Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
3.
Front Immunol ; 14: 1175348, 2023.
Article in English | MEDLINE | ID: mdl-37223095

ABSTRACT

Background and aim: Type I interferons (IFNs) are highly expressed in the gut mucosa of celiac disease (CD) gut mucosa and stimulates immune response prompted by gluten ingestion, but the processes that maintain the production of these inflammatory molecules are not well understood. Adenosine deaminase acting on RNA 1 (ADAR1), an RNA-editing enzyme, plays a crucial role in inhibiting self or viral RNAs from activating auto-immune mediated responses, most notably within the type-I IFN production pathway. The aim of this study was to assess whether ADAR1 could contribute to the induction and/or progression of gut inflammation in patients with celiac disease. Material and methods: ADAR1 expression was assessed by Real time PCR and Western blotting in duodenal biopsy taken from inactive and active celiac disease (CD) patients and normal controls (CTR). To analyze the role of ADAR1 in inflamed CD mucosa, lamina propria mononuclear cells (LPMC) were isolated from inactive CD and ADAR1 was silenced in with a specific antisense oligonucleotide (AS) and then incubated with a synthetic analogue of viral dsRNA (poly I:C). IFN-inducing pathways (IRF3, IRF7) in these cells were evaluated with Western blotting and inflammatory cytokines were evaluated with flow cytometry. Lastly, the role of ADAR1 was investigated in a mouse model of poly I:C-driven small intestine atrophy. Results: Reduced ADAR1 expression was seen in duodenal biopsies compared to inactive CD and normal controls. Ex vivo organ cultures of duodenal mucosal biopsies, taken from inactive CD patients, stimulated with a peptic-tryptic digest of gliadin displayed a decreased expression of ADAR1. ADAR1 silencing in LPMC stimulated with a synthetic analogue of viral dsRNA strongly boosted the activation of IRF3 and IRF7 and the production of type-I IFN, TNF-α and IFN-γ. Administration of ADAR1 antisense but not sense oligonucleotide to mice with poly I:C-induced intestinal atrophy, significantly increased gut damage and inflammatory cytokines production. Conclusions: These data show that ADAR1 is an important regulator of intestinal immune homeostasis and demonstrate that defective ADAR1 expression could provide to amplifying pathogenic responses in CD intestinal mucosa.


Subject(s)
Celiac Disease , Animals , Mice , Celiac Disease/genetics , Adenosine Deaminase/genetics , Intestinal Mucosa , RNA, Double-Stranded , Atrophy , Cytokines , Poly I
4.
Int J Mol Sci ; 24(3)2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36768150

ABSTRACT

Decades of research have investigated the mechanisms that lead to the origin of cancer, striving to identify tumor-initiating cells. These cells, also known as cancer stem cells, are characterized by the ability to self-renew, to give rise to differentiated tumor populations, and on a larger scale, are deemed responsible not only for tumor initiation but also for recurrent tumors, often resistant to chemotherapy and radiotherapy. Long noncoding RNAs are RNA molecules longer than 200 nt, lacking the ability to code for proteins, with recognized roles as fine regulators of gene expression. They can exert these functions through a variety of mechanisms, acting at almost all steps of gene expression, from modulation of the epigenetic state of chromatin to modulation of protein stability. In all cases, lncRNAs do not work alone, but they always interact with other RNA molecules, either coding or non-coding, or with protein factors. In this review, we summarize the latest results obtained about the involvement of lncRNAs in the initiating cells of several types of tumors, and highlight the different mechanisms through which they work, while discussing how the modulation of a lncRNA can affect several aspects of tumor onset and progression.


Subject(s)
RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Neoplasm Recurrence, Local/genetics , Chromatin , Neoplastic Stem Cells/metabolism , Gene Expression Regulation, Neoplastic
5.
Mol Oncol ; 17(2): 238-260, 2023 02.
Article in English | MEDLINE | ID: mdl-36495079

ABSTRACT

Glioblastoma (GBM) cancer stem cells (GSCs) contribute to GBM's origin, recurrence, and resistance to treatment. However, the understanding of how mRNA expression patterns of GBM subtypes are reflected at global proteome level in GSCs is limited. To characterize protein expression in GSCs, we performed in-depth proteogenomic analysis of patient-derived GSCs by RNA-sequencing and mass-spectrometry. We quantified > 10 000 proteins in two independent GSC panels and propose a GSC-associated proteomic signature characterizing two distinct phenotypic conditions; one defined by proteins upregulated in proneural and classical GSCs (GPC-like), and another by proteins upregulated in mesenchymal GSCs (GM-like). The GM-like protein set in GBM tissue was associated with necrosis, recurrence, and worse overall survival. Through proteogenomics, we discovered 252 non-canonical peptides in the GSCs, i.e., protein sequences that are variant or derive from genome regions previously considered non-protein-coding, including variants of the heterogeneous ribonucleoproteins implicated in RNA splicing. In summary, GSCs express two protein sets that have an inverse association with clinical outcomes in GBM. The discovery of non-canonical protein sequences questions existing gene models and pinpoints new protein targets for research in GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/metabolism , Proteomics , Brain Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Cell Line, Tumor
6.
Cancers (Basel) ; 14(9)2022 May 06.
Article in English | MEDLINE | ID: mdl-35565433

ABSTRACT

The most widely accepted hypothesis for the development of glioblastoma suggests that glioblastoma stem-like cells (GSCs) are crucially involved in tumor initiation and recurrence as well as in the occurrence of chemo- and radio-resistance. Mesenchyme homeobox 2 (MEOX2) is a transcription factor overexpressed in glioblastoma, whose expression is negatively correlated with patient survival. Starting from our observation that MEOX2 expression is strongly enhanced in six GSC lines, we performed shRNA-mediated knock-down experiments in two different GSC lines and found that MEOX2 depletion resulted in the inhibition of cell growth and sphere-forming ability and an increase in apoptotic cell death. By a deep transcriptome analysis, we identified a core group of genes modulated in response to MEOX2 knock-down. Among these genes, the repressed ones are largely enriched in genes involved in the hypoxic response and glycolytic pathway, two strictly related pathways that contribute to the resistance of high-grade gliomas to therapies. An in silico study of the regulatory regions of genes differentially expressed by MEOX2 knock-down revealed that they mainly consisted of GC-rich regions enriched for Sp1 and Klf4 binding motifs, two main regulators of metabolism in glioblastoma. Our results show, for the first time, the involvement of MEOX2 in the regulation of genes of GSC metabolism, which is essential for the survival and growth of these cells.

7.
RNA Biol ; 18(sup1): 75-87, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34224323

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) are enzymes that convert adenosines to inosines in double-stranded RNAs (RNA editing A-to-I). ADAR1 and ADAR2 were previously reported as HIV-1 proviral factors. The aim of this study was to investigate the composition of the ADAR2 ribonucleoprotein complex during HIV-1 expression. By using a dual-tag affinity purification procedure in cells expressing HIV-1 followed by mass spectrometry analysis, we identified 10 non-ribosomal ADAR2-interacting factors. A significant fraction of these proteins was previously found associated to the Long INterspersed Element 1 (LINE1 or L1) ribonucleoparticles and to regulate the life cycle of L1 retrotransposons. Considering that we previously demonstrated that ADAR1 is an inhibitor of LINE-1 retrotransposon activity, we investigated whether also ADAR2 played a similar function. To reach this goal, we performed specific cell culture retrotransposition assays in cells overexpressing or ablated for ADAR2. These experiments unveil a novel function of ADAR2 as suppressor of L1 retrotransposition. Furthermore, we showed that ADAR2 binds the basal L1 RNP complex.Overall, these data support the role of ADAR2 as regulator of L1 life cycle.


Subject(s)
Adenosine Deaminase/metabolism , Long Interspersed Nucleotide Elements , RNA Editing , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , HEK293 Cells , HeLa Cells , Humans , RNA-Binding Proteins/genetics
8.
Methods Mol Biol ; 2181: 269-286, 2021.
Article in English | MEDLINE | ID: mdl-32729086

ABSTRACT

The type I interferonopathies comprise a heterogenous group of monogenic diseases associated with a constitutive activation of type I interferon signaling.The elucidation of the genetic causes of this group of diseases revealed an alteration of nucleic acid processing and signaling.ADAR1 is among the genes found mutated in patients with this type of disorders.This enzyme catalyzes the hydrolytic deamination of adenosines in inosines within a double-stranded RNA target (RNA editing of A to I). This RNA modification is widespread in human cells and deregulated in a variety of human diseases, ranging from cancers to neurological abnormalities.In this review, we briefly summarize the knowledge about the RNA editing alterations occurring in patients with mutations in ADAR1 gene and how these alterations might cause the inappropriate IFN activation.


Subject(s)
Genetic Diseases, Inborn/genetics , Interferon Type I/genetics , RNA Editing/physiology , Adenosine Deaminase/genetics , Aortic Diseases/genetics , Autoimmune Diseases of the Nervous System/genetics , Dental Enamel Hypoplasia/genetics , Humans , Immunity, Innate/genetics , Interferon Type I/metabolism , Metacarpus/abnormalities , Muscular Diseases/genetics , Nervous System Malformations/genetics , Odontodysplasia/genetics , Osteoporosis/genetics , RNA, Double-Stranded/genetics , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Vascular Calcification/genetics
9.
Int J Mol Sci ; 21(8)2020 Apr 17.
Article in English | MEDLINE | ID: mdl-32316617

ABSTRACT

N6-methyladenosine (m6A) is one of the most widespread and abundant internal messenger RNA modifications found in eukaryotes. Emerging evidence suggests that this modification is strongly linked to the activation and inhibition of cancer pathways and is associated with prognostically significant tumour subtypes. The present review describes the dynamic nature of m6A regulator enzymes, as methyltransferases, demethylases and m6A binding proteins, and points out thevalue of the balance among these proteins in regulating gene expression, cell metabolism and cancer development. The main focus of this review is on the roles of m6A modification in glioblastoma, the most aggressive and invariably lethal brain tumour. Although the study of m6A in glioblastoma is a young one, and papers in this field can yield divergent conclusions, the results collected so far clearly demonstrate that modulation of mRNA m6A levels impacts multiple aspects of this tumour, including growth, glioma stem cells self-renewal, and tumorigenesis, suggesting that mRNA m6A modification may serve as a promising target for glioblastoma therapy. We also present recent data about another type of epitranscriptomic modification, the methylation of cytosine at a specific site of 28S rRNA, as it was recently shown to affect the biology of glioma cells, with high potential of clinical implications.


Subject(s)
Adenosine/analogs & derivatives , Brain Neoplasms/genetics , Glioblastoma/genetics , Adenosine/metabolism , Cytosine/chemistry , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Methylation , RNA, Ribosomal, 28S/genetics
10.
Int J Mol Sci ; 20(10)2019 May 21.
Article in English | MEDLINE | ID: mdl-31117166

ABSTRACT

Glioblastoma (GBM) is a primary brain tumor whose prognosis is inevitably dismal, leading patients to death in about 15 months from diagnosis. Tumor cells in the mass of the neoplasm are in continuous exchange with cells of the stromal microenvironment, through the production of soluble molecules, among which chemokines play prominent roles. CXCL14 is a chemokine with a pro-tumor role in breast and prostate carcinoma, where it is secreted by cancer associated fibroblasts, and contributes to tumor growth and invasion. We previously observed that CXCL14 expression is higher in GBM tissues than in healthy white matter. Here, we study the effects of exogenously supplemented CXCL14 on key tumorigenic properties of human GBM cell lines. We show that CXCL14 enhances the migration ability and the proliferation of U87MG and LN229 GBM cell lines. None of these effects was affected by the use of AMD3100, an inhibitor of CXCR4 receptor, suggesting that the observed CXCL14 effects are not mediated by this receptor. We also provide evidence that CXCL14 enhances the sphere-forming ability of glioblastoma stem cells, considered the initiating cells, and is responsible for tumor onset, growth and recurrence. In support of our in vitro results, we present data from several GBM expression datasets, demonstrating that CXCL14 expression is inversely correlated with overall survival, that it is enriched at the leading edge of the tumors and in infiltrating tumor areas, and it characterizes mesenchymal and NON G-CIMP tumors, known to have a particularly bad prognosis. Overall, our results point to CXCL14 as a protumorigenic chemokine in GBM.


Subject(s)
Cell Movement , Cell Proliferation , Chemokines, CXC/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/physiopathology , Humans , Tumor Microenvironment
11.
Virology ; 523: 121-128, 2018 10.
Article in English | MEDLINE | ID: mdl-30119013

ABSTRACT

The HIV-1 accessory protein Vpr displays various activities that can favor viral replication such as G2 cell cycle arrest. Vpr also modulates host gene expression, although this property is poorly characterized. Here, we investigated the effect of Vpr on L-selectin (CD62L), which crucially controls leukocytes circulation and generation of immune responses against pathogens. We report that Vpr up-regulates CD62L mRNA level when individually expressed in Jurkat T cells as well as during HIV-1 infection of primary CD4+ T cells. Vpr mutant analysis and use of inhibitors suggest that the effect of Vpr on CD62L occurs independently of G2 arrest but requires activation of the ATR kinase. Yet, induction of CD62L expression by Vpr is contrasted by down-regulation of CD62L protein by Nef that, together with Vpu, induces a net reduction of cell-surface CD62L on HIV-1-infected cells, which may impact viral spread and evasion of immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV-1/genetics , Human Immunodeficiency Virus Proteins/genetics , L-Selectin/genetics , Viral Regulatory and Accessory Proteins/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , CD4-Positive T-Lymphocytes/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation , HIV-1/metabolism , Histones/genetics , Histones/metabolism , Host-Pathogen Interactions , Human Immunodeficiency Virus Proteins/metabolism , Humans , Jurkat Cells , L-Selectin/metabolism , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Viral Regulatory and Accessory Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism
12.
Oncotarget ; 9(21): 15512-15525, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29643989

ABSTRACT

The still largely obscure molecular events in the glioblastoma oncogenesis, a primary brain tumor characterized by an inevitably dismal prognosis, impel for investigation. The importance of Long noncoding RNAs as regulators of gene expression has recently become evident. Among them, H19 has a recognized oncogenic role in several types of human tumors and was shown to correlate to some oncogenic aspects of glioblastoma cells. Here we, hypothesyze that in glioblastoma H19 exerts its function through the interaction with the catalytic subunit of the PRC2 complex, EZH2. By employing a factor analysis on a SAGE dataset of 12 glioblastoma samples, we show that H19 expression in glioblastoma tissues correlates with that of several genes involved in glioblastoma growth and progression. H19 knock-down reduces viability, migration and invasiveness of two distinct human glioblastoma cell lines. Most importantly, we provide a mechanistic perspective about the role of H19 in glioblastoma cells, by showing that its expression is inversely linked to that of NKD1, a negative regulator of Wnt pathway, suggesting that H19 might regulate NKD1 transcription via EZH2-induced H3K27 trimethylation of its promoter. Indeed, we showed that H19 binds EZH2 in glioblastoma cells, and that EZH2 binding to NKD1 and other promoters is impaired by H19 silencing. In this work we describe H19 as part of an epigenetic modulation program executed by EZH2, that results in the repression of Nkd1. We believe that our results can provide a new piece to the complex puzzle of H19 function in glioblastoma.

13.
Chromosome Res ; 26(1-2): 45-59, 2018 03.
Article in English | MEDLINE | ID: mdl-29396793

ABSTRACT

Long interspersed element-1 (LINE-1 or L1) retrotransposons represent the only functional family of autonomous transposable elements in humans and formed 17% of our genome. Even though most of the human L1 sequences are inactive, a limited number of copies per individual retain the ability to mobilize by a process termed retrotransposition. The ongoing L1 retrotransposition may result in insertional mutagenesis that could lead to negative consequences such as genetic disease and cancer. For this reason, cells have evolved several mechanisms of defense to restrict L1 activity. Among them, a critical role for cellular deaminases [activation-induced deaminase (AID)/apolipoprotein B mRNA-editing catalytic polypeptide-like (APOBEC) and adenosine deaminases that act on RNA (ADAR) enzymes] has emerged. The majority of the AID/APOBEC family of proteins are responsible for the deamination of cytosine to uracil (C-to-U editing) within DNA and RNA targets. The ADARs convert adenosine bases to inosines (A-to-I editing) within double-stranded RNA (dsRNA) targets. This review will discuss the current understanding of the regulation of LINE-1 retrotransposition mediated by these enzymes.


Subject(s)
APOBEC Deaminases/metabolism , Adenosine Deaminase/metabolism , Cytidine Deaminase/metabolism , Long Interspersed Nucleotide Elements , Retroelements , DNA/metabolism , Humans , RNA/metabolism , RNA, Double-Stranded/metabolism
14.
RNA Biol ; 14(11): 1485-1491, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28640667

ABSTRACT

ADAR1 is an enzyme that belongs to the Adenosine Deaminases Acting on RNA (ADARs) family. These enzymes deaminate adenosines to inosines (RNA editing A-to-I) within double-stranded RNA regions in transcripts. Since inosines are recognized as guanosines by the cellular machinery, RNA editing mediated by ADARs can either lead to the formation of an altered protein (recoding) or affect different aspects of RNA metabolism. Recently, a proteomic analysis led to the identification of novel ADAR1-associated factors and found that a good fraction of them is shared with the Long Interspersed Element 1 (LINE-1 or L1) ribonucleoparticles (RNPs). This evidence suggested a possible role of ADAR1 in regulating the L1 life cycle. By taking advantage of the use of cell culture retrotransposition assays, a novel function of this deaminase as an inhibitor of L1 retrotransposition was demonstrated. These results pave the way toward a better comprehension of the mechanisms of restriction of retrotransposons.


Subject(s)
Adenosine Deaminase/genetics , Genome, Human , Long Interspersed Nucleotide Elements , RNA Editing , RNA-Binding Proteins/genetics , RNA/genetics , Adenosine/metabolism , Adenosine Deaminase/metabolism , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/pathology , Biological Assay , HeLa Cells , Humans , Inosine/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , RNA/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
15.
Oncotarget ; 8(9): 15704-15715, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28152517

ABSTRACT

BACKGROUND: An impaired HBsAg-secretion can increase HBV oncogenic-properties. Here, we investigate genetic-determinants in HBsAg correlated with HBV-induced hepatocellular carcinoma (HCC), and their impact on HBsAg-secretion and cell-proliferation. METHODS: This study included 128 chronically HBV-infected patients: 23 with HCC (73.9% D; 26.1% A HBV-genotype), and 105 without cirrhosis/HCC (72.4% D, 27.6% A) as reference-group. The impact of mutations on HBsAg-secretion was assessed by measuring the ratio [secreted/intracellular HBsAg] until day 5 post-transfection. The impact of mutations on cell-cycle advancement was assessed by flow-cytometry. RESULTS: Two HBsAg mutations significantly correlated with HCC: P203Q (17.4% [4/23] in HCC vs 1.0% [1/105] in non-HCC, P=0.004); S210R (34.8% [8/23] in HCC vs 3.8% [4/105] in non-HCC, P <0.001); P203Q+S210R (17.4% [4/23] in HCC vs 0% [0/110] in non-HCC, P=0.001). Both mutations reside in trans-membrane C-terminal domain critical for HBsAg-secretion. In in-vitro experiments, P203Q, S210R and P203Q+S210R significantly reduced the ratio [secreted/intracellular HBsAg] compared to wt at each time-point analysed (P <0.05), supporting an impaired HBsAg-secretion. Furthermore, P203Q and P203Q+S210R increased the percentage of cells in S-phase compared to wt, indicating cell-cycle progression (P203Q:26±13%; P203Q+S210R:29±14%; wt:18%±9, P <0.01. Additionally, S210R increased the percentage of cells in G2/M-phase (26±8% for wt versus 33±6% for S210R, P <0.001). CONCLUSIONS: Specific mutations in HBsAg C-terminus significantly correlate with HBV-induced HCC. They hamper HBsAg-secretion and are associated with increased cellular proliferation, supporting their involvement in HCC-development. The identification of viral genetic markers associated with HCC is critical to identify patients at higher HCC-risk that may deserve intensive liver monitoring, and/or early anti-HBV therapy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hepatitis B Surface Antigens/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic/pathology , Liver Neoplasms/pathology , Mutation , Adult , Aged , Carcinoma, Hepatocellular/virology , Cell Cycle , Cell Proliferation , Female , Gene Frequency , Genotype , Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/metabolism , Hepatitis B virus/physiology , Hepatitis B, Chronic/virology , Host-Pathogen Interactions , Humans , Liver Neoplasms/virology , Male , Middle Aged , Multivariate Analysis , Risk Factors
16.
Nucleic Acids Res ; 45(1): 155-168, 2017 01 09.
Article in English | MEDLINE | ID: mdl-27658966

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) are involved in RNA editing that converts adenosines to inosines in double-stranded RNAs. ADAR1 was demonstrated to be functional on different viruses exerting either antiviral or proviral effects. Concerning HIV-1, several studies showed that ADAR1 favors viral replication. The aim of this study was to investigate the composition of the ADAR1 ribonucleoprotein complex during HIV-1 expression. By using a dual-tag affinity purification procedure in cells expressing HIV-1 followed by mass spectrometry analysis, we identified 14 non-ribosomal ADAR1-interacting proteins, most of which are novel. A significant fraction of these proteins were previously demonstrated to be associated to the Long INterspersed Element 1 (LINE1 or L1) ribonucleoparticles and to regulate the life cycle of L1 retrotransposons that continuously re-enter host-genome.Hence, we investigated the function of ADAR1 in the regulation of L1 activity.By using different cell-culture based retrotransposition assays in HeLa cells, we demonstrated a novel function of ADAR1 as suppressor of L1 retrotransposition. Apparently, this inhibitory mechanism does not occur through ADAR1 editing activity. Furthermore, we showed that ADAR1 binds the basal L1 RNP complex. Overall, these data support the role of ADAR1 as regulator of L1 life cycle.


Subject(s)
Adenosine Deaminase/genetics , HIV-1/genetics , Long Interspersed Nucleotide Elements , RNA-Binding Proteins/genetics , Retroelements , Ribonucleoproteins/genetics , Adenosine Deaminase/metabolism , Biological Assay , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , HIV-1/metabolism , HeLa Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Host-Pathogen Interactions , Humans , Molecular Sequence Annotation , Protein Binding , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Signal Transduction
17.
EMBO Rep ; 17(12): 1872-1889, 2016 12.
Article in English | MEDLINE | ID: mdl-27852622

ABSTRACT

MYC deregulation is common in human cancer and has a role in sustaining the aggressive cancer stem cell populations. MYC mediates a broad transcriptional response controlling normal biological programmes, but its activity is not clearly understood. We address MYC function in cancer stem cells through the inducible expression of Omomyc-a MYC-derived polypeptide interfering with MYC activity-taking as model the most lethal brain tumour, glioblastoma. Omomyc bridles the key cancer stemlike cell features and affects the tumour microenvironment, inhibiting angiogenesis. This occurs because Omomyc interferes with proper MYC localization and itself associates with the genome, with a preference for sites occupied by MYC This is accompanied by selective repression of master transcription factors for glioblastoma stemlike cell identity such as OLIG2, POU3F2, SOX2, upregulation of effectors of tumour suppression and differentiation such as ID4, MIAT, PTEN, and modulation of the expression of microRNAs that target molecules implicated in glioblastoma growth and invasion such as EGFR and ZEB1. Data support a novel view of MYC as a network stabilizer that strengthens the regulatory nodes of gene expression networks controlling cell phenotype and highlight Omomyc as model molecule for targeting cancer stem cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, myc , Glioblastoma/genetics , Neoplastic Stem Cells/physiology , Peptide Fragments/genetics , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Angiogenesis Inhibitors , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Proliferation , ErbB Receptors/genetics , Glioblastoma/physiopathology , Humans , Inhibitor of Differentiation Proteins/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Protein Binding , Transcriptional Activation , Tumor Microenvironment/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics
18.
Sci Rep ; 6: 25219, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27142352

ABSTRACT

The cytoplasmic element binding protein 1 (CPEB1) regulates many important biological processes ranging from cell cycle control to learning and memory formation, by controlling mRNA translation efficiency via 3' untranslated regions (3'UTR). In the present study, we show that CPEB1 is significantly downregulated in human Glioblastoma Multiforme (GBM) tissues and that the restoration of its expression impairs glioma cell lines growth. We demonstrate that CPEB1 promotes the expression of the cell cycle inhibitor p27(Kip1) by specifically targeting its 3'UTR, and competes with miR-221/222 binding at an overlapping site in the 3'UTR, thus impairing miR-221/222 inhibitory activity. Upon binding to p27(Kip1) 3'UTR, CPEB1 promotes elongation of poly-A tail and the subsequent translation of p27(Kip1) mRNA. This leads to higher levels of p27(Kip1) in the cell, in turn significantly inhibiting cell proliferation, and confers to CPEB1 a potential value as a tumor suppressor in Glioblastoma.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Gene Expression Regulation , Glioblastoma/pathology , Protein Biosynthesis , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Cell Line, Tumor , Humans , Neuroglia/physiology
19.
Virology ; 485: 475-80, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363218

ABSTRACT

Adenosine deaminase acting on RNA1 (ADAR1) was previously reported to affect HIV-1 replication. We report data showing that ADAR1 interacts with the HIV-1 p55 Gag protein, the major structural protein of the immature virus capsid. Furthermore, we found that the endogenous ADAR1 is incorporated into virions purified from the supernatant of primary HIV-1-infected CD4(+) T lymphocytes. Additional experiments demonstrated that the expression of the p55 Gag protein is sufficient for ADAR1 incorporation into virus-like particles (VLPs). Overall, our data originally support the evidence that ADAR1 can be part of the cell protein array uploaded in HIV-1 particles.


Subject(s)
Adenosine Deaminase/metabolism , HIV-1/physiology , RNA-Binding Proteins/metabolism , Virion , Adenosine Deaminase/chemistry , Cell Line , Humans , Protein Binding , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , gag Gene Products, Human Immunodeficiency Virus/metabolism
20.
PLoS One ; 9(12): e113908, 2014.
Article in English | MEDLINE | ID: mdl-25462981

ABSTRACT

HIV-1 preferentially infects CD4+ T cells, causing fundamental changes that eventually lead to the release of new viral particles and cell death. To investigate in detail alterations in the transcriptome of the CD4+ T cells upon viral infection, we sequenced polyadenylated RNA isolated from Jurkat cells infected or not with HIV-1. We found a marked global alteration of gene expression following infection, with an overall trend toward induction of genes, indicating widespread modification of the host biology. Annotation and pathway analysis of the most deregulated genes showed that viral infection produces a down-regulation of genes associated with the nucleolus, in particular those implicated in regulating the different steps of ribosome biogenesis, such as ribosomal RNA (rRNA) transcription, pre-rRNA processing, and ribosome maturation. The impact of HIV-1 infection on genes involved in ribosome biogenesis was further validated in primary CD4+ T cells. Moreover, we provided evidence by Northern Blot experiments, that host pre-rRNA processing in Jurkat cells might be perturbed during HIV-1 infection, thus strengthening the hypothesis of a crosstalk between nucleolar functions and viral pathogenesis.


Subject(s)
Down-Regulation/genetics , HIV Infections/genetics , HIV Infections/immunology , HIV-1/physiology , Ribosomes/metabolism , CD4-Positive T-Lymphocytes/immunology , Gene Expression Profiling , Humans , Jurkat Cells , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/genetics , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL
...