Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
2.
Front Cell Infect Microbiol ; 14: 1308362, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476167

RESUMEN

Infectious peritonitis is a leading cause of peritoneal functional impairment and a primary factor for therapy discontinuation in peritoneal dialysis (PD) patients. Although bacterial infections are a common cause of peritonitis episodes, emerging evidence suggests a role for viral pathogens. Toll-like receptors (TLRs) specifically recognize conserved pathogen-associated molecular patterns (PAMPs) from bacteria, viruses, and fungi, thereby orchestrating the ensuing inflammatory/immune responses. Among TLRs, TLR3 recognizes viral dsRNA and triggers antiviral response cascades upon activation. Epigenetic regulation, mediated by histone deacetylase (HDAC), has been demonstrated to control several cellular functions in response to various extracellular stimuli. Employing epigenetic target modulators, such as epidrugs, is a current therapeutic option in several cancers and holds promise in treating viral diseases. This study aims to elucidate the impact of TLR3 stimulation on the plasticity of human mesothelial cells (MCs) in PD patients and to investigate the effects of HDAC1-3 inhibition. Treatment of MCs from PD patients with the TLR3 agonist polyinosinic:polycytidylic acid (Poly(I:C)), led to the acquisition of a bona fide mesothelial-to-mesenchymal transition (MMT) characterized by the upregulation of mesenchymal genes and loss of epithelial-like features. Moreover, Poly(I:C) modulated the expression of several inflammatory cytokines and chemokines. A quantitative proteomic analysis of MCs treated with MS-275, an HDAC1-3 inhibitor, unveiled altered expression of several proteins, including inflammatory cytokines/chemokines and interferon-stimulated genes (ISGs). Treatment with MS-275 facilitated MMT reversal and inhibited the interferon signature, which was associated with reduced STAT1 phosphorylation. However, the modulation of inflammatory cytokine/chemokine production was not univocal, as IL-6 and CXCL8 were augmented while TNF-α and CXCL10 were decreased. Collectively, our findings underline the significance of viral infections in acquiring a mesenchymal-like phenotype by MCs and the potential consequences of virus-associated peritonitis episodes for PD patients. The observed promotion of MMT reversal and interferon response inhibition by an HDAC1-3 inhibitor, albeit without a general impact on inflammatory cytokine production, has translational implications deserving further analysis.


Asunto(s)
Benzamidas , Interferón Tipo I , Peritonitis , Piridinas , Virosis , Humanos , Interferón Tipo I/metabolismo , Receptor Toll-Like 3/metabolismo , Epigénesis Genética , Proteómica , Citocinas/metabolismo , Quimiocinas/metabolismo , Poli I-C/farmacología , Receptores Toll-Like/metabolismo , Virosis/genética , Fenotipo , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo
3.
J Exp Clin Cancer Res ; 43(1): 27, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38254102

RESUMEN

BACKGROUND: Peritoneal metastasis, which accounts for 85% of all epithelial ovarian carcinoma (EOC) metastases, is a multistep process that requires the establishment of adhesive interactions between cancer cells and the peritoneal membrane. Interrelations between EOC and the mesothelial stroma are critical to facilitate the metastatic process. No data is available so far on the impact of histone acetylation/deacetylation, a potentially relevant mechanism governing EOC metastasis, on mesothelial cells (MCs)-mediated adhesion. METHODS: Static adhesion and peritoneal clearance experiments were performed pretreating mesenchymal-like MCs and platinum-sensitive/resistant EOC cell lines with MS-275-a Histone deacetylase (HDAC)1-3 pharmacological inhibitor currently used in combination trials. Results were acquired by confocal microscopy and were analyzed with an automated Opera software. The role of HDAC1/2 was validated by genetic silencing. The role of α4-, α5-α1 Integrins and Fibronectin-1 was validated using specific monoclonal antibodies. Quantitative proteomic analysis was performed on primary MCs pretreated with MS-275. Decellularized matrices were generated from either MS-275-exposed or untreated cells to study Fibronectin-1 extracellular secretion. The effect of MS-275 on ß1 integrin activity was assessed using specific monoclonal antibodies. The role of Talin-1 in MCs/EOC adhesion was analyzed by genetic silencing. Talin-1 ectopic expression was validated as a rescue tool from MS-275-induced phenotype. The in vivo effect of MS-275-induced MC remodeling was validated in a mouse model of peritoneal EOC dissemination. RESULTS: Treatment of MCs with non-cytotoxic concentrations of MS-275 caused a consistent reduction of EOC adhesion. Proteomic analysis revealed several pathways altered upon MC treatment with MS-275, including ECM deposition/remodeling, adhesion receptors and actin cytoskeleton regulators. HDAC1/2 inhibition hampered actin cytoskeleton polymerization by downregulating actin regulators including Talin-1, impairing ß1 integrin activation, and leading to abnormal extracellular secretion and distribution of Fibronectin-1. Talin-1 ectopic expression rescued EOC adhesion to MS-275-treated MCs. In an experimental mouse model of metastatic EOC, MS-275 limited tumor invasion, Fibronectin-1 secretion and the sub-mesothelial accumulation of MC-derived carcinoma-associated fibroblasts. CONCLUSION: Our study unveils a direct impact of HDAC-1/2 in the regulation of MC/EOC adhesion and highlights the regulation of MC plasticity by epigenetic inhibition as a potential target for therapeutic intervention in EOC peritoneal metastasis.


Asunto(s)
Benzamidas , Carcinoma Epitelial de Ovario , Adhesión Celular , Histona Desacetilasa 1 , Histona Desacetilasa 2 , Neoplasias Ováricas , Neoplasias Peritoneales , Animales , Femenino , Humanos , Ratones , Citoesqueleto de Actina/metabolismo , Anticuerpos Monoclonales , Carcinoma Epitelial de Ovario/metabolismo , Epitelio , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas , Histona Desacetilasa 1/metabolismo , Integrina alfa5 , Integrina beta1/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/metabolismo , Proteómica , Piridinas , Talina/genética , Talina/metabolismo , Histona Desacetilasa 2/metabolismo , Adhesión Celular/genética
4.
J Med Chem ; 66(14): 9622-9641, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37439550

RESUMEN

The mitochondrial SIRT3 modulates several biological pathways such as cancer, metabolism, and hypoxia-related diseases. Recently, we discovered new 1,4-dihydropyridines, compounds 2 and 3, the latter being a SIRT3-specific activator. In the present work, a novel 2- and 3-related small series of compounds have been developed, with 3c displaying the strongest SIRT3 binding and activation, with a KD of 29 µM and 387% of enzyme activation. Differently, 3d was the best in enhancing glutamate dehydrogenase activity and deacetylating K68- and K122-acMnSOD in triple-negative MDA-MB-231 breast cancer cells. Tested in CAL-62 thyroid cancer and MDA-MB-231 cells, 3d displayed the strongest time- and dose-dependent reduction of cell viability and clonogenicity at a single-digit micromolar level, along with cell death, in both normoxia and hypoxia conditions. Moreover, 3d downregulated not only hypoxia-induced factors, such as HIF-1α, EPAS-1, and CA-IX, but also epithelial-mesenchymal transition master regulators and extracellular matrix components such as SNAIL1, ZEB1, SLUG, COL1A2, MMP2, and MMP9, markedly hampering MDA-MB-231 cell migration.


Asunto(s)
Neoplasias , Sirtuina 3 , Humanos , Supervivencia Celular , Línea Celular Tumoral , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia
6.
Cell Biosci ; 13(1): 44, 2023 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-36871010

RESUMEN

In recent years, progress in nanotechnology provided new tools to treat cancer more effectively. Advances in biomaterials tailored for drug delivery have the potential to overcome the limited selectivity and side effects frequently associated with traditional therapeutic agents. While autophagy is pivotal in determining cell fate and adaptation to different challenges, and despite the fact that it is frequently dysregulated in cancer, antitumor therapeutic strategies leveraging on or targeting this process are scarce. This is due to many reasons, including the very contextual effects of autophagy in cancer, low bioavailability and non-targeted delivery of existing autophagy modulatory compounds. Conjugating the versatile characteristics of nanoparticles with autophagy modulators may render these drugs safer and more effective for cancer treatment. Here, we review current standing questions on the biology of autophagy in tumor progression, and precursory studies and the state-of-the-art in harnessing nanomaterials science to enhance the specificity and therapeutic potential of autophagy modulators.

7.
Cell Death Dis ; 14(1): 32, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36650140

RESUMEN

YES-associated protein (YAP) is a transcriptional cofactor with a key role in the regulation of several physio-pathological cellular processes, by integrating multiple cell autonomous and microenvironmental cues. YAP is the main downstream effector of the Hippo pathway, a tumor-suppressive signaling able to transduce several extracellular signals. The Hippo pathway acts restraining YAP activity, since its activation induces YAP phosphorylation and cytoplasmic sequestration. However, recent observations indicate that YAP activity can be also modulated by Hippo independent/integrating pathways, still largely unexplored. In this study, we demonstrated the role of the extracellular signal-regulated kinase 5 (ERK5)/mitogen-activated protein kinase in the regulation of YAP activity. By means of ERK5 inhibition/silencing and overexpression experiments, and by using as model liver stem cells, hepatocytes, and hepatocellular carcinoma (HCC) cell lines, we provided evidence that ERK5 is required for YAP-dependent gene expression. Mechanistically, ERK5 controls the recruitment of YAP on promoters of target genes and its physical interaction with the transcriptional partner TEAD; moreover, it mediates the YAP activation occurring in cell adhesion, migration, and TGFß-induced EMT of liver cells. Furthermore, we demonstrated that ERK5 signaling modulates YAP activity in a LATS1/2-independent manner. Therefore, our observations identify ERK5 as a novel upstream Hippo-independent regulator of YAP activity, thus unveiling a new target for therapeutic approaches aimed at interfering with its function.


Asunto(s)
Hepatocitos , Proteína Quinasa 7 Activada por Mitógenos , Proteínas Señalizadoras YAP , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Neoplasias Hepáticas/patología , Proteína Quinasa 7 Activada por Mitógenos/genética , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo , Hepatocitos/metabolismo , Células Madre
8.
Eur J Med Chem ; 247: 115022, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36549114

RESUMEN

After over 30 years of research, the development of HDAC inhibitors led to five FDA/Chinese FDA-approved drugs and many others under clinical or preclinical investigation to treat cancer and non-cancer diseases. Herein, based on our recent development of pyridine-based isomers as HDAC inhibitors, we report a series of novel 5-acylamino-2-pyridylacrylic- and -picolinic hydroxamates and 2'-aminoanilides 5-8 as anticancer agents. The hydroxamate 5d proved to be quite HDAC3/6-selective exhibiting IC50 values of 80 and 11 nM, respectively, whereas the congener 5e behaved as inhibitor of HDAC1-3, -6, -8, and -10 (class I/IIb-selective inhibitor) at nanomolar level. Compound 5e provided a huge antiproliferative activity (nanomolar IC50 values) against both haematological and solid cancer cell lines. In leukaemia U937 cells, the hydroxamate 5d and the 2'-aminoanilide 8f induced remarkable cell death after 48 h, with 76% and 100% pre-G1 phase arrest, respectively, showing a stronger effect with respect to SAHA and MS-275 used as reference compounds. In U937 cells, the highest dose- and time-dependent cytodifferentiation was obtained by the 2'-aminoanilide 8d (up to 35% of CD11c positive/propidium iodide negative cells at 5 µM for 48 h). The same 8d and the hydroxamates 5d and 5e were the most effective in inducing p21 protein expression in the same cell line. Mechanistically, 5d, 5e, 8d and 8f increased mRNA expression of p21, BAX and BAK, downregulated cyclin D1 and BCL-2 and modulated pro- and anti-apoptotic microRNAs towards apoptosis induction. Finally, 5e strongly arrested proliferation in nine different haematological cancer cell lines, with dual-digit nanomolar potency towards MV4-11, Kasumi-1, and NB4, being more potent than mocetinostat, used as reference drug.


Asunto(s)
Antineoplásicos , MicroARNs , Neoplasias , Humanos , Inhibidores de Histona Desacetilasas/farmacología , Línea Celular Tumoral , Proliferación Celular , Antineoplásicos/farmacología , Ácidos Hidroxámicos/farmacología , Apoptosis , Piridinas/farmacología , Histona Desacetilasa 1
9.
Front Cell Infect Microbiol ; 13: 1257683, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38162580

RESUMEN

Background: Despite the significant progress achieved in understanding the pathology and clinical management of SARS-CoV-2 infection, still pathogenic and clinical issues need to be clarified. Treatment with modulators of epigenetic targets, i.e., epidrugs, is a current therapeutic option in several cancers and could represent an approach in the therapy of viral diseases. Results: Aim of this study was the analysis of the role of histone deacetylase (HDAC) inhibition in the modulation of SARS-CoV-2 infection of mesothelial cells (MCs).MeT5A cells, a pleura MC line, were pre-treated with different specific class I and IIb HDAC inhibitors. Unexpectedly, treatment with HDAC1-3 inhibitors significantly increased ACE2/TMPRSS2 expression, suggesting a role in favoring SARS-CoV-2 infection. We focused our analysis on the most potent ACE2/TMPRSS2 inducer among the inhibitors analysed, MS-275, a HDAC1-3 inhibitor. ACE2/TMPRSS2 expression was validated by Western Blot (WB) and immunofluorescence. The involvement of HDAC inhibition in receptor induction was confirmed by HDAC1/HDAC2 silencing. In accordance to the ACE2/TMPRSS2 expression data, MS-275 increased SARS-CoV-2 replication and virus propagation in Vero E6 cells.Notably, MS-275 was able to increase ACE2/TMPRSS2 expression and SARS-CoV-2 production, although to a lesser extent, also in the lung adenocarcinoma cell line Calu-3 cells.Mechanistically, treatment with MS-275 increased H3 and H4 histone acetylation at ACE2/TMPRSS2 promoters, increasing their transcription. Conclusion: This study highlights a previously unrecognized effect of HDAC1-3 inhibition in increasing SARS-CoV-2 cell entry, replication and productive infection correlating with increased expression of ACE2 and TMPRSS2. These data, while adding basic insight into COVID-19 pathogenesis, warn for the use of HDAC inhibitors in SARS-CoV-2 patients.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/metabolismo , Pulmón/metabolismo , Células Epiteliales , Histona Desacetilasa 1/metabolismo
10.
Cell Death Dis ; 13(11): 965, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396626

RESUMEN

Histone acetylation/deacetylation play an essential role in modifying chromatin structure and in regulating cell plasticity in eukaryotic cells. Therefore, histone deacetylase (HDAC) pharmacological inhibitors are promising tools in the therapy of fibrotic diseases and in cancer. Peritoneal fibrosis is a pathological process characterized by many cellular and molecular alterations, including the acquisition of invasive/pro-fibrotic abilities by mesothelial cells (MCs) through induction of mesothelial to mesenchymal transition (MMT). The aim of this study was to characterize the molecular mechanism of the antifibrotic role of HDAC1 inhibition. Specifically, treatment with MS-275, an HDAC1-3 inhibitor previously known to promote MMT reversal, induced the expression of several TGFBRI mRNA-targeting miRNAs. Among them, miR-769-5p ectopic expression was sufficient to promote MMT reversal and to limit MC migration and invasion, whereas miR-769-5p silencing further enhanced mesenchymal gene expression. These results were confirmed by HDAC1 genetic silencing. Interestingly, miR-769-5p silencing maintained mesenchymal features despite HDAC1 inhibition, thus indicating that it is necessary to drive MMT reversal induced by HDAC1 inhibition. Besides TGFBRI, miR-769-5p was demonstrated to target SMAD2/3 and PAI-1 expression directly. When analyzing molecular mechanisms underlying miR-769-5p expression, we found that the transcription factor Wilms' tumor 1 (WT1), a master gene controlling MC development, binds to the miR-769-5p promoter favoring its expression. Interestingly, both WT1 expression and binding to miR-769-5p promoter were increased by HDAC1 inhibition and attenuated by TGFß1 treatment. Finally, we explored the significance of these observations in the cell-to-cell communication: we evaluated the ability of miR-769-5p to be loaded into extracellular vesicles (EVs) and to promote MMT reversal in recipient mesenchymal-like MCs. Treatment of fibrotic MCs with EVs isolated from miR-769-5p over-expressing MCs promoted the down-regulation of specific mesenchymal targets and the reacquisition of an epithelial-like morphology. In conclusion, we highlighted an HDAC1-WT1-miR-769-5p axis potentially relevant for therapies aimed at counteracting organ fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal , MicroARNs , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Epitelio/metabolismo , MicroARNs/metabolismo
11.
Oncoscience ; 9: 49-51, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36110328

RESUMEN

Long non-coding RNAs (lncRNAs) exert central pathophysiological roles through the regulation of gene expression both at transcriptional and post-transcriptional levels. The characterization of lncRNAs' interactome is disclosing several new mechanisms that control disease onset and progression thus opening the way to the development of new pioneering therapeutic approaches. Regarding the lncRNA HOTAIR, found upregulated in several cancers and in liver fibrosis, it has been proved as a potential therapeutic target. HOTAIR acts as a ceRNA for several miRNAs and it directly interacts with chromatin remodelling complexes (e.g. PRC2 and LSD1/NuRD complexes). In this regard, we recently reported the transcription factor SNAIL-mediated recruitment of HOTAIR/PRC2 complex on specific chromatin sites causing epithelial genes' repression through epigenetic chromatin modifications. Conversely, HOTAIR is repressed by the liver-enriched transcriptional factor HNF4a that binds to both HOTAIR promoter and distant enhancer and impairs the formation of a chromatin loop between these genomic regions. In a therapeutic perspective, we design and validated the first example of a dominant negative lncRNA molecule (HOTAIR-sbid) that covers the HOTAIR portion involved in the interaction with SNAIL while is devoid of the domain of interaction with EZH2. Functionally, HOTAIR-sbid expression impairs SNAIL/EZH2/endogenous HOTAIR interaction; thus, PRC2 complex is not recruited on SNAIL-target chromatin sites (i.e. epithelial genes' promoters). Accordingly, the cells rescue an epithelial phenotype, reduce EMT and, in turn, migratory, invasive and anchorage independent growth abilities. This approach promises high level of specificity and limited off-target effects. Future investigations should enhance RNAs' stability and should design strategies for the delivery of these molecules to specific target cells.

13.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35055098

RESUMEN

Heterogeneous nuclear ribonucleoproteins (hnRNPs) control gene expression by acting at multiple levels and are often deregulated in epithelial tumors; however, their roles in the fine regulation of cellular reprogramming, specifically in epithelial-mesenchymal transition (EMT), remain largely unknown. Here, we focused on the hnRNP-Q (also known as SYNCRIP), showing by molecular analysis that in hepatocytes it acts as a "mesenchymal" gene, being induced by TGFß and modulating the EMT. SYNCRIP silencing limits the induction of the mesenchymal program and maintains the epithelial phenotype. Notably, in HCC invasive cells, SYNCRIP knockdown induces a mesenchymal-epithelial transition (MET), negatively regulating their mesenchymal phenotype and significantly impairing their migratory capacity. In exploring possible molecular mechanisms underlying these observations, we identified a set of miRNAs (i.e., miR-181-a1-3p, miR-181-b1-3p, miR-122-5p, miR-200a-5p, and miR-let7g-5p), previously shown to exert pro- or anti-EMT activities, significantly impacted by SYNCRIP interference during EMT/MET dynamics and gathered insights, suggesting the possible involvement of this RNA binding protein in their transcriptional regulation.


Asunto(s)
Carcinoma Hepatocelular/etiología , Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/genética , Hepatocitos/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Neoplasias Hepáticas/etiología , Animales , Biomarcadores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Susceptibilidad a Enfermedades , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hepatocitos/patología , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , MicroARNs/genética , Fenotipo , Interferencia de ARN , Proteínas de Unión al ARN
14.
Front Mol Biosci ; 8: 752616, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34901152

RESUMEN

Although lung fibrosis has a major impact in COVID-19 disease, its pathogenesis is incompletely understood. In particular, no direct evidence of pleura implication in COVID-19-related fibrotic damage has been reported so far. In this study, the expression of epithelial cytokeratins and Wilms tumor 1 (WT1), specific markers of mesothelial cells (MCs), was analyzed in COVID-19 and unrelated pleura autoptic samples. SARS-CoV-2 replication was analyzed by RT-PCR and confocal microscopy in MeT5A, a pleura MC line. SARS-CoV-2 receptors were analyzed by RT-PCR and western blot. Inflammatory cytokines from the supernatants of SARS-CoV-2-infected MeT5A cells were analysed by Luminex and ELLA assays. Immunohistochemistry of COVID-19 pleura patients highlighted disruption of pleura monolayer and fibrosis of the sub-mesothelial stroma, with the presence of MCs with fibroblastoid morphology in the sub-mesothelial stroma, but no evidence of direct infection in vivo. Interestingly, we found evidence of ACE2 expression in MCs from pleura of COVID-19 patients. In vitro analysis shown that MeT5A cells expressed ACE2, TMPRSS2, ADAM17 and NRP1, plasma membrane receptors implicated in SARS-CoV-2 cell entry and infectivity. Moreover, MeT5A cells sustained SARS-CoV-2 replication and productive infection. Infected MeT5A cells produced interferons, inflammatory cytokines and metalloproteases. Overall, our data highlight the potential role of pleura MCs as promoters of the fibrotic reaction and regulators of the immune response upon SARS-CoV-2 infection.

15.
RNA Biol ; 18(sup1): 75-87, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34224323

RESUMEN

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.


Asunto(s)
Adenosina Desaminasa/metabolismo , Elementos de Nucleótido Esparcido Largo , Edición de ARN , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/genética , Células HEK293 , Células HeLa , Humanos , Proteínas de Unión al ARN/genética
16.
Front Immunol ; 12: 607204, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33854496

RESUMEN

Peritoneal fibrosis is characterized by abnormal production of extracellular matrix proteins leading to progressive thickening of the submesothelial compact zone of the peritoneal membrane. This process may be caused by a number of insults including pathological conditions linked to clinical practice, such as peritoneal dialysis, abdominal surgery, hemoperitoneum, and infectious peritonitis. All these events may cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy. Among the cellular processes implicated in these peritoneal alterations is the generation of myofibroblasts from mesothelial cells and other cellular sources that are central in the induction of fibrosis and in the subsequent functional deterioration of the peritoneal membrane. Myofibroblast generation and activity is actually integrated in a complex network of extracellular signals generated by the various cellular types, including leukocytes, stably residing or recirculating along the peritoneal membrane. Here, the main extracellular factors and the cellular players are described with emphasis on the cross-talk between immune system and cells of the peritoneal stroma. The understanding of cellular and molecular mechanisms underlying fibrosis of the peritoneal membrane has both a basic and a translational relevance, since it may be useful for setup of therapies aimed at counteracting the deterioration as well as restoring the homeostasis of the peritoneal membrane.


Asunto(s)
Comunicación Celular , Susceptibilidad a Enfermedades , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Peritoneo/inmunología , Peritoneo/metabolismo , Células del Estroma/metabolismo , Animales , Biomarcadores , Comunicación Celular/inmunología , Citocinas/metabolismo , Células Epiteliales/metabolismo , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/patología , Peritoneo/patología , Peritonitis/complicaciones , Peritonitis/etiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
17.
Cancer Res ; 81(1): 103-113, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33158813

RESUMEN

HOTAIR is a lncRNA overexpressed in several epithelial cancers and strongly correlated with invasion. This lncRNA was proven a pivotal element of the epithelial-to-mesenchymal transition (EMT), a transdifferentiation process triggering metastasis. Snail, master inducer of EMT, requires HOTAIR to recruit EZH2 on specific epithelial target genes (i.e., HNF4α, E-cadherin, and HNF1α) and cause their repression. Here, we designed a HOTAIR deletion mutant form, named HOTAIR-sbid, including the putative Snail-binding domain but depleted of the EZH2-binding domain. HOTAIR-sbid acted as a dominant negative of the endogenous HOTAIR. In both murine and human tumor cells, HOTAIR-sbid impaired the ability of HOTAIR to bind Snail and, in turn, trigger H3K27me3/EZH2-mediated repression of Snail epithelial target genes. Notably, HOTAIR-sbid expression was proven to reduce cellular motility, invasiveness, anchorage-independent growth, and responsiveness to TGFß-induced EMT. These data provide evidence on a lncRNA-based strategy to effectively impair the function of a master EMT-transcriptional factor. SIGNIFICANCE: This study defines an innovative RNA-based strategy to interfere with a pivotal function of the tumor-related lncRNA HOTAIR, comprising a dominant negative mutant that was computationally designed and that impairs epithelial-to-mesenchymal transition.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/patología , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Hepatocitos/patología , ARN Largo no Codificante/genética , Factores de Transcripción de la Familia Snail/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Mutación , ARN Largo no Codificante/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/genética
18.
Acta bioquím. clín. latinoam ; 54(3): 257-266, set. 2020. graf, tab
Artículo en Español | LILACS | ID: biblio-1130600

RESUMEN

EL HOMA-IR (homeostasis model assessment-insulin-resistance) es un estimador de insulinorresistencia (IR) pero depende de la determinación de insulina. Los índices triglicéridos-glucosa (T-G)-circunferencia de la cintura (CC) (T-G-CC) o triglicéridos-glucosa-índice de masa corporal (TG- IMC) podrían ser sustitutos. Los objetivos de este trabajo consistieron en investigar en personas con riesgo de desarrollar diabetes tipo 2 (DT2): a) los índices T-G, T-G-CC y T-G-IMC como estimadores de HOMA-IR>2,1; b) determinar su poder discriminante. Se realizó un estudio prospectivo en el que se estudiaron 223 individuos ≥45 años con riesgo de desarrollar diabetes tipo 2 (DT2). La relación T-G se calculó como ln [triglicéridos (mg/dL) x glucemia (mg/dL)/2]. La relación T-G-CC y T-G-IMC fue el producto de T-G por CC o IMC. Se utilizó análisis de regresión logística y se calcularon las áreas bajo las curvas ROC (receiver operating characteristic curves) (ABC) para comparar las asociaciones de T-G, T-G-CC y T-G-IMC con HOMA-IR>2,1. Mediante análisis discriminante se evaluó la clasificación de los sujetos entre HOMA-IR>2,1 y HOMA-IR≤2,1. ABC, sensibilidad, especificidad, poder predictivo positivo y negativo para T-G-CC y T-G-IMC fueron mayores que para T-G, con los siguientes valores de corte: T-G=8,75, T-G-CC=821 y T-G-IMC=255. Los odds ratios (OR) para HOMA-IR>2,1, ajustados para confusores, fueron: T-G>8,75, OR: 4,85 (IC 95% 2,73-8,62); T-G-CC>821, OR: 10,41 (IC 95% 5,55-19,53); T-GIMC> 255, OR: 10,41 (IC 95% 5,55-19,53). Con el análisis discriminante T-G>8,75 clasificó correctamente 69,2% individuos con HOMA-IR≤2,1 y 68,3% con HOMA-IR>2,1; T-G-CC y T-G-IMC clasificaron 74,4% y 78,2% respectivamente (p<0,001 en todos los casos). Se concluyó que T-GCC> 821 y T-G-IMC>255 fueron mejores estimadores de HOMA-IR>2,1 que T-G>8,75. Estas son determinaciones simples y accesibles y podrían ser útiles en la práctica clínica y en estudios epidemiológicos.


HOMA-IR ((homeostasis model assessment-insulin-resistance) is a surrogate estimator of insulin resistance (IR) but it depends on insulin determination. Triglyceride-glucose-waist circumference (T-G-WC) or triglyceride-glucose-body mass index (BMI) (T-G-BMI) could be substitutes. The objectives of this work were: to investigate in people at risk of developing type 2 diabetes (T2D): a) T-G, T-G-CC and T-G-BMI as estimators of HOMA-IR>2.1 and b) to determine their discriminating power. A prospective study was conducted studying 223 individuals ≥45 years of age at risk of developing type 2 diabetes (T2D). The T-G ratio was calculated as ln [triglycerides (mg/dL) x glycemia (mg/dL)/2]. The T-G-CC and T-G-BMI ratio was the product of T-G by CC or BMI. Logistic regression analysis was used and the areas under the receiver operating characteristic curves (ROC) curves were calculated to compare the associations of T-G, T-G-CC and T-G-BMI with HOMA-IR>2.1. Using a discriminant analysis, the classification of the subjects between HOMA-IR>2.1 or HOMA-IR≤2.1 was evaluated. AUC, sensitivity, specificity, positive and negative predictive powers for T-G-CC and T-G-BMI were higher than for T-G, with the following cut-off values: TG=8.75, T-G-CC=821 and T-G-BMI=255. Odds ratios (OR) for HOMA-IR>2.1, adjusted for confounders, were: T-G>8.75, OR 4.85 (95% CI 2.73-8.62); T-G-CC>821, OR 10.41 (95% CI 5.55-19.53); T-G-BMI>255, OR 10.41 (95% CI 5.55-19.53). With the discriminant analysis T-G>8.75, 69.2% correctly classified with HOMA-IR≤2.1 and 68.3% with HOMA-IR>2.1; T-G-CC and T-G-BMI correctly classified 74.4% and 78.2% respectively (p <0.001 in all cases). It is concluded that T-G-CC>821 and T-G-BMI>255 were better estimators of HOMA-IR>2.1 than T-G>8.75. T-G-WC and T-G-BMI are simple and reliable determinations and could be useful in clinical practice and epidemiological studies.


O HOMA-IR (homeostasis model assessment-insulin-resistance) e um estimador de resistencia a insulina (RI), mas depende da determinacao da insulina. Triglicerideos-glicose (T-G), circunferencia da cintura (CC) (T-G-CC) ou triglicerideos-glicose-indice de massa corporal (T-G-IMC) poderiam ser substitutos. Os objetivos desse trabalho foram investigar em pessoas com risco de desenvolver diabetes tipo 2 (DT2): a) os indices T-G, T-G-CC e T-G-IMC como estimadores de HOMA-IR> 2,1; b) determinar seu poder discriminante. Um estudo prospectivo foi realizado em 223 pessoas ≥45 anos com risco de desenvolver diabetes tipo 2 (DT2). A razao T-G foi calculada como ln [triglicerideos (mg/dL) x glicemia (mg/dL)/2]. A razao T-G-CC e T-G-IMC foi o produto de T-G por CC ou IMC. A analise de regressao logistica foi utilizada e as areas sob as curvas ROC (receiver operating features) ABC foram calculadas para comparar as associacoes de T-G, T-G-CC e T-G-IMC com HOMA-IR>2.1. Por meio de analise discriminante, avaliou-se a classificacao dos sujeitos entre HOMA-IR>2,1 e HOMA-IR≤2,1. ABC, sensibilidade, especificidade, poder preditivo positivo e negativo para TG-CC e TG-IMC foram maiores que para TG, com os seguintes valores de corte: TG=8,75, TG-CC=821 e TG-IMC=255. Odds Ratios (OR) para HOMA-IR>2,1, ajustados para fatores de confusao, foram: TG>8,75, OR 4,85 (IC95% 2,73-8,62); T-G-CC>821, OR 10,41 (IC 95% 5,55-19,53); T-G-IMC>255, OR 10,41 (IC 95% 5,55-19,53). Com a analise discriminante T-G>8,75, 69,2% foram classificados corretamente com HOMA-IR≤2,1 e 68,3% com HOMA-IR>2,1; T-G-CC e T-G-IMC classificaram 74,4% e 78,2%, respectivamente (p<0,001 em todos os casos). Conclui-se que T-G-CC>821 e TG- IMC>255 foram melhores estimadores de HOMA-IR>2,1 que T-G>8,75. Elas sao determinacoes simples e acessiveis e poderiam ser uteis na pratica clinica e em estudos epidemiologicos.


Asunto(s)
Humanos , Triglicéridos , Poder Psicológico , Estudios Epidemiológicos , Modelos Logísticos , Oportunidad Relativa , Factores de Confusión Epidemiológicos , Curva ROC , Sensibilidad y Especificidad , Clasificación , Área Bajo la Curva , Cortejo , Diabetes Mellitus Tipo 2 , Diabetes Mellitus Tipo 2/complicaciones , Glucosa , Objetivos , Insulina , Personas , Organización y Administración , Asociación , Glucemia , Resistencia a la Insulina , Índice de Masa Corporal , Análisis Discriminante , Riesgo , Análisis de Regresión , Circunferencia de la Cintura
19.
Cell Death Dis ; 11(8): 647, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32811813

RESUMEN

Despite their emerging relevance to fully understand disease pathogenesis, we have as yet a poor understanding as to how biomechanical signals are integrated with specific biochemical pathways to determine cell behaviour. Mesothelial-to-mesenchymal transition (MMT) markers colocalized with TGF-ß1-dependent signaling and yes-associated protein (YAP) activation across biopsies from different pathologies exhibiting peritoneal fibrosis, supporting mechanotransduction as a central driving component of these class of fibrotic lesions and its crosstalk with specific signaling pathways. Transcriptome and proteome profiling of the response of mesothelial cells (MCs) to linear cyclic stretch revealed molecular changes compatible with bona fide MMT, which (i) overlapped with established YAP target gene subsets, and were largely dependent on endogenous TGF-ß1 signaling. Importantly, TGF-ß1 blockade blunts the transcriptional upregulation of these gene signatures, but not the mechanical activation and nuclear translocation of YAP per se. We studied the role therein of caveolin-1 (CAV1), a plasma membrane mechanotransducer. Exposure of CAV1-deficient MCs to cyclic stretch led to a robust upregulation of MMT-related gene programs, which was blunted upon TGF-ß1 inhibition. Conversely, CAV1 depletion enhanced both TGF-ß1 and TGFBRI expression, whereas its re-expression blunted mechanical stretching-induced MMT. CAV1 genetic deficiency exacerbated MMT and adhesion formation in an experimental murine model of peritoneal ischaemic buttons. Taken together, these results support that CAV1-YAP/TAZ fine-tune the fibrotic response through the modulation of MMT, onto which TGF-ß1-dependent signaling coordinately converges. Our findings reveal a cooperation between biomechanical and biochemical signals in the triggering of MMT, representing a novel potential opportunity to intervene mechanically induced disorders coursing with peritoneal fibrosis, such as post-surgical adhesions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Caveolina 1/metabolismo , Fibrosis Peritoneal/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Caveolina 1/fisiología , Caveolinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Diálisis Peritoneal/métodos , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/patología , Peritoneo/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Adherencias Tisulares/metabolismo , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Señalizadoras YAP
20.
Int J Mol Sci ; 21(11)2020 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-32481626

RESUMEN

The reduction of oxygen partial pressure in growing tumors triggers numerous survival strategies driven by the transcription factor complex HIF1 (Hypoxia Inducible Factor-1). Recent evidence revealed that HIF1 promotes rapid and effective phenotypic changes through the induction of non-coding RNAs, whose contribution has not yet been fully described. Here we investigated the role of the hypoxia-induced, long non-coding RNA H19 (lncH19) and its intragenic miRNA (miR-675-5p) into HIF1-Wnt crosstalk. During hypoxic stimulation, colorectal cancer cell lines up-regulated the levels of both the lncH19 and its intragenic miR-675-5p. Loss of expression experiments revealed that miR-675-5p inhibition, in hypoxic cells, hampered ß-catenin nuclear localization and its transcriptional activity, while lncH19 silencing did not induce the same effects. Interestingly, our data revealed that miRNA inhibition in hypoxic cells restored the activity of Glycogen Synthase Kinase 3ß (GSK-3ß) reducing the amount of P-Ser9 kinase, thus unveiling a role of the miR-675-5p in controlling GSK-3ß activity. Bioinformatics analyses highlighted the serine/threonine-protein phosphatases PPP2CA, responsible for GSK-3ß activation, among the miR-675-5p targets, thus indicating the molecular mediator through which miR-675-5p may control ß-catenin nuclear localization. In conclusion, here we demonstrated that the inhibition of the hypoxia-induced non-coding RNA miR-675-5p hampered the nuclear localization of ß-catenin by regulating GSK-3ß activity, thus proposing the miR-675-5p as a new therapeutic target for the treatment of colorectal cancer.


Asunto(s)
Hipoxia de la Célula , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , MicroARNs/metabolismo , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Biología Computacional , Células HCT116 , Humanos , Estimación de Kaplan-Meier , Microscopía Fluorescente , Mutación , Unión Proteica , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA