ijms-logo

Journal Browser

Journal Browser

Epigenetic Mechanisms and Human Pathology

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (31 March 2021) | Viewed by 53799

Special Issue Editors


E-Mail Website
Guest Editor
Department of Genetics, Wroclaw Medical University, 50-368 Wroclaw, Poland
Interests: DNA methylation; epigenetics; protein tyrosine phosphatases; tyrosine receptor; dephosphorylation
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Co-Guest Editor
Department of Genetics, Wroclaw Medical University, 50-368 Wroclaw, Poland
Interests: epigenetics; bioinformatics; cancer microenvironment; cancer immunology; tumor subtyping
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Growing evidence indicates that addressing the direct effects of genetic and environmental factors might be insufficient to understand the mechanisms underlying the complexity of human pathologies. The recognition of epigenetic mechanisms has largely improved our understanding of human diseases. The term "epigenetics" refers to several molecular mechanisms that impact DNA expression without altering its sequence. Epigenetic regulation of gene expression includes three main mechanisms, i.e., DNA methylation, histone modifications, and non-protein-coding RNAs (npcRNAs), which play a fundamental role in all molecular processes in living organisms. Consequently, any alterations in these mechanisms may result in a whole range of human pathologies including pediatric, psychiatric, neurologic, endocrine disorders, as well as cancer and non-communicable chronic adulthood diseases. The mostly reversible nature of epigenetic alterations provides grounds for developing novel treatment targets and personalized therapies.

This Special Issue of the International Journal of Molecular Sciences entitled: “Epigenetic Mechanisms and Human Pathology” shall serve as a forum to present state-of-the-art and critical reviews, as well as original papers discussing the application of epigenetic approaches to the etiology, diagnostics, and therapy of a variety of diseases.

Contributions from leading international investigators will guarantee a broad and comprehensive analysis of potential epigenetic mechanisms as well as related clinical problems.

Note: You are warmly welcomed to contribute your outstanding research outcomes to our second volume “Epigenetic Mechanisms and Human Pathology 2.0”.

Prof. Dr. Maria M. Sasiadek
Dr. Pawel Karpinski
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Epigenetics
  • human pathology
  • cancer
  • DNA methylation
  • npcRNA, histone modification
  • personalized therapy

Published Papers (13 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

15 pages, 6742 KiB  
Article
Poly(ADP-Ribose) Polymerase 1 Promotes Inflammation and Fibrosis in a Mouse Model of Chronic Pancreatitis
by Tarek El-Hamoly, Zoltán Hajnády, Máté Nagy-Pénzes, Edina Bakondi, Zsolt Regdon, Máté A. Demény, Katalin Kovács, Csaba Hegedűs, Sahar S. Abd El-Rahman, Éva Szabó, József Maléth, Péter Hegyi and László Virág
Int. J. Mol. Sci. 2021, 22(7), 3593; https://doi.org/10.3390/ijms22073593 - 30 Mar 2021
Cited by 10 | Viewed by 3309
Abstract
Chronic pancreatitis (CP) is an inflammatory disease of the pancreas characterized by ductal obstructions, tissue fibrosis, atrophy and exocrine and endocrine pancreatic insufficiency. However, our understanding is very limited concerning the disease’s progression from a single acute inflammation, via recurrent acute pancreatitis (AP) [...] Read more.
Chronic pancreatitis (CP) is an inflammatory disease of the pancreas characterized by ductal obstructions, tissue fibrosis, atrophy and exocrine and endocrine pancreatic insufficiency. However, our understanding is very limited concerning the disease’s progression from a single acute inflammation, via recurrent acute pancreatitis (AP) and early CP, to the late stage CP. Poly(ADP-ribose) polymerase 1 (PARP1) is a DNA damage sensor enzyme activated mostly by oxidative DNA damage. As a co-activator of inflammatory transcription factors, PARP1 is a central mediator of the inflammatory response and it has also been implicated in acute pancreatitis. Here, we set out to investigate whether PARP1 contributed to the pathogenesis of CP. We found that the clinically used PARP inhibitor olaparib (OLA) had protective effects in a murine model of CP induced by multiple cerulein injections. OLA reduced pancreas atrophy and expression of the inflammatory mediators TNFα and interleukin-6 (IL-6), both in the pancreas and in the lungs. Moreover, there was significantly less fibrosis (Masson’s trichrome staining) in the pancreatic sections of OLA-treated mice compared to the cerulein-only group. mRNA expression of the fibrosis markers TGFβ, smooth muscle actin (SMA), and collagen-1 were markedly reduced by OLA. CP was also induced in PARP1 knockout (KO) mice and their wild-type (WT) counterparts. Inflammation and fibrosis markers showed lower expression in the KO compared to the WT mice. Moreover, reduced granulocyte infiltration (tissue myeloperoxidase activity) and a lower elevation of serum amylase and lipase activity could also be detected in the KO mice. Furthermore, primary acinar cells isolated from KO mice were also protected from cerulein-induced toxicity compared to WT cells. In summary, our data suggest that PARP inhibitors may be promising candidates for repurposing to treat not only acute but chronic pancreatitis as well. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

17 pages, 1901 KiB  
Article
VTRNA2-1: Genetic Variation, Heritable Methylation and Disease Association
by Pierre-Antoine Dugué, Chenglong Yu, Timothy McKay, Ee Ming Wong, Jihoon Eric Joo, Helen Tsimiklis, Fleur Hammet, Maryam Mahmoodi, Derrick Theys, kConFab, John L. Hopper, Graham G. Giles, Roger L. Milne, Jason A. Steen, James G. Dowty, Tu Nguyen-Dumont and Melissa C. Southey
Int. J. Mol. Sci. 2021, 22(5), 2535; https://doi.org/10.3390/ijms22052535 - 3 Mar 2021
Cited by 13 | Viewed by 2854
Abstract
VTRNA2-1 is a metastable epiallele with accumulating evidence that methylation at this region is heritable, modifiable and associated with disease including risk and progression of cancer. This study investigated the influence of genetic variation and other factors such as age and adult lifestyle [...] Read more.
VTRNA2-1 is a metastable epiallele with accumulating evidence that methylation at this region is heritable, modifiable and associated with disease including risk and progression of cancer. This study investigated the influence of genetic variation and other factors such as age and adult lifestyle on blood DNA methylation in this region. We first sequenced the VTRNA2-1 gene region in multiple-case breast cancer families in which VTRNA2-1 methylation was identified as heritable and associated with breast cancer risk. Methylation quantitative trait loci (mQTL) were investigated using a prospective cohort study (4500 participants with genotyping and methylation data). The cis-mQTL analysis (334 variants ± 50 kb of the most heritable CpG site) identified 43 variants associated with VTRNA2-1 methylation (p < 1.5 × 10−4); however, these explained little of the methylation variation (R2 < 0.5% for each of these variants). No genetic variants elsewhere in the genome were found to strongly influence VTRNA2-1 methylation. SNP-based heritability estimates were consistent with the mQTL findings (h2 = 0, 95%CI: −0.14 to 0.14). We found no evidence that age, sex, country of birth, smoking, body mass index, alcohol consumption or diet influenced blood DNA methylation at VTRNA2-1. Genetic factors and adult lifestyle play a minimal role in explaining methylation variability at the heritable VTRNA2-1 cluster. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

17 pages, 4025 KiB  
Article
Effect of Chronic Western Diets on Non-Alcoholic Fatty Liver of Male Mice Modifying the PPAR-γ Pathway via miR-27b-5p Regulation
by Jian Zhang, Catherine A Powell, Matthew K Kay, Ravi Sonkar, Sunitha Meruvu and Mahua Choudhury
Int. J. Mol. Sci. 2021, 22(4), 1822; https://doi.org/10.3390/ijms22041822 - 12 Feb 2021
Cited by 17 | Viewed by 3268
Abstract
Western diets contribute to metabolic diseases. However, the effects of various diets and epigenetic mechanisms are mostly unknown. Here, six week-old C57BL/6J male and female mice were fed with a low-fat diet (LFD), high-fat diet (HFD), and high-fat high-fructose diet (HFD-HF) for 20 [...] Read more.
Western diets contribute to metabolic diseases. However, the effects of various diets and epigenetic mechanisms are mostly unknown. Here, six week-old C57BL/6J male and female mice were fed with a low-fat diet (LFD), high-fat diet (HFD), and high-fat high-fructose diet (HFD-HF) for 20 weeks. We determined that HFD-HF or HFD mice experienced significant metabolic dysregulation compared to the LFD. HFD-HF and HFD-fed male mice showed significantly increased body weight, liver size, and fasting glucose levels with downregulated PPARγ, SCD1, and FAS protein expression. In contrast, female mice were less affected by HFD and HFD-HF. As miR-27b contains a seed sequence in PPARγ, it was discovered that these changes are accompanied by male-specific upregulation of miR-27b-5p, which is even more pronounced in the HFD-HF group (p < 0.01 vs. LFD) compared to the HFD group (p < 0.05 vs. LFD). Other miR-27 subtypes were increased but not significantly. HFD-HF showed insignificant changes in fibrosis markers when compared to LFD. Interestingly, fat ballooning in hepatocytes was increased in HFD-fed mice compared to HFD-HF fed mice, however, the HFD-HF liver showed an increase in the number of small cells. Here, we concluded that chronic Western diet-composition administered for 20 weeks may surpass the non-alcoholic fatty liver (NAFL) stage but may be at an intermediate stage between fatty liver and fibrosis via miR-27b-5p-induced PPARγ downregulation. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

14 pages, 1682 KiB  
Article
Reprogramming and Differentiation of Cutaneous Squamous Cell Carcinoma Cells in Recessive Dystrophic Epidermolysis Bullosa
by Avina Rami, Łukasz Łaczmański, Jagoda Jacków-Nowicka and Joanna Jacków
Int. J. Mol. Sci. 2021, 22(1), 245; https://doi.org/10.3390/ijms22010245 - 29 Dec 2020
Cited by 5 | Viewed by 2647
Abstract
The early onset and rapid progression of cutaneous squamous cell carcinoma (cSCC) leads to high mortality rates in individuals with recessive dystrophic epidermolysis bullosa (RDEB). Currently, the molecular mechanisms underlying cSCC development in RDEB are not well understood and there are limited therapeutic [...] Read more.
The early onset and rapid progression of cutaneous squamous cell carcinoma (cSCC) leads to high mortality rates in individuals with recessive dystrophic epidermolysis bullosa (RDEB). Currently, the molecular mechanisms underlying cSCC development in RDEB are not well understood and there are limited therapeutic options. RDEB-cSCC arises through the accumulation of genetic mutations; however, previous work analyzing gene expression profiles have not been able to explain its aggressive nature. Therefore, we generated a model to study RDEB-cSCC development using cellular reprograming and re-differentiation technology. We compared RDEB-cSCC to cSCC that were first reprogrammed into induced pluripotent stem cells (RDEB-cSCC-iPSC) and then differentiated back to keratinocytes (RDEB-cSCC-iKC). The RDEB-cSCC-iKC cell population had reduced proliferative capacities in vitro and in vivo, suggesting that reprogramming and re-differentiation leads to functional changes. Finally, we performed RNA-seq analysis for RDEB-cSCC, RDEB-cSCC-iPSC, and RDEB-cSCC-iKC and identified different gene expression signatures between these cell populations. Taken together, this cell culture model offers a valuable tool to study cSCC and provides a novel way to identify potential therapeutic targets for RDEB-cSCC. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

13 pages, 1558 KiB  
Article
Comparison of Bisulfite Pyrosequencing and Methylation-Specific qPCR for Methylation Assessment
by Loretta De Chiara, Virginia Leiro-Fernandez, Mar Rodríguez-Girondo, Diana Valverde, María Isabel Botana-Rial and Alberto Fernández-Villar
Int. J. Mol. Sci. 2020, 21(23), 9242; https://doi.org/10.3390/ijms21239242 - 3 Dec 2020
Cited by 11 | Viewed by 2579
Abstract
Different methodological approaches are available to assess DNA methylation biomarkers. In this study, we evaluated two sodium bisulfite conversion-dependent methods, namely pyrosequencing and methylation-specific qPCR (MS-qPCR), with the aim of measuring the closeness of agreement of methylation values between these two methods and [...] Read more.
Different methodological approaches are available to assess DNA methylation biomarkers. In this study, we evaluated two sodium bisulfite conversion-dependent methods, namely pyrosequencing and methylation-specific qPCR (MS-qPCR), with the aim of measuring the closeness of agreement of methylation values between these two methods and its effect when setting a cut-off. Methylation of tumor suppressor gene p16/INK4A was evaluated in 80 lung cancer patients from which cytological lymph node samples were obtained. Cluster analyses were used to establish methylated and unmethylated groups for each method. Agreement and concordance between pyrosequencing and MS-qPCR was evaluated with Pearson’s correlation, Bland–Altman, Cohen’s kappa index and ROC curve analyses. Based on these analyses, cut-offs were derived for MS-qPCR. An acceptable correlation (Pearson’s R2 = 0.738) was found between pyrosequencing (PYRmean) and MS-qPCR (NMP; normalized methylation percentage), providing similar clinical results when categorizing data as binary using cluster analysis. Compared to pyrosequencing, MS-qPCR tended to underestimate methylation for values between 0 and 15%, while for methylation >30% overestimation was observed. The estimated cut-off for MS-qPCR data based on cluster analysis, kappa-index agreement and ROC curve analysis were much lower than that derived from pyrosequencing. In conclusion, our results indicate that independently of the approach used for estimating the cut-off, the methylation percentage obtained through MS-qPCR is lower than that calculated for pyrosequencing. These differences in data and therefore in the cut-off should be examined when using methylation biomarkers in the clinical practice. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

18 pages, 984 KiB  
Article
DNA Methylation at Birth Predicts Intellectual Functioning and Autism Features in Children with Fragile X Syndrome
by Claudine M Kraan, Emma K Baker, Marta Arpone, Minh Bui, Ling Ling, Dinusha Gamage, Lesley Bretherton, Carolyn Rogers, Michael J Field, Tiffany L Wotton, David Francis, Matt F Hunter, Jonathan Cohen, David J Amor and David E Godler
Int. J. Mol. Sci. 2020, 21(20), 7735; https://doi.org/10.3390/ijms21207735 - 19 Oct 2020
Cited by 10 | Viewed by 3402
Abstract
Fragile X syndrome (FXS) is a leading single-gene cause of intellectual disability (ID) with autism features. This study analysed diagnostic and prognostic utility of the Fragile X-Related Epigenetic Element 2 DNA methylation (FREE2m) assessed by Methylation Specific-Quantitative Melt Analysis and the EpiTYPER system, [...] Read more.
Fragile X syndrome (FXS) is a leading single-gene cause of intellectual disability (ID) with autism features. This study analysed diagnostic and prognostic utility of the Fragile X-Related Epigenetic Element 2 DNA methylation (FREE2m) assessed by Methylation Specific-Quantitative Melt Analysis and the EpiTYPER system, in retrospectively retrieved newborn blood spots (NBS) and newly created dried blood spots (DBS) from 65 children with FXS (~2–17 years). A further 168 NBS from infants from the general population were used to establish control reference ranges, in both sexes. FREE2m analysis showed sensitivity and specificity approaching 100%. In FXS males, NBS FREE2m strongly correlated with intellectual functioning and autism features, however associations were not as strong for FXS females. Fragile X mental retardation 1 gene (FMR1) mRNA levels in blood were correlated with FREE2m in both NBS and DBS, for both sexes. In females, DNAm was significantly increased at birth with a decrease in childhood. The findings support the use of FREE2m analysis in newborns for screening, diagnostic and prognostic testing in FXS. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

Review

Jump to: Research

21 pages, 637 KiB  
Review
The Emerging Role of Chromatin Remodeling Complexes in Ovarian Cancer
by Ieva Vaicekauskaitė, Rasa Sabaliauskaitė, Juozas Rimantas Lazutka and Sonata Jarmalaitė
Int. J. Mol. Sci. 2022, 23(22), 13670; https://doi.org/10.3390/ijms232213670 - 8 Nov 2022
Cited by 11 | Viewed by 2922
Abstract
Ovarian cancer (OC) is the fifth leading cause of women’s death from cancers. The high mortality rate is attributed to the late presence of the disease and the lack of modern diagnostic tools, including molecular biomarkers. Moreover, OC is a highly heterogeneous disease, [...] Read more.
Ovarian cancer (OC) is the fifth leading cause of women’s death from cancers. The high mortality rate is attributed to the late presence of the disease and the lack of modern diagnostic tools, including molecular biomarkers. Moreover, OC is a highly heterogeneous disease, which contributes to early treatment failure. Thus, exploring OC molecular mechanisms could significantly enhance our understanding of the disease and provide new treatment options. Chromatin remodeling complexes (CRCs) are ATP-dependent molecular machines responsible for chromatin reorganization and involved in many DNA-related processes, including transcriptional regulation, replication, and reparation. Dysregulation of chromatin remodeling machinery may be related to cancer development and chemoresistance in OC. Some forms of OC and other gynecologic diseases have been associated with mutations in specific CRC genes. Most notably, ARID1A in endometriosis-related OC, SMARCA4, and SMARCB1 in hypercalcemic type small cell ovarian carcinoma (SCCOHT), ACTL6A, CHRAC1, RSF1 amplification in high-grade serous OC. Here we review the available literature on CRCs’ involvement in OC to improve our understanding of its development and investigate CRCs as possible biomarkers and treatment targets for OC. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

15 pages, 357 KiB  
Review
Epigenetic Regulation of Glycosylation in Cancer and Other Diseases
by Rossella Indellicato and Marco Trinchera
Int. J. Mol. Sci. 2021, 22(6), 2980; https://doi.org/10.3390/ijms22062980 - 15 Mar 2021
Cited by 11 | Viewed by 3185
Abstract
In the last few decades, the newly emerging field of epigenetic regulation of glycosylation acquired more importance because it is unraveling physiological and pathological mechanisms related to glycan functions. Glycosylation is a complex process in which proteins and lipids are modified by the [...] Read more.
In the last few decades, the newly emerging field of epigenetic regulation of glycosylation acquired more importance because it is unraveling physiological and pathological mechanisms related to glycan functions. Glycosylation is a complex process in which proteins and lipids are modified by the attachment of monosaccharides. The main actors in this kind of modification are the glycoenzymes, which are translated from glycosylation-related genes (or glycogenes). The expression of glycogenes is regulated by transcription factors and epigenetic mechanisms (mainly DNA methylation, histone acetylation and noncoding RNAs). This review focuses only on these last ones, in relation to cancer and other diseases, such as inflammatory bowel disease and IgA1 nephropathy. In fact, it is clear that a deeper knowledge in the fine-tuning of glycogenes is essential for acquiring new insights in the glycan field, especially if this could be useful for finding novel and personalized therapeutics. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
17 pages, 716 KiB  
Review
DNA Methylation in T-Cell Acute Lymphoblastic Leukemia: In Search for Clinical and Biological Meaning
by Natalia Maćkowska, Monika Drobna-Śledzińska, Michał Witt and Małgorzata Dawidowska
Int. J. Mol. Sci. 2021, 22(3), 1388; https://doi.org/10.3390/ijms22031388 - 30 Jan 2021
Cited by 10 | Viewed by 3659
Abstract
Distinct DNA methylation signatures, related to different prognosis, have been observed across many cancers, including T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological neoplasm. By global methylation analysis, two major phenotypes might be observed in T-ALL: hypermethylation related to better outcome and hypomethylation, [...] Read more.
Distinct DNA methylation signatures, related to different prognosis, have been observed across many cancers, including T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological neoplasm. By global methylation analysis, two major phenotypes might be observed in T-ALL: hypermethylation related to better outcome and hypomethylation, which is a candidate marker of poor prognosis. Moreover, DNA methylation holds more than a clinical meaning. It reflects the replicative history of leukemic cells and most likely different mechanisms underlying leukemia development in these T-ALL subtypes. The elucidation of the mechanisms and aberrations specific to (epi-)genomic subtypes might pave the way towards predictive diagnostics and precision medicine in T-ALL. We present the current state of knowledge on the role of DNA methylation in T-ALL. We describe the involvement of DNA methylation in normal hematopoiesis and T-cell development, focusing on epigenetic aberrations contributing to this leukemia. We further review the research investigating distinct methylation phenotypes in T-ALL, related to different outcomes, pointing to the most recent research aimed to unravel the biological mechanisms behind differential methylation. We highlight how technological advancements facilitated broadening the perspective of the investigation into DNA methylation and how this has changed our understanding of the roles of this epigenetic modification in T-ALL. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

18 pages, 374 KiB  
Review
The Functions of the Demethylase JMJD3 in Cancer
by Anna Sanchez, Fatma Zohra Houfaf Khoufaf, Mouhamed Idrissou, Frédérique Penault-Llorca, Yves-Jean Bignon, Laurent Guy and Dominique Bernard-Gallon
Int. J. Mol. Sci. 2021, 22(2), 968; https://doi.org/10.3390/ijms22020968 - 19 Jan 2021
Cited by 15 | Viewed by 4182
Abstract
Cancer is a major cause of death worldwide. Epigenetic changes in response to external (diet, sports activities, etc.) and internal events are increasingly implicated in tumor initiation and progression. In this review, we focused on post-translational changes in histones and, more particularly, the [...] Read more.
Cancer is a major cause of death worldwide. Epigenetic changes in response to external (diet, sports activities, etc.) and internal events are increasingly implicated in tumor initiation and progression. In this review, we focused on post-translational changes in histones and, more particularly, the tri methylation of lysine from histone 3 (H3K27me3) mark, a repressive epigenetic mark often under- or overexpressed in a wide range of cancers. Two actors regulate H3K27 methylation: Jumonji Domain-Containing Protein 3 demethylase (JMJD3) and Enhancer of zeste homolog 2 (EZH2) methyltransferase. A number of studies have highlighted the deregulation of these actors, which is why this scientific review will focus on the role of JMJD3 and, consequently, H3K27me3 in cancer development. Data on JMJD3’s involvement in cancer are classified by cancer type: nervous system, prostate, blood, colorectal, breast, lung, liver, ovarian, and gastric cancers. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
37 pages, 2242 KiB  
Review
Aberrant Activity of Histone–Lysine N-Methyltransferase 2 (KMT2) Complexes in Oncogenesis
by Elzbieta Poreba, Krzysztof Lesniewicz and Julia Durzynska
Int. J. Mol. Sci. 2020, 21(24), 9340; https://doi.org/10.3390/ijms21249340 - 8 Dec 2020
Cited by 16 | Viewed by 6413
Abstract
KMT2 (histone-lysine N-methyltransferase subclass 2) complexes methylate lysine 4 on the histone H3 tail at gene promoters and gene enhancers and, thus, control the process of gene transcription. These complexes not only play an essential role in normal development but have also been [...] Read more.
KMT2 (histone-lysine N-methyltransferase subclass 2) complexes methylate lysine 4 on the histone H3 tail at gene promoters and gene enhancers and, thus, control the process of gene transcription. These complexes not only play an essential role in normal development but have also been described as involved in the aberrant growth of tissues. KMT2 mutations resulting from the rearrangements of the KMT2A (MLL1) gene at 11q23 are associated with pediatric mixed-lineage leukemias, and recent studies demonstrate that KMT2 genes are frequently mutated in many types of human cancers. Moreover, other components of the KMT2 complexes have been reported to contribute to oncogenesis. This review summarizes the recent advances in our knowledge of the role of KMT2 complexes in cell transformation. In addition, it discusses the therapeutic targeting of different components of the KMT2 complexes. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

15 pages, 1004 KiB  
Review
EZH2 as a Potential Target for NAFLD Therapy
by Hyun Jung Lim and Mirang Kim
Int. J. Mol. Sci. 2020, 21(22), 8617; https://doi.org/10.3390/ijms21228617 - 16 Nov 2020
Cited by 17 | Viewed by 4519
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex disease that is affected by genetic predisposition and epigenetic modification. Deregulation of epigenetic pathways is now recognized as a frequent event in NAFLD, and understanding the mechanistic roles of these epigenetic factors may lead to [...] Read more.
Non-alcoholic fatty liver disease (NAFLD) is a complex disease that is affected by genetic predisposition and epigenetic modification. Deregulation of epigenetic pathways is now recognized as a frequent event in NAFLD, and understanding the mechanistic roles of these epigenetic factors may lead to new strategies for NAFLD treatment. Enhancer of zeste homolog 2 (EZH2) catalyzes methylation on Lys 27 of histone H3, which leads to chromatin compaction and gene silencing. EZH2 regulates embryonic development and cell lineage determination and is related to many human diseases. Recent studies show that EZH2 has critical roles in liver development, homeostasis, and regeneration. Moreover, aberrant activation of EZH2 promotes NAFLD progression. Several EZH2 inhibitors have been developed and studied both in vitro and in clinical trials. In this review, we summarize our current understanding of the role of EZH2 in NAFLD and highlight its potential as a novel therapeutic target for NAFLD treatment. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Graphical abstract

18 pages, 1231 KiB  
Review
From 1957 to Nowadays: A Brief History of Epigenetics
by Paul Peixoto, Pierre-François Cartron, Aurélien A. Serandour and Eric Hervouet
Int. J. Mol. Sci. 2020, 21(20), 7571; https://doi.org/10.3390/ijms21207571 - 14 Oct 2020
Cited by 69 | Viewed by 8991
Abstract
Due to the spectacular number of studies focusing on epigenetics in the last few decades, and particularly for the last few years, the availability of a chronology of epigenetics appears essential. Indeed, our review places epigenetic events and the identification of the main [...] Read more.
Due to the spectacular number of studies focusing on epigenetics in the last few decades, and particularly for the last few years, the availability of a chronology of epigenetics appears essential. Indeed, our review places epigenetic events and the identification of the main epigenetic writers, readers and erasers on a historic scale. This review helps to understand the increasing knowledge in molecular and cellular biology, the development of new biochemical techniques and advances in epigenetics and, more importantly, the roles played by epigenetics in many physiological and pathological situations. Full article
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology)
Show Figures

Figure 1

Back to TopTop