkers for predicting cognitive impairment risk in older adults. This article also explores the potential roles of nutritional deficiencies in programming cognitive disorders during the perinatal period in offspring, as well as the promise of gut microbiome-targeted therapeutics with epigenetic effects to prevent or alleviate cognitive dysfunctions in infants, middle-aged adults, and older adults. Unsolved challenges of gut microbiome-targeted therapeutics in mitigating cognitive dysfunctions for translation into clinical practice are discussed, lastly. Full article
Show Figures

Figure 1

35 pages, 570 KiB  
Review
Epigenetic Mechanisms in Aging: Extrinsic Factors and Gut Microbiome
by Alejandro Borrego-Ruiz and Juan J. Borrego
Genes 2024, 15(12), 1599; https://doi.org/10.3390/genes15121599 - 14 Dec 2024
Cited by 5 | Viewed by 3374
Abstract
Background/Objectives: Aging is a natural physiological process involving biological and genetic pathways. Growing evidence suggests that alterations in the epigenome during aging result in transcriptional changes, which play a significant role in the onset of age-related diseases, including cancer, cardiovascular disease, diabetes, and [...] Read more.
Background/Objectives: Aging is a natural physiological process involving biological and genetic pathways. Growing evidence suggests that alterations in the epigenome during aging result in transcriptional changes, which play a significant role in the onset of age-related diseases, including cancer, cardiovascular disease, diabetes, and neurodegenerative disorders. For this reason, the epigenetic alterations in aging and age-related diseases have been reviewed, and the major extrinsic factors influencing these epigenetic alterations have been identified. In addition, the role of the gut microbiome and its metabolites as epigenetic modifiers has been addressed. Results: Long-term exposure to extrinsic factors such as air pollution, diet, drug use, environmental chemicals, microbial infections, physical activity, radiation, and stress provoke epigenetic changes in the host through several endocrine and immune pathways, potentially accelerating the aging process. Diverse studies have reported that the gut microbiome plays a critical role in regulating brain cell functions through DNA methylation and histone modifications. The interaction between genes and the gut microbiome serves as a source of adaptive variation, contributing to phenotypic plasticity. However, the molecular mechanisms and signaling pathways driving this process are still not fully understood. Conclusions: Extrinsic factors are potential inducers of epigenetic alterations, which may have important implications for longevity. The gut microbiome serves as an epigenetic effector influencing host gene expression through histone and DNA modifications, while bidirectional interactions with the host and the underexplored roles of microbial metabolites and non-bacterial microorganisms such as fungi and viruses highlight the need for further research. Full article
(This article belongs to the Section Epigenomics)
38 pages, 4137 KiB  
Review
Epigenetic Explorations of Neurological Disorders, the Identification Methods, and Therapeutic Avenues
by Zeba Firdaus and Xiaogang Li
Int. J. Mol. Sci. 2024, 25(21), 11658; https://doi.org/10.3390/ijms252111658 - 30 Oct 2024
Cited by 3 | Viewed by 2937
Abstract
Neurodegenerative disorders are major health concerns globally, especially in aging societies. The exploration of brain epigenomes, which consist of multiple forms of DNA methylation and covalent histone modifications, offers new and unanticipated perspective into the mechanisms of aging and neurodegenerative diseases. Initially, chromatin [...] Read more.
Neurodegenerative disorders are major health concerns globally, especially in aging societies. The exploration of brain epigenomes, which consist of multiple forms of DNA methylation and covalent histone modifications, offers new and unanticipated perspective into the mechanisms of aging and neurodegenerative diseases. Initially, chromatin defects in the brain were thought to be static abnormalities from early development associated with rare genetic syndromes. However, it is now evident that mutations and the dysregulation of the epigenetic machinery extend across a broader spectrum, encompassing adult-onset neurodegenerative diseases. Hence, it is crucial to develop methodologies that can enhance epigenetic research. Several approaches have been created to investigate alterations in epigenetics on a spectrum of scales—ranging from low to high—with a particular focus on detecting DNA methylation and histone modifications. This article explores the burgeoning realm of neuroepigenetics, emphasizing its role in enhancing our mechanistic comprehension of neurodegenerative disorders and elucidating the predominant techniques employed for detecting modifications in the epigenome. Additionally, we ponder the potential influence of these advancements on shaping future therapeutic approaches. Full article
Show Figures

Figure 1

13 pages, 1566 KiB  
Article
Are Methylation Patterns in the KALRN Gene Associated with Cognitive and Depressive Symptoms? Findings from the Moli-sani Cohort
by Miriam Shasa Quiccione, Alfonsina Tirozzi, Giulia Cassioli, Martina Morelli, Simona Costanzo, Antonietta Pepe, Francesca Bracone, Sara Magnacca, Chiara Cerletti, Danilo Licastro, Augusto Di Castelnuovo, Maria Benedetta Donati, Giovanni de Gaetano, Licia Iacoviello and Alessandro Gialluisi
Int. J. Mol. Sci. 2024, 25(19), 10317; https://doi.org/10.3390/ijms251910317 - 25 Sep 2024
Cited by 4 | Viewed by 1187
Abstract
The KALRN gene (encoding kalirin) has been implicated in several neuropsychiatric and neurodegenerative disorders. However, genetic evidence supporting this implication is limited and targeted epigenetic analyses are lacking. Here, we tested associations between epigenetic variation in KALRN and interindividual variation in depressive symptoms [...] Read more.
The KALRN gene (encoding kalirin) has been implicated in several neuropsychiatric and neurodegenerative disorders. However, genetic evidence supporting this implication is limited and targeted epigenetic analyses are lacking. Here, we tested associations between epigenetic variation in KALRN and interindividual variation in depressive symptoms (PHQ9) and cognitive (MoCA) performance, in an Italian population cohort (N = 2409; mean (SD) age: 67 (9) years; 55% women). First, we analyzed the candidate region chr3:124584826–124584886 (hg38), within the KALRN promoter, through pyrosequencing of 1385 samples. Then, we widened the investigated region by analyzing 137 CpGs annotated to the whole gene, rescued from epigenome-wide (Illumina EPIC) data from 1024 independent samples from the same cohort. These were tested through stepwise regression models adjusted for age, sex, circulating leukocytes fractions, education, prevalent health conditions and lifestyles. We observed no statistically significant associations with methylation levels in the three CpGs tested through pyrosequencing, or in the gene-wide association analysis with MoCA score. However, we observed a statistically significant association between PHQ9 and cg13549966 (chr3:124106738; β (Standard Error) = 0.28 (0.08), Bonferroni-corrected p = 0.025), located close to the transcription start site of the gene. This association was driven by a polychoric factor tagging somatic depressive symptoms (β (SE) = 0.127 (0.064), p = 0.048). This evidence underscores the importance of studying epigenetic variation within the KALRN gene and the role that it may play in brain diseases, particularly in atypical depression, which is often characterized by somatic symptoms. Full article
(This article belongs to the Special Issue Depression: From Molecular Basis to Therapy)
Show Figures

Figure 1

25 pages, 1298 KiB  
Review
From the Environment to Molecular Interactions of Nanoplastics: Unraveling the Neurotoxic Impacts and the Implications in Neurodegenerative Processes
by Chiara Urani, Raffaella Barbieri, Susanna Alloisio and Marina Tesauro
Appl. Sci. 2024, 14(16), 7280; https://doi.org/10.3390/app14167280 - 19 Aug 2024
Cited by 5 | Viewed by 2642
Abstract
Nanoplastics (NPs) represent an escalating hazard to both humans and the ecosystem due to their pervasive presence. This review delves into (i) the widespread occurrence of NPs across the different environmental matrices, including food; (ii) routes and estimates for human exposure; (iii) the [...] Read more.
Nanoplastics (NPs) represent an escalating hazard to both humans and the ecosystem due to their pervasive presence. This review delves into (i) the widespread occurrence of NPs across the different environmental matrices, including food; (ii) routes and estimates for human exposure; (iii) the mechanisms of blood–brain barrier (BBB) crossing; and (iv) implications for human health, with a specific focus on molecular features associated with neurotoxicity and neurodegenerative processes. The impact of NPs on the central nervous system, their ability to cross the BBB and the underpinning mechanisms, the potential to initiate neurotoxicity by fostering β-amyloid aggregation, and their interactions with metallo-enzymes (such as superoxide dismutase) are elucidated. The analysis of transcriptomics and epigenomic results, including microRNA dysregulation, unveil how NPs could contribute to neurological disorders. The need for considering overlaps among diverse pathogenetic mechanisms when probing the effects of NPs is discussed. Additional urgent needs are the development of reliable in vitro models for neurotoxicity studies able to mimic the complexity of the nervous system and the exposure of such models to more environmentally relevant NPs. Finally, the development of extremely sensitive detection and analysis methodologies to quantify NPs in environmental and biological matrices is a pressing priority. Full article
Show Figures

Figure 1

32 pages, 5113 KiB  
Review
Pediatric Hemispheric High-Grade Gliomas and H3.3-G34 Mutation: A Review of the Literature on Biological Features and New Therapeutic Strategies
by Marta Bonada, Matilde Pittarello, Emerson De Fazio, Alessandro Gans, Paolo Alimonti, Hasan Slika, Federico Legnani, Francesco Di Meco and Betty Tyler
Genes 2024, 15(8), 1038; https://doi.org/10.3390/genes15081038 - 6 Aug 2024
Cited by 1 | Viewed by 4716
Abstract
Pediatric high-grade glioma (pHGG) encompasses a wide range of gliomas with different genomic, epigenomic, and transcriptomic features. Almost 50% of pHGGs present a mutation in genes coding for histone 3, including the subtype harboring the H3.3-G34 mutation. In this context, histone mutations are [...] Read more.
Pediatric high-grade glioma (pHGG) encompasses a wide range of gliomas with different genomic, epigenomic, and transcriptomic features. Almost 50% of pHGGs present a mutation in genes coding for histone 3, including the subtype harboring the H3.3-G34 mutation. In this context, histone mutations are frequently associated with mutations in TP53 and ATRX, along with PDGFRA and NOTCH2NL amplifications. Moreover, the H3.3-G34 histone mutation induces epigenetic changes in immune-related genes and exerts modulatory functions on the microenvironment. Also, the functionality of the blood–brain barrier (BBB) has an impact on treatment response. The prognosis remains poor with conventional treatments, thus eliciting the investigation of additional and alternative therapies. Promising molecular targets include PDGFRA amplification, BRAF mutation, EGFR amplification, NF1 loss, and IDH mutation. Considering that pHGGs harboring the H3.3-G34R mutation appear to be more susceptible to immunotherapies (ITs), different options have been recently explored, including immune checkpoint inhibitors, antibody mediated IT, and Car-T cells. This review aims to summarize the knowledge concerning cancer biology and cancer-immune cell interaction in this set of pediatric gliomas, with a focus on possible therapeutic options. Full article
(This article belongs to the Special Issue Epigenetic Modifications for Cancer Therapy)
Show Figures

Figure 1

23 pages, 1498 KiB  
Review
Substance-Induced Psychiatric Disorders, Epigenetic and Microbiome Alterations, and Potential for Therapeutic Interventions
by Shabnam Nohesara, Hamid Mostafavi Abdolmaleky and Sam Thiagalingam
Brain Sci. 2024, 14(8), 769; https://doi.org/10.3390/brainsci14080769 - 30 Jul 2024
Cited by 4 | Viewed by 3184
Abstract
Substance use disorders (SUDs) are complex biopsychosocial diseases that cause neurocognitive deficits and neurological impairments by altering the gene expression in reward-related brain areas. Repeated drug use gives rise to alterations in DNA methylation, histone modifications, and the expression of microRNAs in several [...] Read more.
Substance use disorders (SUDs) are complex biopsychosocial diseases that cause neurocognitive deficits and neurological impairments by altering the gene expression in reward-related brain areas. Repeated drug use gives rise to alterations in DNA methylation, histone modifications, and the expression of microRNAs in several brain areas that may be associated with the development of psychotic symptoms. The first section of this review discusses how substance use contributes to the development of psychotic symptoms via epigenetic alterations. Then, we present more evidence about the link between SUDs and brain epigenetic alterations. The next section presents associations between paternal and maternal exposure to substances and epigenetic alterations in the brains of offspring and the role of maternal diet in preventing substance-induced neurological impairments. Then, we introduce potential therapeutic agents/approaches such as methyl-rich diets to modify epigenetic alterations for alleviating psychotic symptoms or depression in SUDs. Next, we discuss how substance use–gut microbiome interactions contribute to the development of neurological impairments through epigenetic alterations and how gut microbiome-derived metabolites may become new therapeutics for normalizing epigenetic aberrations. Finally, we address possible challenges and future perspectives for alleviating psychotic symptoms and depression in patients with SUDs by modulating diets, the epigenome, and gut microbiome. Full article
(This article belongs to the Special Issue New Insights in Psychiatric Disorder Psychopharmacology)
Show Figures

Figure 1

34 pages, 2074 KiB  
Review
Placental Epigenome Impacts Fetal Development: Effects of Maternal Nutrients and Gut Microbiota
by Sanjay Basak, Rahul Mallick, Boga Navya Sree and Asim K. Duttaroy
Nutrients 2024, 16(12), 1860; https://doi.org/10.3390/nu16121860 - 13 Jun 2024
Cited by 8 | Viewed by 6575
Abstract
Evidence is emerging on the role of maternal diet, gut microbiota, and other lifestyle factors in establishing lifelong health and disease, which are determined by transgenerationally inherited epigenetic modifications. Understanding epigenetic mechanisms may help identify novel biomarkers for gestation-related exposure, burden, or disease [...] Read more.
Evidence is emerging on the role of maternal diet, gut microbiota, and other lifestyle factors in establishing lifelong health and disease, which are determined by transgenerationally inherited epigenetic modifications. Understanding epigenetic mechanisms may help identify novel biomarkers for gestation-related exposure, burden, or disease risk. Such biomarkers are essential for developing tools for the early detection of risk factors and exposure levels. It is necessary to establish an exposure threshold due to nutrient deficiencies or other environmental factors that can result in clinically relevant epigenetic alterations that modulate disease risks in the fetus. This narrative review summarizes the latest updates on the roles of maternal nutrients (n-3 fatty acids, polyphenols, vitamins) and gut microbiota on the placental epigenome and its impacts on fetal brain development. This review unravels the potential roles of the functional epigenome for targeted intervention to ensure optimal fetal brain development and its performance in later life. Full article
Show Figures

Figure 1

Back to TopTop