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










Publication year range
1.
Int J Mol Med ; 53(5)2024 05.
Article in English | MEDLINE | ID: mdl-38488030

ABSTRACT

DNA methylation is an epigenetic modification that plays a key role in several cellular processes mediating the fine regulation of gene expression. Aberrant DNA methylation is observed in a wide range of pathologies, including cancer. Since these DNA modifications are transferred to the cell progenies and are stable over the time, the analysis of DNA methylation status has been proposed for diagnostic and prognostic purposes in cancer. Currently, DNA bisulfite conversion is the gold standard method for the high­throughput analysis of DNA methylation alterations. However, bisulfite treatment induces DNA fragmentation affecting its quality for the downstream analyses. In this field, it is mandatory to identify novel methods to overcome the limits of conventional approaches. In the present study, the Methylation­Sensitive Restriction Enzyme­droplet digital PCR (MSRE­ddPCR) assay was developed as a novel sensitive method for the analysis of DNA methylation of short genomic regions, combining the MSRE assay with the high­sensitivity ddPCR and using an exogenous methylation sequence as control. Setup and validation experiments were performed analyzing a methylation hotspot of the Solute Carrier Family 22 Member 17 in DNA samples derived from melanoma cell lines as well as from tissues and serum samples obtained from patients with melanoma and healthy controls. Compared with the standard MSRE approaches, the MSRE­ddPCR assay is more appropriate for the analysis of DNA methylation (methDNA) in samples with low amounts of DNA (up to 0.651 ng) showing a greater sensitivity. These findings suggested the potential clinical application of MSRE­ddPCR paving the way to the analysis of other methDNA hotspots in different tumors.


Subject(s)
DNA Methylation , Melanoma , Sulfites , Humans , DNA Methylation/genetics , Melanoma/diagnosis , Melanoma/genetics , Polymerase Chain Reaction/methods , DNA/genetics
2.
Genes (Basel) ; 15(3)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38540380

ABSTRACT

Initially described as a triad of immunodeficiency, congenital heart defects and hypoparathyroidism, 22q11.2 deletion syndrome (22q11.2DS) now encompasses a great amount of abnormalities involving different systems. Approximately 85% of patients share a 3 Mb 22q11.2 region of hemizygous deletion in which 46 protein-coding genes are included. However, the hemizygosity of the genes of this region cannot fully explain the clinical phenotype and the phenotypic variability observed among patients. Additional mutations in genes located outside the deleted region, leading to "dual diagnosis", have been described in 1% of patients. In some cases, the hemizygosity of the 22q11.2 region unmasks autosomal recessive conditions due to additional mutations on the non-deleted allele. Some of the deleted genes play a crucial role in gene expression regulation pathways, involving the whole genome. Typical miRNA expression patterns have been identified in 22q11.2DS, due to an alteration in miRNA biogenesis, affecting the expression of several target genes. Also, a methylation epi-signature in CpG islands differentiating patients from controls has been defined. Herein, we summarize the evidence on the genetic and epigenetic mechanisms implicated in the pathogenesis of the clinical manifestations of 22q11.2 DS. The review of the literature confirms the hypothesis that the 22q11.2DS phenotype results from a network of interactions between deleted protein-coding genes and altered epigenetic regulation.


Subject(s)
DiGeorge Syndrome , Heart Defects, Congenital , MicroRNAs , Humans , DiGeorge Syndrome/genetics , Epigenesis, Genetic , Phenotype , Heart Defects, Congenital/genetics
3.
Cell Commun Signal ; 22(1): 120, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38347585

ABSTRACT

BACKGROUND: Breast cancer (BC) is the most frequent tumor entity in women worldwide with a high chance of therapeutic response in early- and non-metastatic disease stages. Among all BC subtypes, triple-negative BC (TNBC) is the most challenging cancer subtype lacking effective molecular targets due to the particular enrichment of cancer stem cells (CSCs), frequently leading to a chemoresistant phenotype and metastasis. The Ubiquitin Specific Peptidase 22 (USP22) is a deubiquitinase that has been frequently associated with a CSC-promoting function and intimately implicated in resistance to conventional therapies, tumor relapse, metastasis and overall poor survival in a broad range of cancer entities, including BC. To date, though, the role of USP22 in TNBC has been only superficially addressed. METHODS: The current study utilized the MMTV-cre, Usp22fl/fl transgenic mouse model to study the involvement of USP22 in the stem cell-like properties of the growing mammary tissue. Additionally, we combined high-throughput transcriptomic analyses with publicly available patient transcriptomic data and utilized TNBC culture models to decipher the functional role of USP22 in the CSC characteristics of this disease. RESULTS: Interestingly, we identified that USP22 promotes CSC properties and drug tolerance by supporting the oxidative phosphorylation program, known to be largely responsible for the poor response to conventional therapies in this particularly aggressive BC subtype. CONCLUSIONS: This study suggests a novel tumor-supportive role of USP22 in sustaining cellular respiration to facilitate the drug-tolerant behavior of HER2+-BC and TNBC cells. Therefore, we posit USP22 as a promising therapeutic target to optimize standard therapies and combat the aggressiveness of these malignancies. Video Abstract.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Respiration , Disease Models, Animal , Neoplasm Recurrence, Local , Triple Negative Breast Neoplasms/pathology , Ubiquitin Thiolesterase
4.
Genes (Basel) ; 13(3)2022 03 17.
Article in English | MEDLINE | ID: mdl-35328081

ABSTRACT

Phelan-McDermid syndrome (PMS) is a multi-systemic disorder characterized by both genetic and phenotypic variability. Genetic abnormalities causing PMS span from pathogenic variants of the SHANK3 gene to chromosomal rearrangements affecting the 22q13 region and leading to the loss of up to over nine megabases. The clinical presentation of individuals with PMS includes intellectual disability, neonatal hypotonia, delayed or absent speech, developmental delay, and minor dysmorphic facial features. Several other features may present with differences in age of onset and/or severity: seizures, autism, regression, sleep disorders, gastrointestinal problems, renal disorders, dysplastic toenails, and disrupted thermoregulation. Among the causes of this phenotypic variability, the size of the 22q13 deletion has effects that may be influenced by environmental factors interacting with haploinsufficiency or hemizygous variants of certain genes. Another mechanism linking environmental factors and phenotypic variability in PMS involves the loss of one copy of genes like BRD1 or CYP2D6, located at 22q13 and involved in the regulation of genomic methylation or pharmacokinetics, which are also influenced by external agents, such as diet and drugs. Overall, several non-mutually exclusive genetic and epigenetic mechanisms interact with environmental factors and may contribute to the clinical variability observed in individuals with PMS. Characterization of such factors will help to better manage this disorder.


Subject(s)
Chromosome Disorders , Nerve Tissue Proteins , Biological Variation, Population , Chromosome Deletion , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosomes, Human, Pair 22 , Humans , Infant, Newborn , Nerve Tissue Proteins/genetics
5.
EMBO Rep ; 23(5): e52606, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35297148

ABSTRACT

Mitochondrial dysfunction can either extend or decrease Caenorhabditis elegans lifespan, depending on whether transcriptionally regulated responses can elicit durable stress adaptation to otherwise detrimental lesions. Here, we test the hypothesis that enhanced metabolic flexibility is sufficient to circumvent bioenergetic abnormalities associated with the phenotypic threshold effect, thereby transforming short-lived mitochondrial mutants into long-lived ones. We find that CEST-2.2, a carboxylesterase mainly localizes in the intestine, may stimulate the survival of mitochondrial deficient animals. We report that genetic manipulation of cest-2.2 expression has a minor lifespan impact on wild-type nematodes, whereas its overexpression markedly extends the lifespan of complex I-deficient gas-1(fc21) mutants. We profile the transcriptome and lipidome of cest-2.2 overexpressing animals and show that CEST-2.2 stimulates lipid metabolism and fatty acid beta-oxidation, thereby enhancing mitochondrial respiratory capacity through complex II and LET-721/ETFDH, despite the inherited genetic lesion of complex I. Together, our findings unveil a metabolic pathway that, through the tissue-specific mobilization of lipid deposits, may influence the longevity of mitochondrial mutant C. elegans.


Subject(s)
Caenorhabditis elegans Proteins , Longevity , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Lipid Metabolism/genetics , Longevity/genetics , Mitochondria/metabolism
6.
J Clin Immunol ; 41(8): 1853-1864, 2021 11.
Article in English | MEDLINE | ID: mdl-34435264

ABSTRACT

PURPOSE: Chromosome 22q11.2 deletion syndrome is a common inborn error of immunity. The early consequences of thymic hypoplasia are low T cell numbers. Later in life, atopy, autoimmunity, inflammation, and evolving hypogammaglobulinemia can occur and the causes of these features are not understood. This study utilized an unbiased discovery approach to define alterations in histone modifications. Our goal was to identify durable chromatin changes that could influence cell behavior. METHODS: CD4 T cells and CD19 B cells underwent ChIP-seq analysis using antibodies to H3K4me3, H3K27ac, and H4ac. RNA effects were defined in CD4 T cells by RNA-seq. Serum cytokines were examined by Luminex. RESULTS: Histone marks of transcriptional activation at CD4 T cell promoters and enhancers were globally increased. The promoter activation signature had elements related to T cell activation and inflammation, concordant with effects seen in the transcriptome. B cells, in contrast, had a minimally altered epigenetic landscape in 22q11.2. Both cell types had an "edge" effect with markedly altered chromatin adjacent to the deletion. CONCLUSIONS: People with 22q11.2 deletion have altered CD4 T cell chromatin and a transcriptome concordant with the changes in the epigenome. These effects support a disease model where qualitative changes to T cells occur in addition to quantitative defects that have been well characterized. This study offers unique insight into qualitative differences in the T cells in 22q11.2 deletion, an aspect that has received limited attention.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DiGeorge Syndrome/immunology , Adolescent , Adult , B-Lymphocytes/immunology , Chromatin , Cytokines/blood , DiGeorge Syndrome/blood , Female , Histones , Humans , Male , Young Adult
7.
Philos Trans R Soc Lond B Biol Sci ; 376(1826): 20200112, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33866817

ABSTRACT

For decades, it was thought that the only heritable information transmitted from one individual to another was that encoded in the DNA sequence. However, it has become increasingly clear that this is not the case and that the transmission of molecules from within the cytoplasm of the gamete also plays a significant role in heritability. The roundworm, Caenorhabditis elegans, has emerged as one of the leading model organisms in which to study the mechanisms of transgenerational epigenetic inheritance (TEI). Collaborative efforts over the past few years have revealed that RNA molecules play a critical role in transmitting transgenerational responses, but precisely how they do so is as yet uncertain. In addition, the role of histone modifications in epigenetic inheritance is increasingly apparent, and RNA and histones interact in a way that we do not yet fully understand. Furthermore, both exogenous and endogenous RNA molecules, as well as other environmental triggers, are able to induce heritable epigenetic changes that affect transcription across the genome. In most cases, these epigenetic changes last only for a handful of generations, but occasionally can be maintained much longer: perhaps indefinitely. In this review, we discuss the current understanding of the role of RNA and histones in TEI, as well as making clear the gaps in our knowledge. We also speculate on the evolutionary implications of epigenetic inheritance, particularly in the context of a short-lived, clonally propagating species. This article is part of the theme issue 'How does epigenetics influence the course of evolution?'


Subject(s)
Caenorhabditis elegans/genetics , Chromatin/genetics , Epigenesis, Genetic , MicroRNAs/genetics , RNA, Helminth/genetics , Animals
8.
Cancers (Basel) ; 13(8)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920268

ABSTRACT

As a member of the 11-gene "death-from-cancer" gene expression signature, ubiquitin-specific protease 22 (USP22) has been considered an oncogene in various human malignancies, including colorectal cancer (CRC). We recently identified an unexpected tumor-suppressive function of USP22 in CRC and detected intestinal inflammation after Usp22 deletion in mice. We aimed to investigate the function of USP22 in intestinal inflammation as well as inflammation-associated CRC. We evaluated the effects of a conditional, intestine-specific knockout of Usp22 during dextran sodium sulfate (DSS)-induced colitis and in a model for inflammation-associated CRC. Mice were analyzed phenotypically and histologically. Differentially regulated genes were identified in USP22-deficient human CRC cells and the occupancy of active histone markers was determined using chromatin immunoprecipitation. The knockout of Usp22 increased inflammation-associated symptoms after DSS treatment locally and systemically. In addition, Usp22 deletion resulted in increased inflammation-associated colorectal tumor growth. Mechanistically, USP22 depletion in human CRC cells induced a profound upregulation of secreted protein acidic and rich in cysteine (SPARC) by affecting H3K27ac and H2Bub1 occupancy on the SPARC gene. The induction of SPARC was confirmed in vivo in our intestinal Usp22-deficient mice. Together, our findings uncover that USP22 controls SPARC expression and inflammation intensity in colitis and CRC.

9.
Acta Pharm Sin B ; 11(3): 694-707, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33777676

ABSTRACT

Targeting immune checkpoints such as programmed cell death protein 1 (PD-1) and programmed death ligand-1 (PD-L1) have been approved for treating melanoma, gastric cancer (GC) and bladder cancer with clinical benefit. Nevertheless, many patients failed to respond to anti-PD-1/PD-L1 treatment, so it is necessary to seek an alternative strategy for traditional PD-1/PD-L1 targeting immunotherapy. Here with the data from The Cancer Genome Atlas (TCGA) and our in-house tissue library, PD-L1 expression was found to be positively correlated with the expression of ubiquitin-specific processing protease 7 (USP7) in GC. Furthermore, USP7 directly interacted with PD-L1 in order to stabilize it, while abrogation of USP7 attenuated PD-L1/PD-1 interaction and sensitized cancer cells to T cell killing in vitro and in vivo. Besides, USP7 inhibitor suppressed GC cells proliferation by stabilizing P53 in vitro and in vivo. Collectively, our findings indicate that in addition to inhibiting cancer cells proliferation, USP7 inhibitor can also downregulate PD-L1 expression to enhance anti-tumor immune response simultaneously. Hence, these data posit USP7 inhibitor as an anti-proliferation agent as well as a novel therapeutic agent in PD-L1/PD-1 blockade strategy that can promote the immune response of the tumor.

10.
World J Biol Psychiatry ; 22(1): 46-57, 2021 01.
Article in English | MEDLINE | ID: mdl-32212948

ABSTRACT

OBJECTIVES: 22q11.2 deletion syndrome (DS) is the strongest known genetic risk for schizophrenia. Methylome screening was conducted to elucidate possible involvement of epigenetic alterations in the emergence of schizophrenia spectrum disorders (SZ-SD) in 22q11.2DS. METHODS: Sixteen adult men with/without SZ-SD were recruited from a 22q11.2DS cohort and underwent genome-wide DNA methylation profile analysis. Differentially methylated probes (DMPs) and regions (DMRs) were analysed using the ChAMP software. RESULTS: The DMPs (p-value <10-6) and DMRs (p-valueArea <0.01) were enriched in two gene sets, 'imprinting genes' and 'chr6p21', a region overlapping the MHC locus. Most of the identified imprinting genes are involved in neurodevelopment and located in clusters under imprinting control region (ICR) regulation, including PEG10, SGCE (7q21.3), GNAS, GNAS-AS1 (20q13.32) and SNHG14, SNURF-SNRPN, SNORD115 (15q11.2). The differentially methylated genes from the MHC locus included immune HLA-genes and non-immune genes, RNF39, PPP1R18 and NOTCH4, implicated in neurodevelopment and synaptic plasticity. The most significant DMR is located in MHC locus and covered the transcription regulator ZFP57 that is required for control and maintenance of gene imprinting at multiple ICRs. CONCLUSIONS: The differential methylation in imprinting genes and in chr6p21-22 indicate the neurodevelopmental nature of 22q11.2DS-related SZ and the major role of MHC locus in the risk to develop SZ.


Subject(s)
DiGeorge Syndrome , Schizophrenia , Adult , DNA Methylation/genetics , DiGeorge Syndrome/genetics , Genome , Genomic Imprinting/genetics , Humans , Male , Schizophrenia/genetics
11.
Front Genet ; 10: 1365, 2019.
Article in English | MEDLINE | ID: mdl-32117416

ABSTRACT

Chromosome 22q11.2 deletion syndrome (22q11.2del) is a complex, multi-organ disorder noted for its varying severity and penetrance among those affected. The clinical problems comprise congenital malformations; cardiac problems including outflow tract defects, hypoplasia of the thymus, hypoparathyroidism, and/or dysmorphic facial features. Additional clinical issues that can appear over time are autoimmunity, renal insufficiency, developmental delay, malignancy and neurological manifestations such as schizophrenia. The majority of individuals with 22q11.2del have a 3 Mb deletion of DNA on chromosome 22, leading to a haploinsufficiency of ~106 genes, which comprise coding RNAs, noncoding RNAs, and pseudogenes. The consequent haploinsufficiency of many of the coding genes are well described, including the key roles of T-box Transcription Factor 1 (TBX1) and DiGeorge Critical Region 8 (DGCR8) in the clinical phenotypes. However, the haploinsufficiency of these genes alone cannot account for the tremendous variation in the severity and penetrance of the clinical complications among those affected. Recent RNA and DNA sequencing approaches are uncovering novel genetic and epigenetic differences among 22q11.2del patients that can influence disease severity. In this review, the role of coding and non-coding genes, including microRNAs (miRNA) and long noncoding RNAs (lncRNAs), will be discussed in relation to their bearing on 22q11.2del with an emphasis on TBX1.

12.
Cell Rep ; 16(10): 2666-2685, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27568567

ABSTRACT

Human haploinsufficiency of the transcription factor Tcf4 leads to a rare autism spectrum disorder called Pitt-Hopkins syndrome (PTHS), which is associated with severe language impairment and development delay. Here, we demonstrate that Tcf4 haploinsufficient mice have deficits in social interaction, ultrasonic vocalization, prepulse inhibition, and spatial and associative learning and memory. Despite learning deficits, Tcf4(+/-) mice have enhanced long-term potentiation in the CA1 area of the hippocampus. In translationally oriented studies, we found that small-molecule HDAC inhibitors normalized hippocampal LTP and memory recall. A comprehensive set of next-generation sequencing experiments of hippocampal mRNA and methylated DNA isolated from Tcf4-deficient and WT mice before or shortly after experiential learning, with or without administration of vorinostat, identified "memory-associated" genes modulated by HDAC inhibition and dysregulated by Tcf4 haploinsufficiency. Finally, we observed that Hdac2 isoform-selective knockdown was sufficient to rescue memory deficits in Tcf4(+/-) mice.


Subject(s)
DNA Methylation/genetics , Memory , Neuronal Plasticity/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Autistic Disorder/complications , Autistic Disorder/pathology , Autistic Disorder/physiopathology , CpG Islands/genetics , DNA Methylation/drug effects , Disease Models, Animal , Facies , Gene Expression Profiling , Gene Knockdown Techniques , Hippocampus/metabolism , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Hyperventilation/complications , Hyperventilation/genetics , Hyperventilation/pathology , Hyperventilation/physiopathology , Intellectual Disability/complications , Intellectual Disability/genetics , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Mice , Motor Activity/drug effects , Neuronal Plasticity/drug effects , Prepulse Inhibition/drug effects , Transcription Factor 7-Like 2 Protein/genetics , Transcription, Genetic/drug effects , Vorinostat
13.
Environ Mol Mutagen ; 57(3): 200-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26801515

ABSTRACT

Persistent organic pollutants (POPs), such as PCBs (polychlorinated biphenyls) and DDT [1,1,1-trichloro-2,2-bis(4-chlorophenyl)ethane], are environmental contaminants with potential endocrine disrupting activity. DNA methylation levels in peripheral blood lymphocytes have been associated with serum concentrations of POPs in Greenland Inuit and Korean populations. Greenland Inuits are characterized by the highest worldwide POP levels. In this cross-sectional study we evaluated the relationship between serum POP concentrations and DNA methylation levels in sperm of non-occupationally exposed fertile men from Greenland, Warsaw (Poland), and Kharkiv (Ukraine). Serum levels of PCB-153 [1,2,4-trichloro-5-(2,4,5-trichlorophenyl)benzene], as a proxy of the total PCBs body burden, and of p,p'-DDE [1,1-dichloro-2,2-bis(4-chlorophenyl)ethylene], the main metabolite of DDT were measured. Sperm DNA methylation level was assessed globally by flow cytometric (FCM) immunodetection of 5-methyl-cytosines and at specific repetitive DNA sequences (Alu, LINE-1, Satα) by PCR-pyrosequencing after bisulfite conversion. Multivariate linear regression analysis was applied to investigate correlations between serum POP concentrations and DNA methylation. No consistent associations between exposure to POPs and sperm DNA methylation at repetitive DNA sequences were detected. A statistically significant global decrease in methylation was associated with exposure to either POP by FCM analysis. This is the first study to investigate environmental exposure to POPs and DNA methylation levels considering sperm as the target cells. Although POP exposure appears to have a limited negative impact on sperm DNA methylation levels in adult males, the global hypomethylation detected by one of the methods applied suggests that further investigation is warranted.


Subject(s)
DNA Methylation/drug effects , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Spermatozoa/drug effects , Adult , Alu Elements , DDT/toxicity , Dichlorodiphenyl Dichloroethylene/blood , Dichlorodiphenyl Dichloroethylene/toxicity , Environmental Exposure/analysis , Environmental Pollutants/blood , Epigenesis, Genetic/drug effects , Greenland , Humans , Long Interspersed Nucleotide Elements , Male , Poland , Polychlorinated Biphenyls/blood , Polychlorinated Biphenyls/toxicity , Spermatozoa/physiology , Ukraine
14.
Hum Pathol ; 46(7): 1006-14, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25971547

ABSTRACT

This study aimed to evaluate the expression of H2B monoubiquitination enzyme (uH2B) and ubiquitin-specific protease enzyme 22 (USP22) in colon carcinoma and establish a correlation between the expression of these enzymes and clinicopathological parameters. The modification levels of uH2B and USP22 in 20 noncancerous and 129 cancerous colon samples were studied by immunohistochemistry. We used a dual-rated semiquantitative method to classify the expression according to 3 levels and analyzed these results. uH2B was abundant in the normal colon epithelium, but its expression was decreased in colon cancers (P < .001); the uH2B modification level correlated with tumor differentiation (P < .001), lymph node metastasis (P = .017), distant metastasis (P = .036), and tumor stage (P = .039). The USP22 expression in colon carcinoma was higher than that in normal tissues (P = .007) and negatively correlated with the degree of differentiation (P = .006), invasion (P = .025), lymph node metastasis (P = .026), and tumor stage (P = .044). uH2B and USP22 expression negatively correlated (r = -0.401, P < .001). Patients with uH2B-negative and USP22-positive staining were found to have lower survival rates (30.737 ± 2.866 versus 51.667 ± 2.286 months, P < .001). Positive uH2B and negative USP22 expression remained a statistically significant prognostic indicator in a multivariate Cox regression analysis (hazard ratio, 2.557; 95% confidence interval, 1.043-6.269; P = .04). We conclude that uH2B displays differential staining patterns according to progressive stages of colon cancer, indicating that uH2B may play an important inhibitory role in carcinogenesis. Increased USP22 expression in colon cancer correlated with reduced uH2B expression, and this expression pattern may contribute to tumor progression.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/enzymology , Colonic Neoplasms/enzymology , Histones/analysis , Thiolester Hydrolases/analysis , Adult , Aged , Aged, 80 and over , Carcinoma/mortality , Carcinoma/secondary , Case-Control Studies , Cell Differentiation , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Staging , Predictive Value of Tests , Proportional Hazards Models , Ubiquitin Thiolesterase , Ubiquitination , Young Adult
15.
Curr Colorectal Cancer Rep ; 11(6): 414-421, 2015 Dec.
Article in English | MEDLINE | ID: mdl-30655730

ABSTRACT

Colorectal cancer stem cells (CSCs) were initially considered to be a subset of undifferentiated tumor cells with well-defined phenotypic and molecular markers. However, emerging evidence indicates instead that colorectal CSCs are heterogeneous subsets of tumor cells that are continuously reshaped by the dynamic interactions between genetic, epigenetic, and immune factors in the tumor microenvironment. Thus, the colorectal CSC phenotypes and responsiveness to therapy may not only be a tumor cell-intrinsic feature, but also depend on tumor-extrinsic microenvironmental factors. Furthermore, emerging evidence also implicates colorectal CSCs in potential immune evasion. Therefore, understanding how colorectal CSC-intrinsic mechanisms cooperate with the extrinsic microenvironmental factors to dynamically shape colorectal CSC resistance to chemotherapy and immunotherapy holds great promise for development of targeted CSC therapies of advanced human CRC.

16.
Worm ; 3: e28641, 2014.
Article in English | MEDLINE | ID: mdl-25254148

ABSTRACT

Endogenous small RNA pathways related to RNA interference (RNAi) play a well-documented role in protecting host genomes from the invasion of foreign nucleic acids. In C. elegans, the PIWI type Argonaute, PRG-1, through an association with 21U-RNAs, mediates a genome surveillance process by constantly scanning the genome for potentially deleterious invading elements. Upon recognition of foreign nucleic acids, PRG-1 initiates a cascade of cytoplasmic and nuclear events that results in heritable epigenetic silencing of these transcripts and their coding genomic loci. If the PRG-1/21U-RNA genome surveillance pathway has the capacity to target most of the C. elegans transcriptome, what mechanisms exist to protect endogenous transcripts from being silenced by this pathway? In this commentary, we discuss three recent publications that implicate the CSR-1 small RNA pathway in the heritable activation of germline transcripts, propose a model as to why not all epialleles behave similarly, and touch on the practical implications of these findings.

17.
Epigenetics ; 9(1): 62-74, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24149573

ABSTRACT

While most eukaryotic genomes contain transposable elements that can provide select evolutionary advantages to a given organism, failure to tightly control the mobility of such transposable elements can result in compromised genomic integrity of both parental and subsequent generations. Together with the Piwi subfamily of Argonaute proteins, small, non-coding Piwi-interacting RNAs (piRNAs) primarily function in the germ line to defend the genome against the potentially deleterious effects that can be caused by transposition. Here, we describe recent discoveries concerning the biogenesis and function of piRNAs in the nematode Caenorhabditis elegans, illuminating how the faithful production of these mature species can impart a robust defense mechanism for the germ line to counteract problems caused by foreign genetic elements across successive generations by contributing to the epigenetic memory of non-self vs. self.


Subject(s)
Caenorhabditis elegans/metabolism , Germ Cells/metabolism , RNA, Small Interfering/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , DNA Transposable Elements , Epigenesis, Genetic , Genomic Instability , Humans , RNA, Small Interfering/genetics , Reproduction
18.
Mol Metab ; 2(4): 337-47, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-24327950

ABSTRACT

Obesity is characterized as an excess accumulation of body fat resulting from a positive energy balance. It is the major risk factor for type 2 diabetes (T2D). The evidence for familial aggregation of obesity and its associated metabolic diseases is substantial. To date, about 150 genetic loci identified in genome-wide association studies (GWAS) are linked with obesity and T2D, each accounting for only a small proportion of the predicted heritability. However, the percentage of overall trait variance explained by these associated loci is modest (~5-10% for T2D, ~2% for BMI). The lack of powerful genetic associations suggests that heritability is not entirely attributable to gene variations. Some of the familial aggregation as well as many of the effects of environmental exposures, may reflect epigenetic processes. This review summarizes our current knowledge on the genetic basis to individual risk of obesity and T2D, and explores the potential role of epigenetic contribution.

19.
J Biol Chem ; 288(33): 24234-46, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23760504

ABSTRACT

Pluripotent embryonic stem cells (ESCs) undergo self-renewal until stimulated to differentiate along specific lineage pathways. Many of the transcriptional networks that drive reprogramming of a self-renewing ESC to a differentiating cell have been identified. However, fundamental questions remain unanswered about the epigenetic programs that control these changes in gene expression. Here we report that the histone ubiquitin hydrolase ubiquitin-specific protease 22 (USP22) is a critical epigenetic modifier that controls this transition from self-renewal to differentiation. USP22 is induced as ESCs differentiate and is necessary for differentiation into all three germ layers. We further report that USP22 is a transcriptional repressor of the locus encoding the core pluripotency factor sex-determining region Y-box 2 (SOX2) in ESCs, and this repression is required for efficient differentiation. USP22 occupies the Sox2 promoter and hydrolyzes monoubiquitin from ubiquitylated histone H2B and blocks transcription of the Sox2 locus. Our study reveals an epigenetic mechanism that represses the core pluripotency transcriptional network in ESCs, allowing ESCs to transition from a state of self-renewal into lineage-specific differentiation programs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endopeptidases/metabolism , Epigenesis, Genetic , SOXB1 Transcription Factors/genetics , Transcription, Genetic , Ubiquitin-Specific Proteases/metabolism , Animals , Cell Line , Cell Proliferation , Endopeptidases/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Histones/metabolism , Mice , Phenotype , Pluripotent Stem Cells/metabolism , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism , Sirtuin 1/metabolism , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics
20.
J Korean Med Sci ; 24(1): 97-103, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19270820

ABSTRACT

Epithelial membrane protein 3 (EMP3) is a trans-membrane signaling molecule with important roles in the regulation of apoptosis, differentiation and invasion of cancer cells, but the detailed is largely still unknown. We analyzed the mRNA levels and methylation statuses of EMP3 in 63 primary breast carcinomas and assessed their correlations with clinicopathologic variables. The expression of EMP3 mRNA in primary breast carcinomas was significantly higher than the expression of 20 normal breast tissues (p<10(-7)). EMP3 overexpression in breast carcinomas was significantly related to histological grade III (p=3.9 x 10(-7)), lymph node metastasis (p=0.003), and strong Her-2 expression (p=3.3 x 10(-6)). Hypermethylation frequencies of EMP3 were detected in 36.5% of breast carcinomas by methylation-specific polymerase chain reaction. However, no significant correlations were found between methylation status of EMP3 and mRNA expression levels as well as other clinical parameters. In conclusion, EMP3 may be a novel marker of tumor aggressiveness. Overexpression of EMP3 in primary breast carcinoma is not associated with DNA methylation.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Membrane Glycoproteins/genetics , Adult , Breast Neoplasms/pathology , Carcinoma/pathology , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Membrane Glycoproteins/metabolism , Middle Aged , Neoplasm Staging , RNA, Messenger/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...