Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(3): e1012060, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38442126

ABSTRACT

The recent discovery of Hepatitis D (HDV)-like viruses across a wide range of taxa led to the establishment of the Kolmioviridae family. Recent studies suggest that kolmiovirids can be satellites of viruses other than Hepatitis B virus (HBV), challenging the strict HBV/HDV-association dogma. Studying whether kolmiovirids are able to replicate in any animal cell they enter is essential to assess their zoonotic potential. Here, we compared replication of three kolmiovirids: HDV, rodent (RDeV) and snake (SDeV) deltavirus in vitro and in vivo. We show that SDeV has the narrowest and RDeV the broadest host cell range. High resolution imaging of cells persistently replicating these viruses revealed nuclear viral hubs with a peculiar RNA-protein organization. Finally, in vivo hydrodynamic delivery of viral replicons showed that both HDV and RDeV, but not SDeV, efficiently replicate in mouse liver, forming massive nuclear viral hubs. Our comparative analysis lays the foundation for the discovery of specific host factors controlling Kolmioviridae host-shifting.


Subject(s)
Hepatitis D , Hepatitis Delta Virus , Mice , Animals , Humans , Rodentia , Hepatitis B virus/genetics , Snakes , Virus Replication , RNA, Viral/genetics
2.
Methods Mol Biol ; 2769: 77-85, 2024.
Article in English | MEDLINE | ID: mdl-38315390

ABSTRACT

Hydrodynamic tail vein injection (HTVi), also called hydrodynamic gene transfer (HGT), is attracting increasing interest for modeling hepatic carcinogenesis. This highly versatile approach reproducibly provides efficient in vivo transfection of hepatocytes with naked DNA. Here, we give an in-depth description of the injection procedure, which is key for the success of the method. HTVi requires the injection of a large volume of a solution containing plasmids into the tail vein of the mouse. The transient right heart overload created by the injection forces the blood to flow back into the hepatic veins, enlarging the endothelial fenestrae and permeabilizing a fraction of hepatocytes for a few seconds. This results in the uptake of plasmids by the permeabilized hepatocytes, giving rise to their in vivo transfection. Including the Sleeping Beauty transposon system among the injected plasmids leads to the stable transfection of a subset of hepatocytes. HTVi is a powerful technique which enables numerous applications in liver cancer biology, such as a study of oncogene cooperation, of tumor heterogeneity, and interaction with the tumor microenvironment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Carcinoma, Hepatocellular/pathology , Hydrodynamics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Hepatocytes , Liver/pathology , Transfection , Plasmids/genetics , Carcinogenesis/pathology , Tumor Microenvironment
3.
EMBO Mol Med ; 16(2): 238-250, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38228803

ABSTRACT

FGF19 hormone has pleiotropic metabolic functions, including the modulation of insulin sensitivity, glucose/lipid metabolism and energy homeostasis. On top of its physiological metabolic role, FGF19 has been identified as a potentially targetable oncogenic driver, notably in hepatocellular carcinoma (HCC). Nevertheless, FGF19 remained an attractive candidate for treatment of metabolic disease, prompting the development of analogs uncoupling its metabolic and tumor-promoting activities. Using pre-clinical mice models of somatic mutation driven HCC, we assessed the oncogenicity of FGF19 in combination with frequent HCC tumorigenic alterations: p53 inactivation, CTNNB1 mutation, CCND1 or MYC overexpression. Our data revealed a strong oncogenic cooperation between FGF19 and MYC. Most importantly, we show that this oncogenic synergy is conserved with a FGF19-analog Aldafermin (NGM282), designed to solely mimic the hormone's metabolic functions. In particular, even a short systemic treatment with recombinant proteins triggered rapid appearance of proliferative foci of MYC-expressing hepatocytes. The fact that FGF19 analog Aldafermin is not fully devoid of the hormone's oncogenic properties raises concerns in the context of its potential use for patients with damaged, mutation-prone liver.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hormones
4.
Elife ; 122023 01 19.
Article in English | MEDLINE | ID: mdl-36656749

ABSTRACT

Quantitative differences in signal transduction are to date an understudied feature of tumour heterogeneity. The MAPK Erk pathway, which is activated in a large proportion of human tumours, is a prototypic example of distinct cell fates being driven by signal intensity. We have used primary hepatocyte precursors transformed with different dosages of an oncogenic form of Ras to model subclonal variations in MAPK signalling. Orthotopic allografts of Ras-transformed cells in immunocompromised mice gave rise to fast-growing aggressive tumours, both at the primary location and in the peritoneal cavity. Fluorescent labelling of cells expressing different oncogene levels, and consequently varying levels of MAPK Erk activation, highlighted the selection processes operating at the two sites of tumour growth. Indeed, significantly higher Ras expression was observed in primary as compared to secondary, metastatic sites, despite the apparent evolutionary trade-off of increased apoptotic death in the liver that correlated with high Ras dosage. Analysis of the immune tumour microenvironment at the two locations suggests that fast peritoneal tumour growth in the immunocompromised setting is abrogated in immunocompetent animals due to efficient antigen presentation by peritoneal dendritic cells. Furthermore, our data indicate that, in contrast to the metastatic-like outgrowth, strong MAPK signalling is required in the primary liver tumours to resist elimination by NK (natural killer) cells. Overall, this study describes a quantitative aspect of tumour heterogeneity and points to a potential vulnerability of a subtype of hepatocellular carcinoma as a function of MAPK Erk signalling intensity.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Carcinoma, Hepatocellular/genetics , Killer Cells, Natural , Liver Neoplasms/genetics , MAP Kinase Signaling System , Signal Transduction , Tumor Microenvironment , ras Proteins/metabolism
5.
Mol Metab ; 60: 101483, 2022 06.
Article in English | MEDLINE | ID: mdl-35367668

ABSTRACT

Fibroblast growth factor 19 (FGF19) is a hormone with pleiotropic metabolic functions, leading to ongoing development of analogues for treatment of metabolic disorders. On the other hand, FGF19 is overexpressed in a sub-group of hepatocellular carcinoma (HCC) patients and has oncogenic properties. It is therefore crucial to precisely define FGF19 effects, notably in the context of chronic exposure to elevated concentrations of the hormone. Here, we used hydrodynamic gene transfer to generate a transgenic mouse model with long-term FGF19 hepatic overexpression. We describe a novel effect of FGF19, namely the stimulation of water intake. This phenotype, lasting at least over a 6-month period, depends on signaling in the central nervous system and is independent of FGF21, although it mimics some of its features. We further show that HCC patients with high levels of circulating FGF19 have a reduced natremia, indicating dipsogenic features. The present study provides evidence of a new activity of FGF19, which could be clinically relevant in the context of FGF19 overexpressing cancers and in the course of treatment of metabolic disorders by FGF19 analogues.


Subject(s)
Carcinoma, Hepatocellular , Fibroblast Growth Factors/metabolism , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/metabolism , Drinking , Fibroblast Growth Factors/genetics , Hormones , Humans , Liver Neoplasms/metabolism , Mice , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism
6.
Pathogens ; 11(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35055994

ABSTRACT

Chronic hepatitis C carries a high risk of development of hepatocellular carcinoma (HCC), triggered by both direct and indirect effects of the virus. We examined cell-autonomous alterations in gene expression profiles associated with hepatitis C viral presence. Highly sensitive single molecule fluorescent in situ hybridization applied to frozen tissue sections of a hepatitis C patient allowed the delineation of clusters of infected hepatocytes. Laser microdissection followed by RNAseq analysis of hepatitis C virus (HCV)-positive and -negative regions from the tumoral and non-tumoral tissues from the same patient revealed HCV-related deregulation of expression of genes in the tumor and in the non-tumoral tissue. However, there was little overlap between both gene sets. Our interest in alterations that increase the probability of tumorigenesis prompted the examination of genes whose expression was increased by the virus in the non-transformed cells and whose level remained high in the tumor. This strategy led to the identification of a novel HCV target gene: GOLT1B, which encodes a protein involved in ER-Golgi trafficking. We further show that GOLT1B expression is induced during the unfolded protein response, that its presence is essential for efficient viral replication, and that its expression is correlated with poor outcome in HCC.

7.
J Hepatol ; 74(4): 893-906, 2021 04.
Article in English | MEDLINE | ID: mdl-33197513

ABSTRACT

BACKGROUND & AIMS: The tumour microenvironment shapes tumour growth through cellular communications that include both direct interactions and secreted factors. The aim of this study was to characterize the impact of the secreted glycoprotein ADAMTSL5, whose role in cancer has not been previously investigated, on hepatocellular carcinoma (HCC). METHODS: ADAMTSL5 methylation status was evaluated through bisulfite sequencing, and publicly available data analysis. ADAMTSL5 RNA and protein expression were assessed in mouse models and HCC patient samples and compared to data from published datasets. Functional studies, including association of ADAMTSL5 depletion with responsiveness to clinically relevant drugs, were performed in cellular and in vivo models. Molecular alterations associated with ADAMTSL5 targeting were determined using proteomics, biochemistry, and reverse-transcription quantitative PCR. RESULTS: Methylome analysis revealed hypermethylated gene body CpG islands at the ADAMTSL5 locus in both mouse and human HCC, correlating with higher ADAMTSL5 expression. ADAMTSL5 targeting interfered with tumorigenic properties of HCC cells in vitro and in vivo, whereas ADAMTSL5 overexpression conferred tumorigenicity to pre-tumoural hepatocytes sensitized to transformation by a modest level of MET receptor expression. Mechanistically, ADAMTSL5 abrogation led to a reduction of several oncogenic inputs relevant to HCC, including reduced expression and/or phosphorylation levels of receptor tyrosine kinases MET, EGFR, PDGFRß, IGF1Rß, or FGFR4. This phenotype was associated with significantly increased sensitivity of HCC cells to clinically relevant drugs, namely sorafenib, lenvatinib, and regorafenib. Moreover, ADAMTSL5 depletion drastically increased expression of AXL, accompanied by a sensitization to bemcentinib. CONCLUSIONS: Our results point to a role for ADAMTSL5 in maintaining the function of key oncogenic signalling pathways, suggesting that it may act as a master regulator of tumorigenicity and drug resistance in HCC. LAY SUMMARY: The environment of cancer cells has profound effects on establishment, progression, and response of a tumour to treatment. Herein, we show that ADAMTSL5, a protein secreted by liver cancer cells and overlooked in cancer so far, is increased in this tumour type, is necessary for tumour formation and supports drug resistance. Adamtsl5 removal conferred sensitivity of liver cancer cells to drugs used in current treatment. This suggests ADAMTSL5 as a potential marker in liver cancer as well as a possible drug target.


Subject(s)
ADAMTS Proteins , ADAMTS5 Protein , Carcinogenesis , Carcinoma, Hepatocellular , Drug Resistance, Neoplasm/physiology , Liver Neoplasms , Signal Transduction , ADAMTS Proteins/genetics , ADAMTS Proteins/metabolism , ADAMTS5 Protein/genetics , ADAMTS5 Protein/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Benzocycloheptenes/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Epigenomics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Sorafenib/pharmacology , Transcriptional Activation , Triazoles/pharmacology , Tumor Microenvironment/physiology
8.
Oncogene ; 39(13): 2676-2691, 2020 03.
Article in English | MEDLINE | ID: mdl-32020053

ABSTRACT

Chromatin organization is essential for appropriate interpretation of the genetic information. Here, we demonstrated that the chromatin-associated proteins HP1 are dispensable for hepatocytes survival but are essential within hepatocytes to prevent liver tumor development in mice with HP1ß being pivotal in these functions. Yet, we found that the loss of HP1 per se is not sufficient to induce cell transformation but renders cells more resistant to specific stress such as the expression of oncogenes and thus in fine, more prone to cell transformation. Molecular characterization of HP1-Triple KO premalignant livers and BMEL cells revealed that HP1 are essential for the maintenance of heterochromatin organization and for the regulation of specific genes with most of them having well characterized functions in liver functions and homeostasis. We further showed that some specific retrotransposons get reactivated upon loss of HP1, correlating with overexpression of genes in their neighborhood. Interestingly, we found that, although HP1-dependent genes are characterized by enrichment H3K9me3, this mark does not require HP1 for its maintenance and is not sufficient to maintain gene repression in absence of HP1. Finally, we demonstrated that the loss of TRIM28 association with HP1 recapitulated several phenotypes induced by the loss of HP1 including the reactivation of some retrotransposons and the increased incidence of liver cancer development. Altogether, our findings indicate that HP1 proteins act as guardians of liver homeostasis to prevent tumor development by modulating multiple chromatin-associated events within both the heterochromatic and euchromatic compartments, partly through regulation of the corepressor TRIM28 activity.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Animals , Cell Line , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Disease Models, Animal , Female , Hepatocytes , Heterochromatin/metabolism , Humans , Liver/cytology , Liver/pathology , Liver Neoplasms/pathology , Male , Mice , Mice, Knockout , Protein Binding/genetics , RNA-Seq , Retroelements/genetics , Tripartite Motif-Containing Protein 28/metabolism
9.
J Virol ; 90(13): 6022-6035, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27099311

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) triggers innate immunity signaling in the infected cell. Replication of the viral genome is dispensable for this phenotype, and we along with others have recently shown that NS5B, the viral RNA-dependent RNA polymerase, synthesizes double-stranded RNA (dsRNA) from cellular templates, thus eliciting an inflammatory response, notably via activation of type I interferon and lymphotoxin ß. Here, we investigated intracellular signal transduction pathways involved in this process. Using HepaRG cells, a model that largely recapitulates the in vivo complexities of the innate immunity receptor signaling, we have confirmed that NS5B triggered increased expression of the canonical pattern recognition receptors (PRRs) specific for dsRNA, namely, RIG-I, MDA5, and Toll-like receptor 3 (TLR3). Unexpectedly, intracellular dsRNA also led to accumulation of NOD1, a receptor classically involved in recognition of bacterial peptidoglycans. NOD1 activation, confirmed by analysis of its downstream targets, was likely due to its interaction with dsRNA and was independent of RIG-I and mitochondrial antiviral signaling protein (MAVS/IPS-1/Cardif/VISA) signaling. It is likely to have a functional significance in the cellular response in the context of HCV infection since interference with the NOD1 pathway severely reduced the inflammatory response elicited by NS5B. IMPORTANCE: In this study, we show that NOD1, a PRR that normally senses bacterial peptidoglycans, is activated by HCV viral polymerase, probably through an interaction with dsRNA, suggesting that NOD1 acts as an RNA ligand recognition receptor. In consequence, interference with NOD1-mediated signaling significantly weakens the inflammatory response to dsRNA. These results add a new level of complexity to the understanding of the cross talk between different classes of pattern recognition receptors and may be related to certain complications of chronic hepatitis C virus infection.


Subject(s)
Hepacivirus/immunology , Nod1 Signaling Adaptor Protein/metabolism , RNA, Double-Stranded/metabolism , RNA-Dependent RNA Polymerase/metabolism , Receptors, Pattern Recognition/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line , Cytoplasm/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Hepacivirus/enzymology , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatocytes/virology , Humans , Immunity, Innate , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Nod1 Signaling Adaptor Protein/genetics , RNA, Double-Stranded/immunology , RNA-Dependent RNA Polymerase/genetics , Receptors, Immunologic , Signal Transduction , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Viral Nonstructural Proteins/genetics
10.
Contemp Oncol (Pozn) ; 19(1A): A62-7, 2015.
Article in English | MEDLINE | ID: mdl-25691824

ABSTRACT

Viruses are considered as causative agents of a significant proportion of human cancers. While the very stringent criteria used for their classification probably lead to an underestimation, only six human viruses are currently classified as oncogenic. In this review we give a brief historical account of the discovery of oncogenic viruses and then analyse the mechanisms underlying the infectious causes of cancer. We discuss viral strategies that evolved to ensure virus propagation and spread can alter cellular homeostasis in a way that increases the probability of oncogenic transformation and acquisition of stem cell phenotype. We argue that a useful way of analysing the convergent characteristics of viral infection and cancer is to examine how viruses affect the so-called cancer hallmarks. This view of infectious origin of cancer is illustrated by examples from hepatitis C infection, which is associated with a high proportion of hepatocellular carcinoma.

11.
Methods Mol Biol ; 1092: 421-36, 2014.
Article in English | MEDLINE | ID: mdl-24318834

ABSTRACT

Targeted cell ablation has proven to be a valuable approach to study in vivo cell functions during organogenesis, tissue homeostasis, and regeneration. Over the last two decades, various approaches have been developed to refine the control of cell ablation. In this review, we give an overview of the distinct genetic tools available for targeted cell ablation, with a particular emphasis on their respective specificity.


Subject(s)
Apoptosis/genetics , Diphtheria Toxin/genetics , Genes, Transgenic, Suicide/genetics , Organogenesis/genetics , Diphtheria Toxin/metabolism , Eukaryotic Initiation Factor-3/genetics , Eukaryotic Initiation Factor-3/metabolism , Homeostasis/genetics , Humans , Molecular Biology/methods , Regeneration/genetics
12.
Nat Commun ; 4: 2065, 2013.
Article in English | MEDLINE | ID: mdl-23817338

ABSTRACT

Cdt1 is required for loading the replicative DNA helicase MCM2/7, a process known as DNA replication licensing. Here we show that 129 mouse strains express a Cdt1 mutated allele with enhanced licensing activity. The mutation, named Δ(6)PEST, involves a six-amino acid deletion within a previously uncharacterized PEST-like domain. Cdt1 Δ(6)PEST and more extensive deletions exhibit increased re-replication and transformation activities that are independent of the Geminin and E3 ligase pathways. This PEST domain negatively regulates cell cycle-dependent chromatin recruitment of Cdt1 in G2/M phases of the cell cycle. Mass spectrometry analysis indicates that Cdt1 is phosphorylated at sites within the deleted PEST domain during mitosis. This study reveals a conserved new regulatory Cdt1 domain crucial for proper DNA licensing activity and suggests a mechanism by which the presence of Cdt1 in G2/M phases does not lead to premature origin licensing. These results also question the usage of 129 mouse strains for knockout analyses.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Mutation/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Carcinogenesis , Cell Extracts , Cell Line , Chromatin/metabolism , Geminin/metabolism , Humans , Mice , Mice, 129 Strain , Mitosis , Models, Biological , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Structure, Tertiary , Sequence Deletion , Ubiquitin-Protein Ligases/metabolism
13.
Development ; 140(10): 2130-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23633510

ABSTRACT

Limb development relies on an exquisite coordination between growth and patterning, but the underlying mechanisms remain elusive. Anterior-posterior and proximal-distal specification initiates in early limb bud concomitantly with the proliferative expansion of limb cells. Previous studies have shown that limb bud growth initially relies on fibroblast growth factors (FGFs) produced in the apical ectodermal ridge (AER-FGFs), the maintenance of which relies on a positive-feedback loop involving sonic hedgehog (Shh) and the BMP antagonist gremlin 1 (Grem1). The positive cross-regulation between Shh and the HoxA and HoxD clustered genes identified an indirect effect of Hox genes on the maintenance of AER-FGFs but the respective function of Shh and Hox genes in this process remains unknown. Here, by uncoupling Hox and Shh function, we show that HoxA and HoxD genes are required for proper AER-FGFs expression, independently of their function in controlling Shh expression. In addition, we provide evidence that the Hox-dependent control of AER-FGF expression is achieved through the regulation of key mesenchymal signals, namely Grem1 and Fgf10, ensuring proper epithelial-mesenchymal interactions. Notably, HoxA and HoxD genes contribute to both the initial activation of Grem1 and the subsequent anterior expansion of its expression domain. We propose that the intricate interactions between Hox genes and the FGF and Shh signaling pathways act as a molecular network that ensures proper limb bud growth and patterning, probably contributing to the coordination of these two processes.


Subject(s)
Extremities/embryology , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Homeodomain Proteins/metabolism , Alleles , Animals , Down-Regulation , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 8/metabolism , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Multigene Family , Mutation , Nerve Tissue Proteins/metabolism , Tomography , Zinc Finger Protein Gli3
14.
PLoS Pathog ; 9(3): e1003234, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555249

ABSTRACT

Exposure to hepatitis C virus (HCV) typically results in chronic infection that leads to progressive liver disease ranging from mild inflammation to severe fibrosis and cirrhosis as well as primary liver cancer. HCV triggers innate immune signaling within the infected hepatocyte, a first step in mounting of the adaptive response against HCV infection. Persistent inflammation is strongly associated with liver tumorigenesis. The goal of our work was to investigate the initiation of the inflammatory processes triggered by HCV viral proteins in their host cell and their possible link with HCV-related liver cancer. We report a dramatic upregulation of the lymphotoxin signaling pathway and more specifically of lymphotoxin-ß in tumors of the FL-N/35 HCV-transgenic mice. Lymphotoxin expression is accompanied by activation of NF-κB, neosynthesis of chemokines and intra-tumoral recruitment of mononuclear cells. Spectacularly, IKKß inactivation in FL-N/35 mice drastically reduces tumor incidence. Activation of lymphotoxin-ß pathway can be reproduced in several cellular models, including the full length replicon and HCV-infected primary human hepatocytes. We have identified NS5B, the HCV RNA dependent RNA polymerase, as the viral protein responsible for this phenotype and shown that pharmacological inhibition of its activity alleviates activation of the pro-inflammatory pathway. These results open new perspectives in understanding the inflammatory mechanisms linked to HCV infection and tumorigenesis.


Subject(s)
Hepacivirus/enzymology , Liver Neoplasms/metabolism , Lymphocytes/immunology , Lymphotoxin-beta/metabolism , RNA-Dependent RNA Polymerase/metabolism , Animals , Cell Line , Chemokines/metabolism , Chemotaxis, Leukocyte , Hepacivirus/pathogenicity , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , I-kappa B Kinase/metabolism , Immunity, Innate , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Liver/metabolism , Liver/pathology , Liver/virology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Lymphocyte Activation , Lymphocytes/virology , Male , Mice , Mice, Transgenic , NF-kappa B , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Signal Transduction , Up-Regulation , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism
15.
Evol Appl ; 6(1): 1-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23397042

ABSTRACT

Since the mid 1970s, cancer has been described as a process of Darwinian evolution, with somatic cellular selection and evolution being the fundamental processes leading to malignancy and its many manifestations (neoangiogenesis, evasion of the immune system, metastasis, and resistance to therapies). Historically, little attention has been placed on applications of evolutionary biology to understanding and controlling neoplastic progression and to prevent therapeutic failures. This is now beginning to change, and there is a growing international interest in the interface between cancer and evolutionary biology. The objective of this introduction is first to describe the basic ideas and concepts linking evolutionary biology to cancer. We then present four major fronts where the evolutionary perspective is most developed, namely laboratory and clinical models, mathematical models, databases, and techniques and assays. Finally, we discuss several of the most promising challenges and future prospects in this interdisciplinary research direction in the war against cancer.

17.
J Hepatol ; 57(5): 1021-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22750466

ABSTRACT

BACKGROUND & AIMS: Apicobasal polarity, which is essential for epithelial structure and function, is targeted by several tumour-related pathogens and is generally perturbed in the course of carcinogenesis. Hepatitis C virus (HCV) infection is associated with a strong risk of hepatocellular carcinoma, typically preceded by dysplastic alterations of cell morphology. We investigated the molecular mechanisms and the functional consequences of HCV-driven perturbations of epithelial polarity. METHODS: We used biochemical, genetic, and cell biology approaches to assess the impact of hepatitis C viral protein NS5A on the polarity and function of hepatocytes and hepatic progenitors. Transgenic animals and xenograft models served for in vivo validation of the results obtained in cell culture. RESULTS: We found that expression of HCV-NS5A in primary hepatic precursors and in immortalized hepatocyte cell lines gave rise to profound modifications of cell polarity, leading to epithelial to mesenchymal transition (EMT). NS5A, either alone or in the context of the full complement of viral proteins in the course of infection, acted through activating Twist2, a transcriptional regulator of EMT. The effects of NS5A were additive to those of TGF-ß, a cytokine abundant in diseased liver and highly relevant to HCV-related pathology. Moreover, NS5A cooperates with oncogenic Ras, giving rise to transformed, invasive cells that are highly tumorigenic in vivo. CONCLUSIONS: Our data suggest that in the context of HCV infection, NS5A favors formation of preneoplastic lesions by disrupting cell polarity and additional oncogenic events cooperate with the viral protein to give rise to motile and invasive tumour cells.


Subject(s)
Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition/physiology , Hepatitis C/complications , Hepatocytes/pathology , Viral Nonstructural Proteins/physiology , Animals , Animals, Genetically Modified , Cell Line , Cell Polarity/physiology , Cells, Cultured , Hepatitis C/metabolism , Hepatitis C/pathology , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins p21(ras)/metabolism , Repressor Proteins/physiology , Risk Factors , Transforming Growth Factor beta/metabolism , Transplantation, Heterologous , Twist-Related Protein 1/physiology
18.
Methods ; 57(2): 158-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22796403

ABSTRACT

Understanding the nature of DNA replication origins in metazoan is quite challenging. In the absence of a genetic assay like in yeast, methods were devised based on the DNA structure, the visualization or quantification of the first nascent strands that are synthesized at origins, or on the localization of origin binding proteins. The purification and quantification of RNA-primed nascent DNA at origins during initiation of DNA synthesis is the most exhaustive and precise method to map active replication origins at present. We have upgraded this method to the level of reproducibility and enrichment required for genome-wide analyses by microarrays or deep sequencing. We detail here the protocol and the controls required at the different steps.


Subject(s)
DNA Replication , DNA/biosynthesis , Replication Origin , Animals , Cell Culture Techniques , Cells, Cultured , DNA/chemistry , DNA/isolation & purification , DNA Cleavage , Exodeoxyribonucleases/chemistry , Genetic Loci , Genome , Homeodomain Proteins/genetics , Humans , Liquid-Liquid Extraction , Oligonucleotide Array Sequence Analysis , RNA Cleavage , Real-Time Polymerase Chain Reaction , Ribonuclease, Pancreatic/chemistry
19.
Proc Natl Acad Sci U S A ; 105(38): 14492-6, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18787116

ABSTRACT

The loxP/Cre recombination system is a widely used tool for mouse functional genomics, in particular for in vivo conditional mutagenesis. Depending on the relative orientation and position of loxP sites, Cre-mediated recombination can result in a variety of targeted genomic rearrangements. It was previously reported that loss of the loxP-carrying chromosome can occur when loxP sites are arranged in inverse orientation. By using a chromosome 2 carrying inverted loxP sites, we found that Cre-mediated recombination not only causes chromosomal loss but also triggers apoptosis. We show that targeted recombination between inverted loxP sites (TRIP) triggers cell death specifically in proliferating Cre-expressing cells, and we provide evidence that TRIP is an efficient tool to ablate proliferating cells within genetically defined cell populations. Furthermore, the procedure requires only a simple, one-step intercross but neither the use of toxins nor the additional step of prodrug injection. With the large repertoire of tissue-specific or inducible Cre-expressing transgenes available, TRIP-mediated cell ablation is valuable to investigate the function of a large variety of cell populations in the context of a whole organism, which includes mechanisms underlying organ development and tissue homeostasis.


Subject(s)
Apoptosis , Attachment Sites, Microbiological/genetics , Integrases/metabolism , Monosomy/genetics , Recombination, Genetic , Animals , Apoptosis/genetics , Cell Proliferation , Cell Survival/genetics , Chromosome Deletion , Embryo, Mammalian/cytology , Mice , Organ Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...