Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Comput Biol ; 17(6): e1009069, 2021 06.
Article in English | MEDLINE | ID: mdl-34166365

ABSTRACT

Despite the unprecedented growth in our understanding of cell biology, it still remains challenging to connect it to experimental data obtained with cells and tissues' physiopathological status under precise circumstances. This knowledge gap often results in difficulties in designing validation experiments, which are usually labor-intensive, expensive to perform, and hard to interpret. Here we propose PHENSIM, a computational tool using a systems biology approach to simulate how cell phenotypes are affected by the activation/inhibition of one or multiple biomolecules, and it does so by exploiting signaling pathways. Our tool's applications include predicting the outcome of drug administration, knockdown experiments, gene transduction, and exposure to exosomal cargo. Importantly, PHENSIM enables the user to make inferences on well-defined cell lines and includes pathway maps from three different model organisms. To assess our approach's reliability, we built a benchmark from transcriptomics data gathered from NCBI GEO and performed four case studies on known biological experiments. Our results show high prediction accuracy, thus highlighting the capabilities of this methodology. PHENSIM standalone Java application is available at https://github.com/alaimos/phensim, along with all data and source codes for benchmarking. A web-based user interface is accessible at https://phensim.tech/.


Subject(s)
Algorithms , Cell Physiological Phenomena , Phenotype , Software , Antineoplastic Agents/pharmacology , Benchmarking , Cell Biology , Cell Line , Cell Line, Tumor , Computational Biology , Computer Simulation , Female , Gene Expression Profiling/statistics & numerical data , Humans , MAP Kinase Kinase Kinases/genetics , Metformin/pharmacology , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , Synthetic Lethal Mutations , Systems Biology , Tumor Necrosis Factor-alpha/genetics
2.
Proc Natl Acad Sci U S A ; 116(28): 14039-14048, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31239343

ABSTRACT

Most normal and tumor cells are protected from tumor necrosis factor α (TNFα)-induced apoptosis. Here, we identify the MAP3 kinase tumor progression locus-2 (TPL2) as a player contributing to the protection of a subset of tumor cell lines. The combination of TPL2 knockdown and TNFα gives rise to a synthetic lethality phenotype via receptor-interacting serine/threonine-protein kinase 1 (RIPK1)-dependent and -independent mechanisms. Whereas wild-type TPL2 rescues the phenotype, its kinase-dead mutant does not. Comparison of the molecular events initiated by small interfering RNA for TPL2 (siTPL2) ± TNFα in treatment-sensitive and -resistant lines revealed that the activation of caspase-8, downstream of miR-21-5p and cFLIP, is the dominant TPL2-dependent event. More important, comparison of the gene expression profiles of all of the tested cell lines results in the clustering of sensitive and resistant lines into distinct groups, providing proof of principle for the feasibility of generating a predictive tool for treatment sensitivity.


Subject(s)
Carcinoma/genetics , Caspase Inhibitors/pharmacology , MAP Kinase Kinase Kinases/genetics , Proto-Oncogene Proteins/genetics , Tumor Necrosis Factor-alpha/genetics , Apoptosis/genetics , Carcinoma/drug therapy , Carcinoma/pathology , Caspase 8/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , HeLa Cells , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Macrophages/metabolism , MicroRNAs/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , RNA, Small Interfering/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Synthetic Lethal Mutations/genetics
3.
Gastroenterology ; 149(4): 981-92.e11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26055138

ABSTRACT

BACKGROUND & AIMS: Persistent activation of the inflammatory response contributes to the development of inflammatory bowel diseases, which increase the risk of colorectal cancer. We aimed to identify microRNAs that regulate inflammation during the development of ulcerative colitis (UC) and progression to colitis-associated colon cancer (CAC). METHODS: We performed a quantitative polymerase chain reaction analysis to measure microRNAs in 401 colon specimens from patients with UC, Crohn's disease, irritable bowel syndrome, sporadic colorectal cancer, or CAC, as well as subjects without these disorders (controls); levels were correlated with clinical features and disease activity of patients. Colitis was induced in mice by administration of dextran sodium sulfate (DSS), and carcinogenesis was induced by addition of azoxymethane; some mice also were given an inhibitor of microRNA214 (miR214). RESULTS: A high-throughput functional screen of the human microRNAome found that miR214 regulated the activity of nuclear factor-κB. Higher levels of miR214 were detected in colon tissues from patients with active UC or CAC than from patients with other disorders or controls and correlated with disease progression. Bioinformatic and genome-wide profile analyses showed that miR214 activates an inflammatory response and is amplified through a feedback loop circuit mediated by phosphatase and tensin homolog (PTEN) and PDZ and LIM domain 2 (PDLIM2). Interleukin-6 induced signal transducer and activator of transcription 3 (STAT3)-mediated transcription of miR214. A miR214 chemical inhibitor blocked this circuit and reduced the severity of DSS-induced colitis in mice, as well as the number and size of tumors that formed in mice given azoxymethane and DSS. In fresh colonic biopsy specimens from patients with active UC, the miR214 inhibitor reduced inflammation by increasing levels of PDLIM2 and PTEN. CONCLUSIONS: Interleukin-6 up-regulates STAT3-mediated transcription of miR214 in colon tissues, which reduces levels of PDLIM2 and PTEN, increases phosphorylation of AKT, and activates nuclear factor-κB. The activity of this circuit correlates with disease activity in patients with UC and progression to colorectal cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Colitis, Ulcerative/prevention & control , Colon/metabolism , Colonic Neoplasms/prevention & control , MicroRNAs/metabolism , RNAi Therapeutics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Azoxymethane , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Line , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dextran Sulfate , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Inflammation Mediators/metabolism , Interleukin-6/metabolism , LIM Domain Proteins/metabolism , Mice , MicroRNAs/genetics , NF-kappa B/metabolism , PTEN Phosphohydrolase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic , Transfection , Tumor Cells, Cultured
4.
Proc Natl Acad Sci U S A ; 109(18): E1082-91, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22451924

ABSTRACT

To address the role of Tpl2, a MAP3K8 that regulates innate/adaptive immunity and inflammation, in intestinal tumorigenesis, we crossed a Tpl2 KO allele into the Apc(min/+) genetic background. Here, we show that Apc(min/+)/Tpl2(-/-) mice exhibit a fivefold increase in the number of intestinal adenomas. Bone marrow transplantation experiments revealed that the enhancement of polyposis was partially hematopoietic cell-driven. Consistent with this observation, Tpl2 ablation promoted intestinal inflammation. IL-10 levels and regulatory T-cell numbers were lower in the intestines of Tpl2(-/-) mice, independent of Apc and polyp status, suggesting that they were responsible for the initiation of the enhancement of tumorigenesis caused by the ablation of Tpl2. The low IL-10 levels correlated with defects in mTOR activation and Stat3 phosphorylation in Toll-like receptor-stimulated macrophages and with a defect in inducible regulatory T-cell generation and function. Both polyp numbers and inflammation increased progressively with time. The rate of increase of both, however, was more rapid in Apc(min/+)/Tpl2(-/-) mice, suggesting that the positive feedback initiated by inflammatory signals originating in developing polyps is more robust in these mice. This may be because these mice have a higher intestinal polyp burden as a result of the enhancement of tumor initiation.


Subject(s)
Genes, APC , Inflammatory Bowel Diseases/etiology , Interleukin-10/biosynthesis , Intestinal Neoplasms/etiology , MAP Kinase Kinase Kinases/deficiency , Proto-Oncogene Proteins/deficiency , T-Lymphocytes, Regulatory/immunology , Adenoma/etiology , Adenoma/genetics , Adenoma/immunology , Animals , Bone Marrow Transplantation , Female , Gene Expression , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/immunology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Models, Immunological , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology
5.
Proc Natl Acad Sci U S A ; 105(6): 1907-12, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18250326

ABSTRACT

A common integration site, cloned from MoMuLV-induced rat T cell lymphomas, was mapped immediately upstream of Not dead yet-1 (Ndy1)/KDM2B, a gene expressed primarily in testis, spleen, and thymus, that is also known as FBXL10 or JHDM1B. Ndy1 encodes a nuclear, chromatin-associated protein that harbors Jumonji C (JmjC), CXXC, PHD, proline-rich, F-box, and leucine-rich repeat domains. Ndy1 and its homolog Ndy2/KDM2A (FBXL11 or JHDM1A), which is also a target of provirus integration in retrovirus-induced lymphomas, encode proteins that were recently shown to possess Jumonji C-dependent histone H3 K36 dimethyl-demethylase or histone H3 K4 trimethyl-demethylase activities. Here, we show that mouse embryo fibroblasts engineered to express Ndy1 or Ndy2 undergo immortalization in the absence of replicative senescence via a JmjC domain-dependent process that targets the Rb and p53 pathways. Knockdown of endogenous Ndy1 or expression of JmjC domain mutants of Ndy1 promote senescence, suggesting that Ndy1 is a physiological inhibitor of senescence in dividing cells and that inhibition of senescence depends on histone H3 demethylation.


Subject(s)
Cell Transformation, Neoplastic , Embryo, Mammalian/chemistry , Oncogene Proteins/physiology , Amino Acid Sequence , Animals , Animals, Newborn , Blotting, Northern , Blotting, Western , Cell Transformation, Viral , Fibroblasts/cytology , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/virology , Mice , Molecular Sequence Data , Moloney murine leukemia virus/physiology , Rats , Rats, Inbred F344 , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...