Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Mol Sci ; 24(14)2023 Jul 09.
Article in English | MEDLINE | ID: mdl-37511023

ABSTRACT

1,3,4-Oxadiazole derivatives are among the most studied anticancer drugs. Previous studies have analyzed the action of different 1,3,4-oxadiazole derivatives and their effects on cancer cells. This study investigated the characterization of two new compounds named 6 and 14 on HeLa and PC-3 cancer cell lines. Based on the previously obtained IC50, cell cycle effects were monitored by flow cytometry. RNA sequencing (RNAseq) was performed to identify differentially expressed genes, followed by functional annotation using gene ontology (GO), KEGG signaling pathway enrichment, and protein-protein interaction (PPI) network analyses. The tubulin polymerization assay was used to analyze the interaction of both compounds with tubulin. The results showed that 6 and 14 strongly inhibited the proliferation of cancer cells by arresting them in the G2/M phase of the cell cycle. Transcriptome analysis showed that exposure of HeLa and PC-3 cells to the compounds caused a marked reprograming of gene expression. Functional enrichment analysis indicated that differentially expressed genes were significantly enriched throughout the cell cycle and cancer-related biological processes. Furthermore, PPI network, hub gene, and CMap analyses revealed that compounds 14 and 6 shared target genes with established microtubule inhibitors, indicating points of similarity between the two molecules and microtubule inhibitors in terms of the mechanism of action. They were also able to influence the polymerization process of tubulin, suggesting the potential of these new compounds to be used as efficient chemotherapeutic agents.


Subject(s)
Antineoplastic Agents , Chalcogens , Neoplasms , Humans , Tubulin/genetics , Tubulin/metabolism , Structure-Activity Relationship , Cell Proliferation , Antineoplastic Agents/pharmacology , HeLa Cells , Tubulin Modulators/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor
2.
Semin Cell Dev Biol ; 97: 26-37, 2020 01.
Article in English | MEDLINE | ID: mdl-31002867

ABSTRACT

Heart failure of ischemic origin is caused by the presence of a large scar resulting from an acute myocardial infarction. Acute myocardial infarction generally occurs when blood supply to the heart is blocked. Regenerative strategies that limit infarct injury would be able to prevent adverse post-ischemic remodelling and maintain the structural support necessary for effective cardiomyocyte contraction. Our understanding of endogenous cardiac regeneration and its biology has exposed a variety of targets for therapeutic approaches, such as non-coding RNAs, DNA methylation, histone modifications, direct cardiac reprogramming, cell transplantation, stimulation of resident cardiomyocytes, proliferation, and inhibition of cardiomyocyte death. In this review, we address the epigenetic mechanisms underlying these strategies and the use of therapeutic epigenetic molecules or epidrugs.


Subject(s)
Cellular Reprogramming/genetics , Epigenesis, Genetic/genetics , Myocytes, Cardiac/drug effects , Regeneration/drug effects , Humans
3.
Int J Cancer ; 139(5): 1106-16, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27074337

ABSTRACT

Squamous cell carcinomas have a range of histopathological manifestations. The parameters that determine this clinically observed heterogeneity are not fully understood. Here, we report the generation of a cell culture model that reflects part of this heterogeneity. We have used the catalytic subunit of human telomerase hTERT and large T to immortalize primary UV-unexposed keratinocytes. Then, mutant HRAS G12V has been introduced to transform these immortal keratinocytes. When injected into immunosuppressed mice, transformed cells grew as xenografts with distinct histopathological characteristics. We observed three major tissue architectures: solid, sarcomatoid and cystic growth types, which were primarily composed of pleomorphic and basaloid cells but in some cases displayed focal apocrine differentiation. We demonstrate that the cells generated represent different stages of skin cancerogenesis and as such can be used to identify novel tumor-promoting alterations such as the overexpression of the PADI2 oncogene in solid-type SCC. Importantly, the cultured cells maintain the characteristics from the xenograft they were derived from while being amenable to manipulation and analysis. The availability of cell lines representing different clinical manifestations opens a new tool to study the stochastic and deterministic factors that cause case-to-case heterogeneity despite departing from the same set of oncogenes and the same genetic background.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Mutation , Phenotype , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression , Genetic Association Studies , Heterografts , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice
4.
J Immunol ; 186(4): 2299-308, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21239708

ABSTRACT

3' Repair exonuclease (Trex1) is the most abundant mammalian 3' → 5' DNA exonuclease with specificity for ssDNA. Trex1 deficiency has been linked to the development of autoimmune disease in mice and humans, causing Aicardi-Goutières syndrome in the latter. In addition, polymorphisms in Trex1 are associated with systemic lupus erythematosus. On the basis of all these observations, it has been hypothesized that Trex1 acts by digesting an endogenous DNA substrate. In this study, we report that Trex1 is regulated by IFN-γ during the activation of primary macrophages. IFN-γ upregulates Trex1 with the time course of an early gene, and this induction occurs at the transcription level. The half-life of mRNA is relatively short (half-life of 70 min). The coding sequence of Trex1 has only one exon and an intron of 260 bp in the promoter in the nontranslated mRNA. Three transcription start sites were detected, the one at -580 bp being the most important. In transient transfection experiments using the Trex1 promoter, we have found that two IFN-γ activation site boxes, as well as an adaptor protein complex 1 box, were required for the IFN-γ-dependent induction. By using EMSA assays and chromatin immune precipitation assays, we determined that STAT1 binds to the IFN-γ activation site boxes. The requirement of STAT1 for Trex1 induction was confirmed using macrophages from Stat1 knockout mice. We also establish that c-Jun protein, but not c-Fos, jun-B, or CREB, bound to the adaptor protein complex 1 box. Therefore, our results indicate that IFN-γ induces the expression of the Trex1 exonuclease through STAT1 and c-Jun.


Subject(s)
Exodeoxyribonucleases/biosynthesis , Exodeoxyribonucleases/genetics , Gene Expression Regulation/immunology , Interferon-gamma/physiology , Macrophage Activation/immunology , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/physiology , Animals , Cell Line , Cells, Cultured , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Macrophage Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Promoter Regions, Genetic/immunology , Transcription Initiation Site , Up-Regulation/genetics , Up-Regulation/immunology
5.
Nat Genet ; 36(7): 738-43, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15208625

ABSTRACT

During skeletal myogenesis, genomic reprogramming toward terminal differentiation is achieved by recruiting chromatin-modifying enzymes to muscle-specific loci. The relative contribution of extracellular signaling cascades in targeting these enzymes to individual genes is unknown. Here we show that the differentiation-activated p38 pathway targets the SWI-SNF chromatin-remodeling complex to myogenic loci. Upon differentiation, p38 kinases were recruited to the chromatin of muscle-regulatory elements. Blockade of p38 alpha/beta repressed the transcription of muscle genes by preventing recruitment of the SWI-SNF complex at these elements without affecting chromatin binding of muscle-regulatory factors and acetyltransferases. The SWI-SNF subunit BAF60 could be phosphorylated by p38 alpha-beta in vitro, and forced activation of p38 alpha/beta in myoblasts by expression of a constitutively active MKK6 (refs. 5,6,7) promoted unscheduled SWI-SNF recruitment to the myogenin promoter. Conversely, inactivation of SWI-SNF enzymatic subunits abrogated MKK6-dependent induction of muscle gene expression. These results identify an unexpected function of differentiation-activated p38 in converting external cues into chromatin modifications at discrete loci, by selectively targeting SWI-SNF to muscle-regulatory elements.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscles/metabolism , Transcription Factors/metabolism , Cell Line , Imidazoles/pharmacology , Muscles/cytology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases
6.
Chem Biol Interact ; 312: 108813, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31494105

ABSTRACT

Rhabdomyosarcoma (RMS) is a pediatric tumor, which arises from muscle precursor cells. Recently, it has been demonstrated that Hippo Pathway (Hpo), a pathway that regulates several physiological and biological features, is involved in RMS tumorigenesis. For instance, an upregulation of the Hpo downstream effector Yes-Associated Protein 1 (YAP) leads to the development of embryonal rhabdomyosarcoma (eRMS) in murine activated muscle satellite cells. On the other hand, the YAP paralog transcriptional co-activator with PDZ-binding motif (TAZ) is overexpressed in alveolar rhabdomyosarcoma (aRMS) patients with poor survival. YAP and TAZ exhibit both cytoplasmic and nuclear functions. In the nucleus, YAP binds TEADs (TEA domain family members) factors and together they constitute a complex that is able either to activate the transcription of several genes such as MYC, Tbx5 and PAX8 or to maintain the stability of others like p73. Due to the key role of YAP and TAZ in cancer, the identification and/or development of new compounds able to block their activity might be an effective antineoplastic strategy. Verteporfin (VP) is a molecule able to stop the formation of YAP/TEAD complex in the nucleus. The aim of this study is to evaluate the action of VP on RMS cell lines. This work shows that VP has an anti-proliferative activity on all RMS cell lines analyzed. Depending on RMS cell lines, VP affects cell cycle differently. Moreover, VP is able to decrease YAP protein levels, and to induce the activation of apoptosis mechanism through the cleavage of PARP-1. In addition, Annexin V assay showed the activation of apoptosis and necrosis after VP treatment. In summary, the ability of VP to disrupt RMS cell proliferation could be a novel and valuable strategy to improve the therapeutic approaches in treating rhabdomyosarcoma.


Subject(s)
Cell Proliferation/drug effects , Verteporfin/pharmacology , Acyltransferases , Cell Cycle Proteins , Cell Line, Tumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Nuclear Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Rhabdomyosarcoma, Alveolar/pathology , Rhabdomyosarcoma, Embryonal/metabolism , Rhabdomyosarcoma, Embryonal/pathology , Transcription Factors/metabolism
7.
Mol Cancer Res ; 14(9): 841-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27280713

ABSTRACT

UNLABELLED: Peptidyl arginine deiminases (PADI) are a family of enzymes that catalyze the poorly understood posttranslational modification converting arginine residues into citrullines. In this study, the role of PADIs in the pathogenesis of colorectal cancer was investigated. Specifically, RNA expression was analyzed and its association with survival in a cohort of 98 colorectal cancer patient specimens with matched adjacent mucosa and 50 controls from donors without cancer. Key results were validated in an independent collection of tumors with matched adjacent mucosa and by mining of a publicly available expression data set. Protein expression was analyzed by immunoblotting for cell lines or IHC for patient specimens that further included 24 cases of adenocarcinoma with adjacent dysplasia and 11 cases of active ulcerative colitis. The data indicate that PADI2 is the dominantly expressed PADI enzyme in colon mucosa and is upregulated during differentiation. PADI2 expression is low or absent in colorectal cancer. Frequently, this occurs already at the stage of low-grade dysplasia. Mucosal PADI2 expression is also low in ulcerative colitis. The expression level of PADI2 in tumor and adjacent mucosa correlates with differential survival: low levels associate with poor prognosis. IMPLICATIONS: Downregulation of PADI2 is an early event in the pathogenesis of colorectal cancer associated with poor prognosis and points toward a possible role of citrullination in modulating tumor cells and their microenvironment. Mol Cancer Res; 14(9); 841-8. ©2016 AACR.


Subject(s)
Colorectal Neoplasms/enzymology , Hydrolases/biosynthesis , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinogenesis , Case-Control Studies , Cell Differentiation/physiology , Cell Line, Tumor , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Down-Regulation , Enterocytes/enzymology , Enterocytes/pathology , HCT116 Cells , HT29 Cells , Humans , Hydrolases/genetics , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Prognosis , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases
8.
Oncogene ; 23(12): 2177-87, 2004 Mar 18.
Article in English | MEDLINE | ID: mdl-14968110

ABSTRACT

The balance between acetylation and deacetylation of histone and nonhistone proteins controls gene expression in a variety of cellular processes, with transcription being activated by acetyltransferases and silenced by deacetylases. We report here the formation and enzymatic characterization of a complex between the acetyltransferase p300 and histone deacetylases. The C/H3 region of p300 was found to co-purify deacetylase activity from nuclear cell extracts. A prototype of class I histone deacetylases, HDAC1, interacts with p300 C/H3 domain in vitro and in vivo. The p300-binding protein E1A competes with HDAC1 for C/H3 binding; and, like E1A, HDAC1 overexpression interferes with either activation of Gal4p300 fusion protein or p300-dependent co-activation of two C/H3-binding proteins, MyoD and p53. The exposure to deacetylase inhibitors could reverse the dominant-negative effect of a C/H3 fragment insulated from the rest of the molecule, on MyoD- and p53-dependent transcription, whereas inhibition by E1A was resistant to trichostatin A. These data support the hypothesis that association between acetyltransferases and deacetylases can control the expression of genes implicated in cellular growth and differentiation, and suggest that the dominant-negative effect of the p300 C/H3 fragment relies on deacetylase recruitment.


Subject(s)
Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Acetyltransferases/chemistry , Adenovirus E1A Proteins/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , HeLa Cells , Histone Deacetylase 1 , Histone Deacetylases/genetics , Humans , Mice , MyoD Protein/genetics , MyoD Protein/metabolism , NIH 3T3 Cells , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Oncotarget ; 6(26): 22375-96, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26090614

ABSTRACT

TREX2 is a 3'-DNA exonuclease specifically expressed in keratinocytes. Here, we investigated the relevance and mechanisms of TREX2 in ultraviolet (UV)-induced skin carcinogenesis. TREX2 expression was up-regulated by chronic UV exposure whereas it was de-regulated or lost in human squamous cell carcinomas (SCCs). Moreover, we identified SNPs in the TREX2 gene that were more frequent in patients with head and neck SCCs than in healthy individuals. In mice, TREX2 deficiency led to enhanced susceptibility to UVB-induced skin carcinogenesis which was preceded by aberrant DNA damage removal and degradation as well as reduced inflammation. Specifically, TREX2 loss diminished the up-regulation of IL12 and IFNγ, key cytokines related to DNA repair and antitumor immunity. In UV-treated keratinocytes, TREX2 promoted DNA repair and passage to late apoptotic stages. Notably, TREX2 was recruited to low-density nuclear chromatin and micronuclei, where it interacted with phosphorylated H2AX histone, which is a critical player in both DNA repair and cell death. Altogether, our data provide new insights in the molecular mechanisms of TREX2 activity and establish cell autonomous and non-cell autonomous functions of TREX2 in the UVB-induced skin response.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Exodeoxyribonucleases/metabolism , Phosphoproteins/metabolism , Skin Neoplasms/enzymology , Ultraviolet Rays/adverse effects , Animals , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , DNA Damage , Exodeoxyribonucleases/genetics , Female , Humans , Keratinocytes/enzymology , Keratinocytes/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Skin Neoplasms/etiology , Skin Neoplasms/pathology
10.
Cell Rep ; 3(3): 661-70, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23478022

ABSTRACT

Direct generation of a homogeneous population of skeletal myoblasts from human embryonic stem cells (hESCs) and formation of three-dimensional contractile structures for disease modeling in vitro are current challenges in regenerative medicine. Previous studies reported on the generation of myoblasts from ESC-derived embryoid bodies (EB), but not from undifferentiated ESCs, indicating the requirement for mesodermal transition to promote skeletal myogenesis. Here, we show that selective absence of the SWI/SNF component BAF60C (encoded by SMARCD3) confers on hESCs resistance to MyoD-mediated activation of skeletal myogenesis. Forced expression of BAF60C enables MyoD to directly activate skeletal myogenesis in hESCs by instructing MyoD positioning and allowing chromatin remodeling at target genes. BAF60C/MyoD-expressing hESCs are epigenetically committed myogenic progenitors, which bypass the mesodermal requirement and, when cultured as floating clusters, give rise to contractile three-dimensional myospheres composed of skeletal myotubes. These results identify BAF60C as a key epigenetic determinant of hESC commitment to the myogenic lineage and establish the molecular basis for the generation of hESC-derived myospheres exploitable for "disease in a dish" models of muscular physiology and dysfunction.


Subject(s)
Cellular Reprogramming , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Muscle Fibers, Skeletal/cytology , Cell Line , Cell Lineage , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Embryonic Stem Cells/cytology , Humans , Mesoderm/cytology , Muscle Contraction , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , MyoD Protein/genetics , MyoD Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Bioarchitecture ; 2(3): 104-109, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22880151

ABSTRACT

Chromatin remodeling by the SWI/SNF complex is required to activate the transcription of myogenic-specific genes. Our work addressed the details of how SWI/SNF is recruited to myogenic regulatory regions in response to differentiation signals. Surprisingly, the muscle determination factor MyoD and the SWI/SNF subunit BAF60c form a complex on the regulatory elements of MyoD-targeted genes in myogenic precursor cells. This Brg1-devoid MyoD-BAF60c complex flags the chromatin of myogenic-differentiation genes before transcription is activated. On differentiation, BAF60c phosphorylation on a conserved threonine by p38 α kinase promotes the incorporation of MyoD-BAF60c into a Brg1-based SWI/SNF complex, which remodels the chromatin and activates transcription of MyoD-target genes. Downregulation of BAF60c expression prevents MyoD access to the chromatin and the proper loading of an active myogenic transcriptosome preventing the expression of hundreds of myogenic genes. Our data support an unprecedented two-step model by which (1) pre-assembled BAF60c-MyoD complex poises the chromatin of myogenic genes for rapid transcription; (2) chromatin-bound BAF60c "senses" the myogenic differentiation cues and recruits an active SWI/SNF complex to remodel the chromatin allowing transcriptional activation.

12.
Semin Cell Dev Biol ; 16(4-5): 596-611, 2005.
Article in English | MEDLINE | ID: mdl-16129633

ABSTRACT

Cellular differentiation entails an extensive reprogramming of the genome toward the expression of discrete subsets of genes, which establish the tissue-specific phenotype. This program is achieved by epigenetic marks of the chromatin at particular loci, and is regulated by environmental cues, such as soluble factors and cell-to-cell interactions. How the intracellular cascades convert the myriad of external stimuli into the nuclear information necessary to reprogram the genome toward specific responses is a question of biological and medical interest. The elucidation of the signaling converting cues from outside the cells into chromatin modifications at individual promoters holds the promise to unveil the targets for selective pharmacological interventions to modulate gene expression for therapeutic purposes. Enhancing muscle regeneration and preventing muscle breakdown are important goals in the therapy of muscular diseases, cancer-associated cachexia and aging-associated sarcopenia. We will summarize the recent progress of our knowledge of the regulation of gene expression by intracellular cascades elicited by external cues during skeletal myogenesis. And will illustrate the potential importance of targeting the chromatin signaling in regenerative medicine--e.g. to boost muscle regeneration.


Subject(s)
Chromatin/genetics , Gene Expression Regulation/drug effects , Gene Targeting/methods , Muscle Development/drug effects , Muscle Development/genetics , Muscle, Skeletal/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chromatin/drug effects , Chromatin/physiology , Humans , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology
13.
Mol Cell ; 20(6): 891-903, 2005 Dec 22.
Article in English | MEDLINE | ID: mdl-16364914

ABSTRACT

Transcriptional and posttranscriptional processes regulate expression of genetic networks in response to environmental cues. The extracellular signal-activated p38 MAP kinase (p38) pathway plays a fundamental role in conversion of myoblasts to differentiated myocytes. p38 phosphorylates specific transcription factors and chromatin-associated proteins promoting assembly of the myogenic transcriptome. Here, we demonstrate that p38 alpha and beta isoforms also control muscle-gene expression posttranscriptionally, by stabilizing critical myogenic transcripts. KSRP, an important factor for AU-rich element (ARE)-directed mRNA decay, undergoes p38-dependent phosphorylation during muscle differentiation. KSRP phosphorylated by p38 displays compromised binding to ARE-containing transcripts and fails to promote their rapid decay, although it retains the ability to interact with the mRNA degradation machinery. Overexpression of KSRP selectively impairs induction of ARE-containing early myogenic transcripts, without affecting p38-mediated transcriptional responses. Our results uncover an unanticipated role for KSRP in establishing a biochemical link between differentiation-activated p38 signaling and turnover of myogenic mRNAs.


Subject(s)
Myoblasts/physiology , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Gene Expression Regulation , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Myoblasts/cytology , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trans-Activators/genetics , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL