Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell ; 186(23): 5068-5083.e23, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37804830

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. However, mechanisms underlying metabolic reprogramming and how altered metabolism in turn enhances tumorigenicity are poorly understood. Here, we report that arginine levels are elevated in murine and patient hepatocellular carcinoma (HCC), despite reduced expression of arginine synthesis genes. Tumor cells accumulate high levels of arginine due to increased uptake and reduced arginine-to-polyamine conversion. Importantly, the high levels of arginine promote tumor formation via further metabolic reprogramming, including changes in glucose, amino acid, nucleotide, and fatty acid metabolism. Mechanistically, arginine binds RNA-binding motif protein 39 (RBM39) to control expression of metabolic genes. RBM39-mediated upregulation of asparagine synthesis leads to enhanced arginine uptake, creating a positive feedback loop to sustain high arginine levels and oncogenic metabolism. Thus, arginine is a second messenger-like molecule that reprograms metabolism to promote tumor growth.


Subject(s)
Arginine , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Arginine/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Lipid Metabolism , Liver Neoplasms/metabolism
2.
Mol Cell ; 82(22): 4246-4261.e11, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36400009

ABSTRACT

Acetyl-coenzyme A (acetyl-CoA) plays an important role in metabolism, gene expression, signaling, and other cellular processes via transfer of its acetyl group to proteins and metabolites. However, the synthesis and usage of acetyl-CoA in disease states such as cancer are poorly characterized. Here, we investigated global acetyl-CoA synthesis and protein acetylation in a mouse model and patient samples of hepatocellular carcinoma (HCC). Unexpectedly, we found that acetyl-CoA levels are decreased in HCC due to transcriptional downregulation of all six acetyl-CoA biosynthesis pathways. This led to hypo-acetylation specifically of non-histone proteins, including many enzymes in metabolic pathways. Importantly, repression of acetyl-CoA synthesis promoted oncogenic dedifferentiation and proliferation. Mechanistically, acetyl-CoA synthesis was repressed by the transcription factors TEAD2 and E2A, previously unknown to control acetyl-CoA synthesis. Knockdown of TEAD2 and E2A restored acetyl-CoA levels and inhibited tumor growth. Our findings causally link transcriptional reprogramming of acetyl-CoA metabolism, dedifferentiation, and cancer.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Acetyl Coenzyme A/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Histones/metabolism , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Carcinogenesis/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
3.
PLoS Comput Biol ; 18(9): e1009767, 2022 09.
Article in English | MEDLINE | ID: mdl-36067230

ABSTRACT

Comprehensive molecular characterization of cancer subtypes is essential for predicting clinical outcomes and searching for personalized treatments. We present bnClustOmics, a statistical model and computational tool for multi-omics unsupervised clustering, which serves a dual purpose: Clustering patient samples based on a Bayesian network mixture model and learning the networks of omics variables representing these clusters. The discovered networks encode interactions among all omics variables and provide a molecular characterization of each patient subgroup. We conducted simulation studies that demonstrated the advantages of our approach compared to other clustering methods in the case where the generative model is a mixture of Bayesian networks. We applied bnClustOmics to a hepatocellular carcinoma (HCC) dataset comprising genome (mutation and copy number), transcriptome, proteome, and phosphoproteome data. We identified three main HCC subtypes together with molecular characteristics, some of which are associated with survival even when adjusting for the clinical stage. Cluster-specific networks shed light on the links between genotypes and molecular phenotypes of samples within their respective clusters and suggest targets for personalized treatments.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Bayes Theorem , Carcinoma, Hepatocellular/genetics , Cluster Analysis , Humans , Liver Neoplasms/genetics , Proteome , Transcriptome
4.
Nat Commun ; 13(1): 2436, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508466

ABSTRACT

Proteogenomic analyses of hepatocellular carcinomas (HCC) have focused on early-stage, HBV-associated HCCs. Here we present an integrated proteogenomic analysis of HCCs across clinical stages and etiologies. Pathways related to cell cycle, transcriptional and translational control, signaling transduction, and metabolism are dysregulated and differentially regulated on the genomic, transcriptomic, proteomic and phosphoproteomic levels. We describe candidate copy number-driven driver genes involved in epithelial-to-mesenchymal transition, the Wnt-ß-catenin, AKT/mTOR and Notch pathways, cell cycle and DNA damage regulation. The targetable aurora kinase A and CDKs are upregulated. CTNNB1 and TP53 mutations are associated with altered protein phosphorylation related to actin filament organization and lipid metabolism, respectively. Integrative proteogenomic clusters show that HCC constitutes heterogeneous subgroups with distinct regulation of biological processes, metabolic reprogramming and kinase activation. Our study provides a comprehensive overview of the proteomic and phophoproteomic landscapes of HCCs, revealing the major pathways altered in the (phospho)proteome.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Proteogenomics , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/metabolism , Mutation , Proteomics , beta Catenin/metabolism
5.
Oncogenesis ; 10(7): 52, 2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34272356

ABSTRACT

Understanding the mechanisms underlying evasive resistance in cancer is an unmet medical need to improve the efficacy of current therapies. In hepatocellular carcinoma (HCC), aberrant expression of hypoxia-inducible factor 1 α (HIF1α) and increased aerobic glycolysis metabolism are drivers of resistance to therapy with the multi-kinase inhibitor Sorafenib. However, it has remained unknown how HIF1α is activated and how its activity and the subsequent induction of aerobic glycolysis promote Sorafenib resistance in HCC. Here, we report the ubiquitin-specific peptidase USP29 as a new regulator of HIF1α and of aerobic glycolysis during the development of Sorafenib resistance in HCC. In particular, we identified USP29 as a critical deubiquitylase (DUB) of HIF1α, which directly deubiquitylates and stabilizes HIF1α and, thus, promotes its transcriptional activity. Among the transcriptional targets of HIF1α is the gene encoding hexokinase 2 (HK2), a key enzyme of the glycolytic pathway. The absence of USP29, and thus of HIF1α transcriptional activity, reduces the levels of aerobic glycolysis and restores sensitivity to Sorafenib in Sorafenib-resistant HCC cells in vitro and in xenograft transplantation mouse models in vivo. Notably, the absence of USP29 and high HK2 expression levels correlate with the response of HCC patients to Sorafenib therapy. Together, the data demonstrate that, as a DUB of HIF1α, USP29 promotes Sorafenib resistance in HCC cells, in parts by upregulating glycolysis, thereby opening new avenues for therapeutically targeting Sorafenib-resistant HCC in patients.

6.
Nat Commun ; 10(1): 5755, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848340

ABSTRACT

Autophagy perturbation represents an emerging therapeutic strategy in cancer. Although LATS1 and LATS2 kinases, core components of the mammalian Hippo pathway, have been shown to exert tumor suppressive activities, here we report a pro-survival role of LATS1 but not LATS2 in hepatocellular carcinoma (HCC) cells. Specifically, LATS1 restricts lethal autophagy in HCC cells induced by sorafenib, the standard of care for advanced HCC patients. Notably, autophagy regulation by LATS1 is independent of its kinase activity. Instead, LATS1 stabilizes the autophagy core-machinery component Beclin-1 by promoting K27-linked ubiquitination at lysine residues K32 and K263 on Beclin-1. Consequently, ubiquitination of Beclin-1 negatively regulates autophagy by promoting inactive dimer formation of Beclin-1. Our study highlights a functional diversity between LATS1 and LATS2, and uncovers a scaffolding role of LATS1 in mediating a cross-talk between the Hippo signaling pathway and autophagy.


Subject(s)
Autophagy/immunology , Carcinoma, Hepatocellular/pathology , Cell Survival/immunology , Liver Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Autophagy/drug effects , Beclin-1/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Datasets as Topic , Disease-Free Survival , Drug Resistance, Neoplasm/immunology , Hippo Signaling Pathway , Humans , Kaplan-Meier Estimate , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Lysine/metabolism , Mice , Mice, Knockout , Organoids , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Stability , Signal Transduction/drug effects , Signal Transduction/immunology , Sorafenib/pharmacology , Sorafenib/therapeutic use , Tumor Suppressor Proteins/immunology , Ubiquitination , Xenograft Model Antitumor Assays
7.
Hypertension ; 73(2): 469-480, 2019 02.
Article in English | MEDLINE | ID: mdl-30580688

ABSTRACT

Primary aldosteronism is a disease of excessive production of adrenal steroid hormones and the most common cause of endocrine hypertension. Primary aldosteronism results mainly from bilateral adrenal hyperplasia or unilateral aldosterone-producing adenoma (APA). Primary aldosteronism cause at the molecular level is incompletely understood and a targeted treatment preventing excessive adrenal steroid production is not available. Here, we perform deep quantitative proteomic and phosphoproteomic profiling of 6 pairs of APA and adjacent nontumoral adrenal cortex. We show that increased steroidogenesis in APA is accompanied by upregulation of steroidogenic enzymes (HSD3B2, CYP21A2, CYP11B2) and of proteins involved in cholesterol uptake (LSR). We demonstrate that HSD3B2 is phosphorylated at Ser95 or 96 and identify a novel phosphorylation site, Ser489, in CYP21A2, suggesting that steroidogenic enzymes are regulated by phosphorylation. Our analysis also reveals altered ECM (extracellular matrix) composition in APA that affects ECM-cell surface interactions and actin cytoskeleton rearrangements. We show that RHOC, a GTPase controlling actin organization in response to extracellular stimuli, is upregulated in APA and promotes expression of the aldosterone synthase gene CYP11B2. Our data also indicate deregulation of protein N-glycosylation and GABAergic signaling in APAs. Finally, we find that mTORC1 (mammalian target of rapamycin complex 1) signaling is the major pathway deregulated in APA. Our study provides a rich resource for future research on the molecular mechanisms of primary aldosteronism.


Subject(s)
Adenoma/metabolism , Aldosterone/biosynthesis , Proteomics/methods , Extracellular Matrix/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/physiology , Signal Transduction , gamma-Aminobutyric Acid/metabolism
8.
Cancer Cell ; 32(6): 807-823.e12, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29232555

ABSTRACT

Dysregulated mammalian target of rapamycin (mTOR) promotes cancer, but underlying mechanisms are poorly understood. We describe an mTOR-driven mouse model that displays hepatosteatosis progressing to hepatocellular carcinoma (HCC). Longitudinal proteomic, lipidomics, and metabolomic analyses revealed that hepatic mTORC2 promotes de novo fatty acid and lipid synthesis, leading to steatosis and tumor development. In particular, mTORC2 stimulated sphingolipid (glucosylceramide) and glycerophospholipid (cardiolipin) synthesis. Inhibition of fatty acid or sphingolipid synthesis prevented tumor development, indicating a causal effect in tumorigenesis. Increased levels of cardiolipin were associated with tubular mitochondria and enhanced oxidative phosphorylation. Furthermore, increased lipogenesis correlated with elevated mTORC2 activity and HCC in human patients. Thus, mTORC2 promotes cancer via formation of lipids essential for growth and energy production.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Fatty Liver/metabolism , Lipogenesis/physiology , Liver Neoplasms/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Animals , Carcinoma, Hepatocellular/etiology , Cell Transformation, Neoplastic/metabolism , Fatty Liver/complications , Humans , Lipids/biosynthesis , Liver Neoplasms/etiology , Mice , Mice, Knockout
9.
Proc Natl Acad Sci U S A ; 113(5): 1381-6, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787912

ABSTRACT

Compensatory signaling pathways in tumors confer resistance to targeted therapy, but the pathways and their mechanisms of activation remain largely unknown. We describe a procedure for quantitative proteomics and phosphoproteomics on snap-frozen biopsies of hepatocellular carcinoma (HCC) and matched nontumor liver tissue. We applied this procedure to monitor signaling pathways in serial biopsies taken from an HCC patient before and during treatment with the multikinase inhibitor sorafenib. At diagnosis, the patient had an advanced HCC. At the time of the second biopsy, abdominal imaging revealed progressive disease despite sorafenib treatment. Sorafenib was confirmed to inhibit MAPK signaling in the tumor, as measured by reduced ribosomal protein S6 kinase phosphorylation. Hierarchical clustering and enrichment analysis revealed pathways broadly implicated in tumor progression and resistance, such as epithelial-to-mesenchymal transition and cell adhesion pathways. Thus, we describe a protocol for quantitative analysis of oncogenic pathways in HCC biopsies and obtained first insights into the effect of sorafenib in vivo. This protocol will allow elucidation of mechanisms of resistance and enable precision medicine.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Phosphoproteins/metabolism , Proteomics , Biopsy , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Niacinamide/therapeutic use , Phosphorylation , Sorafenib
10.
Gut ; 65(12): 2017-2028, 2016 12.
Article in English | MEDLINE | ID: mdl-26276683

ABSTRACT

OBJECTIVE: Chronically HCV-infected orthotopic liver transplantation (OLT) recipients appear to have improved outcomes when their immunosuppressive regimen includes a mammalian target of rapamycin (mTOR) inhibitor. The mechanism underlying this observation is unknown. DESIGN: We used virological assays to investigate mTOR signalling on the HCV replication cycle. Furthermore, we analysed HCV RNA levels of 42 HCV-positive transplanted patients treated with an mTOR inhibitor as part of their immunosuppressive regimen. RESULTS: The mTOR inhibitor rapamycin was found to be a potent inhibitor for HCV RNA replication in Huh-7.5 cells as well as primary human hepatocytes. Half-maximal inhibition was observed at 0.01 µg/mL, a concentration that is in the range of serum levels seen in transplant recipients and does not affect cell proliferation. Early replication cycle steps such as cell entry and RNA translation were not affected. Knockdown of raptor, an essential component of mTORC1, but not rictor, an essential component of mTORC2, inhibited viral RNA replication. In addition, overexpression of raptor led to higher viral RNA replication, demonstrating that mTORC1, but not mTORC2, is required for HCV RNA replication. In 42 HCV-infected liver-transplanted or kidney-transplanted patients who were switched to an mTOR inhibitor, we could verify that mTOR inhibition decreased HCV RNA levels in vivo. CONCLUSIONS: Our data identify mTORC1 as a novel HCV replication factor. These findings suggest an underlying mechanism for the observed benefits of mTOR inhibition in HCV-positive OLT recipients and potentiate further investigation of mTOR-containing regimens in HCV-positive recipients of solid organ transplants.


Subject(s)
Hepacivirus/drug effects , Hepatocytes/drug effects , Multiprotein Complexes/pharmacology , RNA, Viral/drug effects , TOR Serine-Threonine Kinases/pharmacology , Virus Replication/drug effects , Gene Silencing/drug effects , Hepatitis C, Chronic/therapy , Humans , Liver Transplantation , Mechanistic Target of Rapamycin Complex 1 , Treatment Outcome
11.
Proc Natl Acad Sci U S A ; 111(49): 17582-7, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25422452

ABSTRACT

The inflamed tumor microenvironment plays a critical role in tumorigenesis. However, the mechanisms through which immune cells, particularly macrophages, promote tumorigenesis have only been partially elucidated, and the full scope of signaling pathways supplying macrophages with protumorigenic phenotypes still remain largely unknown. Here we report that germ-line absence of c-Jun N-terminal phosphorylation at serines 63 and 73 impedes inflammation-associated hepatocarcinogenesis, yet deleting c-Jun only in hepatocytes does not inhibit hepatocellular carcinoma (HCC) formation. Moreover, in human HCC-bearing livers, c-Jun phosphorylation is found in inflammatory cells, whereas it is mostly absent from malignant hepatocytes. Interestingly, macrophages in livers of mice with chronic hepatitis gradually switch their phenotype along the course of disease. Macrophage phenotype and density are dictated by c-Jun phosphorylation, in vitro and in vivo. Transition of macrophage phenotype, from antitumorigenic to protumorigenic, occurs before tumorigenesis, resulting in the production of various chemokines, including chemokine (C-C motif) ligand 17 (CCL17) and CCL22. Such signals, emanating from the liver microenvironment, direct the recruitment of regulatory T cells, which are known to facilitate HCC growth. Our findings identify c-Jun phosphorylation as a key mediator of macrophage education and point to the recruitment of immunosuppressive regulatory T cells as a possible protumorigenic mechanism.


Subject(s)
Macrophages/cytology , Macrophages/immunology , Proto-Oncogene Proteins c-jun/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Chemokines/metabolism , Gene Expression Regulation, Neoplastic , Hepatitis/metabolism , Hepatocytes/cytology , Humans , Immunity, Innate , Inflammation , Liver/metabolism , Liver Neoplasms/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Phenotype , Phosphorylation , Prognosis , Protein Structure, Tertiary , Tumor Microenvironment
12.
Cell Host Microbe ; 12(1): 71-85, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22817989

ABSTRACT

Virus infection-induced global protein synthesis suppression is linked to assembly of stress granules (SGs), cytosolic aggregates of stalled translation preinitiation complexes. To study long-term stress responses, we developed an imaging approach for extended observation and analysis of SG dynamics during persistent hepatitis C virus (HCV) infection. In combination with type 1 interferon, HCV infection induces highly dynamic assembly/disassembly of cytoplasmic SGs, concomitant with phases of active and stalled translation, delayed cell division, and prolonged cell survival. Double-stranded RNA (dsRNA), independent of viral replication, is sufficient to trigger these oscillations. Translation initiation factor eIF2α phosphorylation by protein kinase R mediates SG formation and translation arrest. This is antagonized by the upregulation of GADD34, the regulatory subunit of protein phosphatase 1 dephosphorylating eIF2α. Stress response oscillation is a general mechanism to prevent long-lasting translation repression and a conserved host cell reaction to multiple RNA viruses, which HCV may exploit to establish persistence.


Subject(s)
Cytoplasmic Granules/metabolism , Hepacivirus/pathogenicity , Hepatitis C/pathology , Host-Pathogen Interactions , Protein Biosynthesis , Cell Division , Cell Line , Cytoplasmic Granules/drug effects , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C/virology , Humans , Interferon-alpha/metabolism , Interferon-alpha/pharmacology , Liver/cytology , Liver/virology , Protein Phosphatase 1/metabolism , RNA, Double-Stranded/metabolism , Virus Replication/genetics , eIF-2 Kinase/metabolism
13.
Hepatology ; 56(6): 2082-93, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22711689

ABSTRACT

UNLABELLED: Persistent infection with hepatitis C virus (HCV) can lead to chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. All current therapies of hepatitis C include interferon-alpha (IFN-α). Moreover, IFN-gamma (IFN-γ), the only type II IFN, strongly inhibits HCV replication in vitro and is the primary mediator of HCV-specific antiviral T-cell responses. However, for both cytokines the precise set of effector protein(s) responsible for replication inhibition is not known. The aim of this study was the identification of IFN-α and IFN-γ stimulated genes (ISGs) responsible for controlling HCV replication. We devised an RNA interference (RNAi)-based "gain of function" screen and identified, in addition to known ISGs earlier reported to suppress HCV replication, several new ones with proven antiviral activity. These include IFIT3 (IFN-induced protein with tetratricopeptide repeats 3), TRIM14 (tripartite motif containing 14), PLSCR1 (phospholipid scramblase 1), and NOS2 (nitric oxide synthase 2, inducible). All ISGs identified in this study were up-regulated both by IFN-α and IFN-γ, demonstrating a substantial overlap of HCV-specific effectors induced by either cytokine. Nevertheless, some ISGs were more specific for IFN-α or IFN-γ, which was most pronounced in case of PLSCR1 and NOS2 that were identified as main effectors of IFN-γ-mediated anti-HCV activity. Combinatorial knockdowns of ISGs suggest additive or synergistic effects demonstrating that with either IFN, inhibition of HCV replication is caused by the combined action of multiple ISGs. CONCLUSION: Our study identifies a number of novel ISGs contributing to the suppression of HCV replication by type I and type II IFN. We demonstrate a substantial overlap of antiviral programs triggered by either cytokine and show that suppression of HCV replication is mediated by the concerted action of multiple effectors.


Subject(s)
Hepacivirus/physiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Virus Replication , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Replicon , Tripartite Motif Proteins , Tumor Cells, Cultured , Up-Regulation/drug effects
14.
Curr Opin Cell Biol ; 23(6): 744-55, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21963299

ABSTRACT

The target of rapamycin (TOR) is a highly conserved serine/threonine kinase and a central controller of cell growth, metabolism and aging. Mammalian TOR (mTOR) is activated in response to nutrients, growth factors and cellular energy. Dysregulated mTOR signaling has been implicated in major disease. Here we review recent findings on the role of mTOR in cancer, metabolic disorders, neurological diseases, and inflammation.


Subject(s)
Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Humans , Mammals/metabolism , Neoplasms/metabolism
15.
Bioinformatics ; 25(17): 2229-35, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19542154

ABSTRACT

MOTIVATION: The reconstruction of signaling pathways from gene knockdown data is a novel research field enabled by developments in RNAi screening technology. However, while RNA interference is a powerful technique to identify genes related to a phenotype of interest, their placement in the corresponding pathways remains a challenging problem. Difficulties are aggravated if not all pathway components can be observed after each knockdown, but readouts are only available for a small subset. We are then facing the problem of reconstructing a network from incomplete data. RESULTS: We infer pathway topologies from gene knockdown data using Bayesian networks with probabilistic Boolean threshold functions. To deal with the problem of underdetermined network parameters, we employ a Bayesian learning approach, in which we can integrate arbitrary prior information on the network under consideration. Missing observations are integrated out. We compute the exact likelihood function for smaller networks, and use an approximation to evaluate the likelihood for larger networks. The posterior distribution is evaluated using mode hopping Markov chain Monte Carlo. Distributions over topologies and parameters can then be used to design additional experiments. We evaluate our approach on a small artificial dataset, and present inference results on RNAi data from the Jak/Stat pathway in a human hepatoma cell line.


Subject(s)
Models, Statistical , RNA Interference , Signal Transduction , Algorithms , Cell Line, Tumor , Computer Simulation , Humans , Janus Kinases/metabolism , STAT Transcription Factors/metabolism
16.
J Clin Invest ; 119(2): 376-86, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19139562

ABSTRACT

There is an association between expression of the MHC class I molecule HLA-B27 and protection following human infection with either HIV or HCV. In both cases, protection has been linked to HLA-B27 presentation of a single immunodominant viral peptide epitope to CD8+ T cells. If HIV mutates the HLA-B27-binding anchor of this epitope to escape the protective immune response, the result is a less-fit virus that requires additional compensatory clustered mutations. Here, we sought to determine whether the immunodominant HLA-B27-restricted HCV epitope was similarly constrained by analyzing the replication competence and immunogenicity of different escape mutants. Interestingly, in most HLA-B27-positive patients chronically infected with HCV, the escape mutations spared the HLA-B27-binding anchor. Instead, the escape mutations were clustered at other sites within the epitope and had only a modest impact on replication competence. Further analysis revealed that the cluster of mutations is required for efficient escape because a combination of mutations is needed to impair T cell recognition of the epitope. Artificially introduced mutations at the HLA-B27-binding anchors were found to be either completely cross-reactive or to lead to substantial loss of fitness. These results suggest that protection by HLA-B27 in HCV infection can be explained by the requirement to accumulate a cluster of mutations within the immunodominant epitope to escape T cell recognition.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HLA-B27 Antigen/physiology , Hepatitis C/immunology , Binding Sites , Cells, Cultured , Epitopes, T-Lymphocyte , HLA-B27 Antigen/chemistry , Humans , Immunodominant Epitopes , Mutation , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...