Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(2)2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38256189

ABSTRACT

Shigellosis, an acute gastroenteritis infection caused by Shigella species, remains a public health burden in developing countries. Recently, many outbreaks due to Shigella sonnei multidrug-resistant strains have been reported in high-income countries, and the lack of an effective vaccine represents a major hurdle to counteract this bacterial pathogen. Vaccine candidates against Shigella sonnei are under clinical development, including a Generalized Modules for Membrane Antigens (GMMA)-based vaccine. The mechanisms by which GMMA-based vaccines interact and activate human immune cells remain elusive. Our previous study provided the first evidence that both adaptive and innate immune cells are targeted and functionally shaped by the GMMA-based vaccine. Here, flow cytometry and confocal microscopy analysis allowed us to identify monocytes as the main target population interacting with the S. sonnei 1790-GMMA vaccine on human peripheral blood. In addition, transcriptomic analysis of this cell population revealed a molecular signature induced by 1790-GMMA mostly correlated with the inflammatory response and cytokine-induced processes. This also impacts the expression of genes associated with macrophages' differentiation and T cell regulation, suggesting a dual function for this vaccine platform both as an antigen carrier and as a regulator of immune cell activation and differentiation.


Subject(s)
Blood Group Antigens , Gastroenteritis , Methylmethacrylates , Vaccines , Humans , Monocytes , Shigella sonnei/genetics , Antigens, Bacterial/genetics
2.
NPJ Vaccines ; 8(1): 34, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36890168

ABSTRACT

The mechanisms by which antibodies confer protection vary across vaccines, ranging from simple neutralization to functions requiring innate immune recruitment via Fc-dependent mechanisms. The role of adjuvants in shaping the maturation of antibody-effector functions remains under investigated. Using systems serology, we compared adjuvants in licensed vaccines (AS01B/AS01E/AS03/AS04/Alum) combined with a model antigen. Antigen-naive adults received two adjuvanted immunizations followed by late revaccination with fractional-dosed non-adjuvanted antigen ( NCT00805389 ). A dichotomy in response quantities/qualities emerged post-dose 2 between AS01B/AS01E/AS03 and AS04/Alum, based on four features related to immunoglobulin titers or Fc-effector functions. AS01B/E and AS03 induced similar robust responses that were boosted upon revaccination, suggesting that memory B-cell programming by the adjuvanted vaccinations dictated responses post non-adjuvanted boost. AS04 and Alum induced weaker responses, that were dissimilar with enhanced functionalities for AS04. Distinct adjuvant classes can be leveraged to tune antibody-effector functions, where selective vaccine formulation using adjuvants with different immunological properties may direct antigen-specific antibody functions.

3.
Bioinformatics ; 38(7): 2062-2063, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35104317

ABSTRACT

SUMMARY: Comparisons of protein structures are critical for developing novel protein designs, annotating protein functions and predicting protein structure. The template modeling score (TM-score) is a widely used but computationally expensive measure of protein similarity that is applicable to a wide variety of structural biology problems. We introduce TMQuery-a continuously updated database containing over eight billion pre-computed TM-score values for every pair of proteins in the Protein Data Bank, allowing researchers to quickly query and download TM-scores via a web interface. AVAILABILITY AND IMPLEMENTATION: Publicly available at https://tmquery.gsk.com/.


Subject(s)
Proteins , Software , Protein Conformation , Proteins/chemistry , Databases, Protein
4.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068078

ABSTRACT

Anthracyclines remain a cornerstone of induction chemotherapy for acute myeloid leukemia (AML). Refractory or relapsed disease due to chemotherapy resistance is a major obstacle in AML management. MicroRNAs (miRNAs) have been observed to be involved in chemoresistance. We previously observed that miR-15a-5p was overexpressed in a subgroup of chemoresistant cytogenetically normal AML patients compared with chemosensitive patients treated with daunorubicin and cytarabine. MiR-15a-5p overexpression in AML cells reduced apoptosis induced by both drugs in vitro. This study aimed to elucidate the mechanisms by which miR-15a-5p contributes to daunorubicin resistance. We showed that daunorubicin induced autophagy in myeloid cell lines. The inhibition of autophagy reduced cell sensitivity to daunorubicin. The overexpression of miR-15a-5p decreased daunorubicin-induced autophagy. Conversely, the downregulation of miR-15a-5p increased daunorubicin-induced autophagy. We found that miR-15a-5p targeted four genes involved in autophagy, namely ATG9a, ATG14, GABARAPL1 and SMPD1. Daunorubicin increased the expression of these four genes, and miR-15a-5p counteracted this regulation. Inhibition experiments with the four target genes showed the functional effect of miR-15a-5p on autophagy. In summary, our results indicated that miR-15a-5p induces chemoresistance in AML cells through the abrogation of daunorubicin-induced autophagy, suggesting that miR-15a-5p could be a promising therapeutic target for chemoresistant AML patients.


Subject(s)
Biomarkers, Tumor/metabolism , Daunorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/drug therapy , MicroRNAs/genetics , Adult , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Autophagy , Biomarkers, Tumor/genetics , Cell Proliferation , Gene Expression Profiling , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Tumor Cells, Cultured
5.
J Cell Mol Med ; 25(1): 575-585, 2021 01.
Article in English | MEDLINE | ID: mdl-33270982

ABSTRACT

Cytarabine and daunorubicin are old drugs commonly used in the treatment of acute myeloid leukaemia (AML). Refractory or relapsed disease because of chemotherapy resistance is a major issue. microRNAs (miRNAs) were incriminated in resistance. This study aimed to identify miRNAs involved in chemoresistance in AML patients and to define their target genes. We focused on cytogenetically normal AML patients with wild-type NPM1 without FLT3-ITD as the treatment of this subset of patients with intermediate-risk cytogenetics is not well established. We analysed baseline AML samples by small RNA sequencing and compared the profile of chemoresistant to chemosensitive AML patients. Among the miRNAs significantly overexpressed in chemoresistant patients, we revealed miR-15a-5p and miR-21-5p as miRNAs with a major role in chemoresistance in AML. We showed that miR-15a-5p and miR-21-5p overexpression decreased apoptosis induced by cytarabine and/or daunorubicin. PDCD4, ARL2 and BTG2 genes were found to be targeted by miR-15a-5p, as well as PDCD4 and BTG2 by miR-21-5p. Inhibition experiments of the three target genes reproduced the functional effect of both miRNAs on chemosensitivity. Our study demonstrates that miR-15a-5p and miR-21-5p are overexpressed in a subgroup of chemoresistant AML patients. Both miRNAs induce chemoresistance by targeting three pro-apoptotic genes PDCD4, ARL2 and BTG2.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , MicroRNAs/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cytarabine/pharmacology , Daunorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Humans , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Principal Component Analysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
6.
Sci Transl Med ; 12(569)2020 11 11.
Article in English | MEDLINE | ID: mdl-33177181

ABSTRACT

The current routine use of adjuvants in human vaccines provides a strong incentive to increase our understanding of how adjuvants differ in their ability to stimulate innate immunity and consequently enhance vaccine immunogenicity. Here, we evaluated gene expression profiles in cells from whole blood elicited in naive subjects receiving the hepatitis B surface antigen formulated with different adjuvants. We identified a core innate gene signature emerging 1 day after the second vaccination and that was shared by the recipients of vaccines formulated with adjuvant systems AS01B, AS01E, or AS03. This core signature associated with the magnitude of the hepatitis B surface-specific antibody response and was characterized by positive regulation of genes associated with interferon-related responses or the innate cell compartment and by negative regulation of natural killer cell-associated genes. Analysis at the individual subject level revealed that the higher immunogenicity of AS01B-adjuvanted vaccine was linked to its ability to induce this signature in most vaccinees even after the first vaccination. Therefore, our data suggest that adjuvanticity is not strictly defined by the nature of the receptors or signaling pathways it activates but by the ability of the adjuvant to consistently induce a core inflammatory signature across individuals.


Subject(s)
Hepatitis B Vaccines , Influenza Vaccines , Adjuvants, Immunologic , Antibodies, Viral , Hepatitis B Surface Antigens/genetics , Humans , Immunogenicity, Vaccine , Vaccination
7.
Gut ; 67(2): 271-283, 2018 02.
Article in English | MEDLINE | ID: mdl-28377388

ABSTRACT

OBJECTIVE: To investigate the beneficial role of prebiotics on endothelial dysfunction, an early key marker of cardiovascular diseases, in an original mouse model linking steatosis and endothelial dysfunction. DESIGN: We examined the contribution of the gut microbiota to vascular dysfunction observed in apolipoprotein E knockout (Apoe-/-) mice fed an n-3 polyunsaturated fatty acid (PUFA)-depleted diet for 12 weeks with or without inulin-type fructans (ITFs) supplementation for the last 15 days. Mesenteric and carotid arteries were isolated to evaluate endothelium-dependent relaxation ex vivo. Caecal microbiota composition (Illumina Sequencing of the 16S rRNA gene) and key pathways/mediators involved in the control of vascular function, including bile acid (BA) profiling, gut and liver key gene expression, nitric oxide and gut hormones production were also assessed. RESULTS: ITF supplementation totally reverses endothelial dysfunction in mesenteric and carotid arteries of n-3 PUFA-depleted Apoe-/- mice via activation of the nitric oxide (NO) synthase/NO pathway. Gut microbiota changes induced by prebiotic treatment consist in increased NO-producing bacteria, replenishment of abundance in Akkermansia and decreased abundance in bacterial taxa involved in secondary BA synthesis. Changes in gut and liver gene expression also occur upon ITFs suggesting increased glucagon-like peptide 1 production and BA turnover as drivers of endothelium function preservation. CONCLUSIONS: We demonstrate for the first time that ITF improve endothelial dysfunction, implicating a short-term adaptation of both gut microbiota and key gut peptides. If confirmed in humans, prebiotics could be proposed as a novel approach in the prevention of metabolic disorders-related cardiovascular diseases.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Fructans/pharmacology , Gastrointestinal Microbiome/drug effects , Prebiotics , Aminopeptidases/genetics , Animals , Antimicrobial Cationic Peptides/genetics , Bacteria/drug effects , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/blood , Carotid Arteries/physiology , Cecum/microbiology , Dietary Supplements , Disease Models, Animal , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/deficiency , Gene Expression/drug effects , Glucagon-Like Peptide 1/biosynthesis , Male , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Neurotensin/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Organic Anion Transporters, Sodium-Dependent/genetics , Proglucagon/genetics , Symporters/genetics , Vasodilation
8.
Immun Inflamm Dis ; 5(3): 346-354, 2017 09.
Article in English | MEDLINE | ID: mdl-28508570

ABSTRACT

INTRODUCTION: While most transcripts arising from the human T Cell Receptor locus reflect fully rearranged genes, several germline transcripts have been identified. We describe a new germline transcript arising from the human TCRB locus. METHODS: cDNA sequencing, promoter, and gene expression analyses were used to characterize the new transcript. RESULTS: The new germline transcript encoded by the human TCRB locus consists of a new exon of 103 bp, which we named TRBX1 (X1), spliced with the first exon of gene segments Cß1 or Cß2. X1 is located upstream of gene segment Dß1 and is therefore deleted from a V-DJ rearranged TCRB locus. The X1-Cß transcripts do not appear to code for a protein. We define their transcription start and minimal promoter. These transcripts are found in populations of mature T lymphocytes from blood or tissues and in T cell clones with a monoallelic TCRB rearrangement. In immature thymocytes, they are already detectable in CD1a- CD34+ CD4- CD8- cells, therefore before completion of the TCRB rearrangements. CONCLUSIONS: The X1 promoter appears to be the ortholog of the mouse pre-Dß1 promoter (PDß1). Like PDß1, its activation is regulated by Eß in T cells and might facilitate the TCRB rearrangement process by contributing to the accessibility of the Dß1 locus.


Subject(s)
Genes, T-Cell Receptor beta , Genetic Loci , Promoter Regions, Genetic , RNA, Messenger/genetics , Transcription, Genetic , Animals , Humans , Mice , RNA, Messenger/biosynthesis
9.
FASEB J ; 31(2): 840-851, 2017 02.
Article in English | MEDLINE | ID: mdl-27856557

ABSTRACT

Activating transcription factor (ATF)3 regulates the expression of inflammation-related genes in several tissues under pathological contexts. In skeletal muscle, atf3 expression increases after exercise, but its target genes remain unknown. We aimed to identify those genes and to determine the influence of ATF3 on muscle adaptation to training. Skeletal muscles of ATF3-knockout (ATF3-KO) and control mice were analyzed at rest, after exercise, and after training. In resting muscles, there was no difference between genotypes in enzymatic activities or fiber type. After exercise, a microarray analysis in quadriceps revealed ATF3 affects genes modulating chemotaxis and chemokine/cytokine activity. Quantitative PCR showed that the mRNA levels of chemokine C-C motif ligand (ccl)8 and chemokine C-X-C motif ligand (cxcl)13 were higher in quadriceps of ATF3-KO mice than in control mice. The same was observed for ccl9 and cxcl13 in soleus. Also in soleus, ccl2, interleukin (il)6, il1ß, and cluster of differentiation (cd)68 mRNA levels increased after exercise only in ATF3-KO mice. Endurance training increased the basal mRNA level of hexokinase-2, hormone sensitive lipase, glutathione peroxidase-1, and myosin heavy chain IIa in quadriceps of control mice but not in ATF3-KO mice. In summary, ATF3 attenuates the expression of inflammation-related genes after exercise and thus facilitates molecular adaptation to training.-Fernández-Verdejo, R., Vanwynsberghe, A. M., Essaghir, A., Demoulin, J.-B., Hai, T., Deldicque, L., Francaux, M. Activating transcription factor 3 attenuates chemokine and cytokine expression in mouse skeletal muscle after exercise and facilitates molecular adaptation to endurance training.


Subject(s)
Activating Transcription Factor 3/metabolism , Muscle, Skeletal/physiology , Activating Transcription Factor 3/genetics , Animals , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Physical Conditioning, Animal , Physical Endurance/physiology
10.
Gut ; 66(4): 620-632, 2017 04.
Article in English | MEDLINE | ID: mdl-27196572

ABSTRACT

OBJECTIVE: To examine the role of hepatocyte myeloid differentiation primary-response gene 88 (MyD88) on glucose and lipid metabolism. DESIGN: To study the impact of the innate immune system at the level of the hepatocyte and metabolism, we generated mice harbouring hepatocyte-specific deletion of MyD88. We investigated the impact of the deletion on metabolism by feeding mice with a normal control diet or a high-fat diet for 8 weeks. We evaluated body weight, fat mass gain (using time-domain nuclear magnetic resonance), glucose metabolism and energy homeostasis (using metabolic chambers). We performed microarrays and quantitative PCRs in the liver. In addition, we investigated the gut microbiota composition, bile acid profile and both liver and plasma metabolome. We analysed the expression pattern of genes in the liver of obese humans developing non-alcoholic steatohepatitis (NASH). RESULTS: Hepatocyte-specific deletion of MyD88 predisposes to glucose intolerance, inflammation and hepatic insulin resistance independently of body weight and adiposity. These phenotypic differences were partially attributed to differences in gene expression, transcriptional factor activity (ie, peroxisome proliferator activator receptor-α, farnesoid X receptor (FXR), liver X receptors and STAT3) and bile acid profiles involved in glucose, lipid metabolism and inflammation. In addition to these alterations, the genetic deletion of MyD88 in hepatocytes changes the gut microbiota composition and their metabolomes, resembling those observed during diet-induced obesity. Finally, obese humans with NASH displayed a decreased expression of different cytochromes P450 involved in bioactive lipid synthesis. CONCLUSIONS: Our study identifies a new link between innate immunity and hepatic synthesis of bile acids and bioactive lipids. This dialogue appears to be involved in the susceptibility to alterations associated with obesity such as type 2 diabetes and NASH, both in mice and humans.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome/genetics , Glucose/metabolism , Hepatocytes/metabolism , Lipid Metabolism/genetics , Metabolome/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Adiposity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Body Weight , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Diet, High-Fat , Gene Expression , Humans , Immunity, Innate/genetics , Insulin Resistance/genetics , Liver/metabolism , Liver X Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/genetics , Obesity/metabolism , PPAR alpha/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , STAT3 Transcription Factor/metabolism
11.
Nat Commun ; 6: 6495, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25757720

ABSTRACT

Obesity is a pandemic disease associated with many metabolic alterations and involves several organs and systems. The endocannabinoid system (ECS) appears to be a key regulator of energy homeostasis and metabolism. Here we show that specific deletion of the ECS synthesizing enzyme, NAPE-PLD, in adipocytes induces obesity, glucose intolerance, adipose tissue inflammation and altered lipid metabolism. We report that Napepld-deleted mice present an altered browning programme and are less responsive to cold-induced browning, highlighting the essential role of NAPE-PLD in regulating energy homeostasis and metabolism in the physiological state. Our results indicate that these alterations are mediated by a shift in gut microbiota composition that can partially transfer the phenotype to germ-free mice. Together, our findings uncover a role of adipose tissue NAPE-PLD on whole-body metabolism and provide support for targeting NAPE-PLD-derived bioactive lipids to treat obesity and related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Gastrointestinal Microbiome/physiology , Glucose Intolerance/metabolism , Obesity/metabolism , Phospholipase D/genetics , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Body Fat Distribution , Cold Temperature , Endocannabinoids/metabolism , Energy Metabolism/physiology , Gene Expression , Glucose Intolerance/genetics , Glucose Intolerance/microbiology , Glucose Intolerance/pathology , Inflammation , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/microbiology , Obesity/pathology , Phospholipase D/deficiency
12.
J Cell Mol Med ; 19(1): 239-48, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25292412

ABSTRACT

Platelet-derived growth factors (PDGF) bind to two related receptor tyrosine kinases, which are encoded by the PDGFRA and PDGFRB genes. Recently, heterozygous PDGFRB mutations have been described in patients diagnosed with idiopathic basal ganglia calcification (IBGC or Fahr disease), a rare inherited neurological disorder. The goal of the present study was to determine whether these mutations had a positive or negative impact on the PDGFRB activity. We first showed that the E1071V mutant behaved like wild-type PDGFRB and may represent a polymorphism unrelated to IBGC. In contrast, the L658P mutant had no kinase activity and failed to activate any of the pathways normally stimulated by PDGF. The R987W mutant activated Akt and MAP kinases but did not induce the phosphorylation of signal transducer and activator of transcription 3 (STAT3) after PDGF stimulation. Phosphorylation of phospholipase Cγ was also decreased. Finally, we showed that the R987W mutant was more rapidly degraded upon PDGF binding compared to wild-type PDGFRB. In conclusion, PDGFRB mutations associated with IBGC impair the receptor signalling. PDGFRB loss of function in IBGC is consistent with recently described inactivating mutations in the PDGF-B ligand. These results raise concerns about the long-term safety of PDGF receptor inhibition by drugs such as imatinib.


Subject(s)
Basal Ganglia Diseases/genetics , Calcinosis/genetics , Mutation/genetics , Neurodegenerative Diseases/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction/genetics , Amino Acid Substitution , Cell Line, Tumor , HEK293 Cells , Humans , Mutant Proteins/metabolism , Phospholipase C gamma/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
13.
PLoS One ; 9(10): e108656, 2014.
Article in English | MEDLINE | ID: mdl-25280005

ABSTRACT

Transforming growth factor-ß (TGFß) is a key mediator of fibrogenesis. TGFß is overexpressed and activated in fibrotic diseases, regulates fibroblast differentiation into myofibroblasts and induces extracellular matrix deposition. Platelet-derived growth factor (PDGF) is also a regulator of fibrogenesis. Some studies showed a link between TGFß and PDGF in certain fibrotic diseases. TGFß induces PDGF receptor alpha expression in scleroderma fibroblasts. PDGF-C and -D are the most recently discovered ligands and also play a role in fibrosis. In this study, we report the first link between TGFß and PDGF-D and -C ligands. In normal fibroblasts, TGFß down-regulated PDGF-D expression and up-regulated PDGF-C expression at the mRNA and protein levels. This phenomenon is not limited to TGFß since other growth factors implicated in fibrosis, such as FGF, EGF and PDGF-B, also regulated PDGF-D and PDGF-C expression. Among different kinase inhibitors, only TGFß receptor inhibitors and the IκB kinase (IKK) inhibitor BMS-345541 blocked the effect of TGFß. However, activation of the classical NF-κB pathway was not involved. Interestingly, in a model of lung fibrosis induced by either bleomycin or silica, PDGF-D was down-regulated, which correlates with the production of TGFß and other fibrotic growth factors. In conclusion, the down-regulation of PDGF-D by TGFß and other growth factors may serve as a negative feedback in the network of cytokines that control fibrosis.


Subject(s)
Down-Regulation/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Transforming Growth Factor beta/pharmacology , Cell Line , Fibroblasts/cytology , Fibroblasts/drug effects , Foreskin/cytology , Foreskin/drug effects , Foreskin/metabolism , Humans , Lymphokines/genetics , Male , Platelet-Derived Growth Factor/genetics , Smad4 Protein/genetics , Smad4 Protein/metabolism , Up-Regulation/drug effects
14.
Methods Mol Biol ; 1164: 109-20, 2014.
Article in English | MEDLINE | ID: mdl-24927839

ABSTRACT

Thanks to high-throughput experiments, biological conditions can be investigated at both the entire genomic and transcriptomic levels. In addition, protein-protein interaction (PPI) data are widely available for well-studied organisms, such as human. In this chapter, we will present an integrative approach that makes use of these data to find the PPI module involving the key regulated transcription factors shared by a number of given conditions. These conditions could be for instance different cancer types. Briefly, for the studied conditions, we need to identify commonly affected chromosomal regions subjected to copy number alterations together with the identification of differentially expressed list of genes in each condition. Transcription factor activity will be inferred from these regulated gene lists. Then, we will define TFs, for which the activity could be explained by an associative effect of both loci copy number alteration and gene expression levels of their coding genes. PPI networks could be mined, afterwards, using appropriate algorithms to find the significant module that connect those TFs together. This module could be viewed as the minimal connected network of TFs, the regulation of which is shared between the investigated conditions.


Subject(s)
Genomics/methods , Protein Interaction Mapping/methods , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , Databases, Genetic , Gene Dosage , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Protein Interaction Maps
15.
Cytokine Growth Factor Rev ; 25(3): 273-83, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24703957

ABSTRACT

For about four decades, platelet-derived growth factors (PDGF) and their receptors have been the subject of intense research, revealing their roles in embryo development and human diseases. Drugs such as imatinib, which selectively inhibit the tyrosine kinase activity of these receptors, have been approved for the treatment of cancers such as gastrointestinal stromal tumors and chronic eosinophilic leukemia. Today, the interest in these factors is still increasing in relationship with new potential clinical applications in cancer, stroke, fibrosis and infectious diseases. This review focuses on the mechanisms of PDGF receptor signaling, with an emphasis on pathways that are important for disease development. Of particular interest, recent studies revealed significant differences between normal and cancer cells regarding signal transduction by these growth factors.


Subject(s)
Gastrointestinal Stromal Tumors/enzymology , Hypereosinophilic Syndrome/enzymology , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Benzamides/therapeutic use , Fibrosis/drug therapy , Fibrosis/enzymology , Fibrosis/pathology , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Humans , Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/pathology , Imatinib Mesylate , Infections/drug therapy , Infections/enzymology , Infections/pathology , Leukemia , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Stroke/drug therapy , Stroke/enzymology , Stroke/pathology
16.
Biochem J ; 460(1): 25-34, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24762137

ABSTRACT

Growth factors inactivate the FOXO (forkhead box O) transcription factors through PI3K (phosphoinositide 3-kinase) and PKB (protein kinase B). By comparing microarray data from multiple model systems, we identified HBP1 (high-mobility group-box protein 1) as a novel downstream target of this pathway. HBP1 mRNA was down-regulated by PDGF (platelet-derived growth factor), FGF (fibroblast growth factor), PI3K and PKB, whereas it was up-regulated by FOXO factors. This observation was confirmed in human and murine fibroblasts as well as in cell lines derived from leukaemia, breast adenocarcinoma and colon carcinoma. Bioinformatics analysis led to the identification of a conserved consensus FOXO-binding site in the HBP1 promoter. By luciferase activity assay and ChIP, we demonstrated that FOXO bound to this site and regulated the HBP1 promoter activity in a PI3K-dependent manner. Silencing of HBP1 by shRNA increased the proliferation of human fibroblasts in response to growth factors, suggesting that HBP1 limits cell growth. Finally, by analysing a transcriptomics dataset from The Cancer Genome Atlas, we observed that HBP1 expression was lower in breast tumours that had lost FOXO expression. In conclusion, HBP1 is a novel target of the PI3K/FOXO pathway and controls cell proliferation in response to growth factors.


Subject(s)
Down-Regulation/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Animals , CHO Cells , Cells, Cultured , Conserved Sequence , Cricetinae , Cricetulus , Fibroblasts/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , HEK293 Cells , High Mobility Group Proteins/biosynthesis , Humans , MCF-7 Cells , Male , Mice , NIH 3T3 Cells , Phosphatidylinositol 3-Kinase/biosynthesis , Promoter Regions, Genetic , Protein Binding/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Repressor Proteins/biosynthesis , Signal Transduction/genetics
17.
Mod Pathol ; 26(5): 685-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23328977

ABSTRACT

Well-differentiated small intestinal neuroendocrine tumors are rare malignancies. They arise from enterochromaffin cells and very little is known about differential microRNA (miRNA) expression. The aim of this study was to identify the miRNA profile of well-differentiated small intestinal neuroendocrine tumors, which may have a critical role in tumor development, progression and potentially develop miRNAs as novel clinical biomarkers. Specimens from two test groups, 24 small intestinal neuroendocrine tumor specimens at different stages of malignancy, are included in this study. Total RNA from the first test group, five primary tumors, five mesentery metastases and five liver metastases was hybridized onto the Affymetrix Genechip miRNA arrays to perform a genome-wide profile. The results were validated by using quantitative real-time PCR (QRT-PCR) and northern blot analyses. We then expanded the investigation to laser capture microdissected small intestinal neuroendocrine tumor cells and immuno-laser capture microdissected normal enterochromaffin cells of the first test group. Furthermore, a second test group, three primary tumors, three mesentery metastases and three liver metastases, was included in the study. Thus, two independent test groups validated the data by QRT-PCR. Moreover, we characterized nine miRNAs, five (miR-96, -182, -183, -196a and -200a), which are upregulated during tumor progression, whereas four (miR-31, -129-5p, -133a and -215) are downregulated. Several online software programs were used to predict potential miRNA target genes to map a number of putative target genes for the aberrantly regulated miRNAs, through an advanced and novel bioinformatics analysis. Our findings provide information about pivotal miRNAs, which may lead to further insights into tumorigenesis, progression mechanisms and novel therapeutic targets recognition.


Subject(s)
Biomarkers, Tumor/genetics , Intestinal Neoplasms/genetics , MicroRNAs/analysis , Neuroendocrine Tumors/genetics , Adult , Aged , Aged, 80 and over , Blotting, Northern , Female , Gene Expression Profiling , Humans , Intestinal Neoplasms/pathology , Laser Capture Microdissection , Male , Middle Aged , Neuroendocrine Tumors/pathology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
18.
Mol Nutr Food Res ; 57(2): 347-59, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23203768

ABSTRACT

SCOPE: Recent data suggest that gut microbiota contributes to the regulation of host lipid metabolism. We report how fermentable dietary fructo-oligosaccharides (FOS) control hepatic steatosis induced by n-3 PUFA depletion, which leads to hepatic alterations similar to those observed in non-alcoholic fatty liver disease patients. METHODS AND RESULTS: C57Bl/6J mice fed an n-3 PUFA-depleted diet for 3 months were supplemented with FOS during the last 10 days of treatment. FOS-treated mice exhibited higher caecal Bifidobacterium spp. and lower Roseburia spp. content. Microarray analysis of hepatic mRNA revealed that FOS supplementation reduced hepatic triglyceride accumulation through a proliferator-activated receptor α-stimulation of fatty acid oxidation and lessened cholesterol accumulation by inhibiting sterol regulatory element binding protein 2-dependent cholesterol synthesis. Cultured precision-cut liver slices confirmed the inhibition of fatty acid oxidation. FOS effects were related to a decreased hepatic micro-RNA33 expression and to an increased colonic glucagon-like peptide 1 production. CONCLUSIONS: The changes in gut microbiota composition by n-3 PUFA-depletion and prebiotics modulate hepatic steatosis by changing gene expression in the liver, a phenomenon that could implicate micro-RNA and gut-derived hormones. Our data underline the advantage of targeting the gut microbiota by colonic nutrients in the management of liver disease.


Subject(s)
Cholesterol/biosynthesis , Dietary Supplements , Fatty Acids, Omega-3/metabolism , Fatty Liver/pathology , Prebiotics , Animals , Bifidobacterium/growth & development , Energy Intake , Fatty Liver/metabolism , Gastrointestinal Tract/microbiology , Gene Expression Regulation , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Lipid Metabolism , Liver/metabolism , Male , Metagenome/physiology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Oligosaccharides/administration & dosage , Oxidative Stress/drug effects , PPAR alpha/genetics , PPAR alpha/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
PLoS One ; 7(10): e48411, 2012.
Article in English | MEDLINE | ID: mdl-23119007

ABSTRACT

Octreotide is a widely used synthetic somatostatin analogue that significantly improves the management of neuroendocrine tumours (NETs). Octreotide acts through somatostatin receptors (SSTRs). However, the molecular mechanisms leading to successful disease control or symptom management, especially when SSTRs levels are low, are largely unknown. We provide novel insights into how octreotide controls NET cells. CNDT2.5 cells were treated from 1 day up to 16 months with octreotide and then were profiled using Affymetrix microarray analysis. Quantitative real-time PCR and western blot analyses were used to validate microarray profiling in silico data. WST-1 cell proliferation assay was applied to evaluate cell growth of CNDT2.5 cells in the presence or absence of 1 µM octreotide at different time points. Moreover, laser capture microdissected tumour cells and paraffin embedded tissue slides from SI-NETs at different stages of disease were used to identify transcriptional and translational expression. Microarrays analyses did not reveal relevant changes in SSTR expression levels. Unexpectedly, six novel genes were found to be upregulated by octreotide: annexin A1 (ANXA1), rho GTPase-activating protein 18 (ARHGAP18), epithelial membrane protein 1 (EMP1), growth/differentiation factor 15 (GDF15), TGF-beta type II receptor (TGFBR2) and tumour necrosis factor (ligand) superfamily member 15 (TNFSF15). Furthermore, these novel genes were expressed in tumour tissues at transcript and protein levels. We suggest that octreotide may use a potential novel framework to exert its beneficial effect as a drug and to convey its action on neuroendocrine cells. Thus, six novel genes may regulate cell growth and differentiation in normal and tumour neuroendocrine cells and have a role in a novel octreotide mechanism system.


Subject(s)
Antineoplastic Agents/pharmacology , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Neuroendocrine Tumors/pathology , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Neoplasm Staging , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Somatostatin/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcriptome/drug effects , Up-Regulation/drug effects
20.
PLoS One ; 7(6): e39666, 2012.
Article in English | MEDLINE | ID: mdl-22761861

ABSTRACT

A universal cancer biomarker candidate for diagnosis is supposed to distinguish, within a broad range of tumors, between healthy and diseased patients. Recently published studies have explored the universal usefulness of some biomarkers in human tumors. In this study, we present an integrative approach to search for potential common cancer biomarkers. Using the TFactS web-tool with a catalogue of experimentally established gene regulations, we could predict transcription factors (TFs) regulated in 305 different human cancer cell lines covering a large panel of tumor types. We also identified chromosomal regions having significant copy number variation (CNV) in these cell lines. Within the scope of TFactS catalogue, 88 TFs whose activity status were explained by their gene expressions and CNVs were identified. Their minimal connected network (MCN) of protein-protein interactions forms a significant module within the human curated TF proteome. Functional analysis of the proteins included in this MCN revealed enrichment in cancer pathways as well as inflammation. The ten most central proteins in MCN are TFs that trans-regulate 157 known genes encoding secreted and transmembrane proteins. In publicly available collections of gene expression data from 8,525 patient tissues, 86 genes were differentially regulated in cancer compared to inflammatory diseases and controls. From TCGA cancer gene expression data sets, 50 genes were significantly associated to patient survival in at least one tumor type. Enrichment analysis shows that these genes mechanistically interact in common cancer pathways. Among these cancer biomarker candidates, TFRC, MET and VEGFA are commonly amplified genes in tumors and their encoded proteins stained positive in more than 80% of malignancies from public databases. They are linked to angiogenesis and hypoxia, which are common in cancer. They could be interesting for further investigations in cancer diagnostic strategies.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Transcription Factors/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Gene Dosage , Gene Expression Regulation , Humans , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Survival Analysis , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...