Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
Add more filters

Country/Region as subject
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.
Nat Immunol ; 17(9): 1093-101, 2016 09.
Article in English | MEDLINE | ID: mdl-27478940

ABSTRACT

The manner in which regulatory T cells (Treg cells) control lymphocyte homeostasis is not fully understood. We identified two Treg cell populations with differing degrees of self-reactivity and distinct regulatory functions. We found that GITR(hi)PD-1(hi)CD25(hi) (Triple(hi)) Treg cells were highly self-reactive and controlled lympho-proliferation in peripheral lymph nodes. GITR(lo)PD-1(lo)CD25(lo) (Triple(lo)) Treg cells were less self-reactive and limited the development of colitis by promoting the conversion of CD4(+) Tconv cells into induced Treg cells (iTreg cells). Although Foxp3-deficient (Scurfy) mice lacked Treg cells, they contained Triple(hi)-like and Triple(lo)-like CD4(+) T cells zsuper> T cells infiltrated the skin, whereas Scurfy Triple(lo)CD4(+) T cells induced colitis and wasting disease. These findings indicate that the affinity of the T cell antigen receptor for self antigen drives the differentiation of Treg cells into distinct subsets with non-overlapping regulatory activities.


Subject(s)
Colitis/immunology , Lymph Nodes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Wasting Syndrome/immunology , Animals , Autoantigens/immunology , Autoimmunity , Cell Differentiation , Cell Proliferation , Cells, Cultured , Clonal Selection, Antigen-Mediated , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/transplantation
4.
EMBO J ; 39(18): e103922, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32812257

ABSTRACT

Translational readthrough, i.e., elongation of polypeptide chains beyond the stop codon, was initially reported for viral RNA, but later found also on eukaryotic transcripts, resulting in proteome diversification and protein-level modulation. Here, we report that AGO1x, an evolutionarily conserved translational readthrough isoform of Argonaute 1, is generated in highly proliferative breast cancer cells, where it curbs accumulation of double-stranded RNAs (dsRNAs) and consequent induction of interferon responses and apoptosis. In contrast to other mammalian Argonaute protein family members with primarily cytoplasmic functions, AGO1x exhibits nuclear localization in the vicinity of nucleoli. We identify AGO1x interaction with the polyribonucleotide nucleotidyltransferase 1 (PNPT1) and show that the depletion of this protein further augments dsRNA accumulation. Our study thus uncovers a novel function of an Argonaute protein in buffering the endogenous dsRNA-induced interferon responses, different than the canonical function of AGO proteins in the miRNA effector pathway. As AGO1x expression is tightly linked to breast cancer cell proliferation, our study thus suggests a new direction for limiting tumor growth.


Subject(s)
Argonaute Proteins/metabolism , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Eukaryotic Initiation Factors/metabolism , Interferons/metabolism , Neoplasm Proteins/metabolism , RNA, Double-Stranded/pharmacology , Signal Transduction/drug effects , Argonaute Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Eukaryotic Initiation Factors/genetics , Exoribonucleases/genetics , Exoribonucleases/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Interferons/genetics , Neoplasm Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/genetics
5.
Am J Pathol ; 193(2): 161-181, 2023 02.
Article in English | MEDLINE | ID: mdl-36410420

ABSTRACT

The roof plate-specific spondin-leucine-rich repeat-containing G-protein coupled receptor 4/5 (LGR4/5)-zinc and ring finger 3 (ZNRF3)/ring finger protein 43 (RNF43) module is a master regulator of hepatic Wnt/ß-catenin signaling and metabolic zonation. However, its impact on nonalcoholic fatty liver disease (NAFLD) remains unclear. The current study investigated whether hepatic epithelial cell-specific loss of the Wnt/ß-catenin modulator Lgr4/5 promoted NAFLD. The 3- and 6-month-old mice with hepatic epithelial cell-specific deletion of both receptors Lgr4/5 (Lgr4/5dLKO) were compared with control mice fed with normal diet (ND) or high-fat diet (HFD). Six-month-old HFD-fed Lgr4/5dLKO mice developed hepatic steatosis and fibrosis but the control mice did not. Serum cholesterol-high-density lipoprotein and total cholesterol levels in 3- and 6-month-old HFD-fed Lgr4/5dLKO mice were decreased compared with those in control mice. An ex vivo primary hepatocyte culture assay and a comprehensive bile acid (BA) characterization in liver, plasma, bile, and feces demonstrated that ND-fed Lgr4/5dLKO mice had impaired BA secretion, predisposing them to develop cholestatic characteristics. Lipidome and RNA-sequencing analyses demonstrated severe alterations in several lipid species and pathways controlling lipid metabolism in the livers of Lgr4/5dLKO mice. In conclusion, loss of hepatic Wnt/ß-catenin activity by Lgr4/5 deletion led to loss of BA secretion, cholestatic features, altered lipid homeostasis, and deregulation of lipoprotein pathways. Both BA and intrinsic lipid alterations contributed to the onset of NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Mice , Non-alcoholic Fatty Liver Disease/metabolism , beta Catenin/metabolism , Leucine/metabolism , Liver/metabolism , Cholesterol/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Mice, Inbred C57BL , Diet, High-Fat/adverse effects
6.
Nature ; 555(7698): 678-682, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29562234

ABSTRACT

Histidine phosphorylation, the so-called hidden phosphoproteome, is a poorly characterized post-translational modification of proteins. Here we describe a role of histidine phosphorylation in tumorigenesis. Proteomic analysis of 12 tumours from an mTOR-driven hepatocellular carcinoma mouse model revealed that NME1 and NME2, the only known mammalian histidine kinases, were upregulated. Conversely, expression of the putative histidine phosphatase LHPP was downregulated specifically in the tumours. We demonstrate that LHPP is indeed a protein histidine phosphatase. Consistent with these observations, global histidine phosphorylation was significantly upregulated in the liver tumours. Sustained, hepatic expression of LHPP in the hepatocellular carcinoma mouse model reduced tumour burden and prevented the loss of liver function. Finally, in patients with hepatocellular carcinoma, low expression of LHPP correlated with increased tumour severity and reduced overall survival. Thus, LHPP is a protein histidine phosphatase and tumour suppressor, suggesting that deregulated histidine phosphorylation is oncogenic.


Subject(s)
Histidine/metabolism , Inorganic Pyrophosphatase/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Humans , Inorganic Pyrophosphatase/deficiency , Inorganic Pyrophosphatase/genetics , Male , Mice , Phosphorylation , Proteomics , Survival Analysis , TOR Serine-Threonine Kinases/metabolism , Tumor Burden , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
7.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649241

ABSTRACT

α1-Antitrypsin (AAT) deficiency is a common genetic disease presenting with lung and liver diseases. AAT deficiency results from pathogenic variants in the SERPINA1 gene encoding AAT and the common mutant Z allele of SERPINA1 encodes for Z α1-antitrypsin (ATZ), a protein forming hepatotoxic polymers retained in the endoplasmic reticulum of hepatocytes. PiZ mice express the human ATZ and are a valuable model to investigate the human liver disease of AAT deficiency. In this study, we investigated differential expression of microRNAs (miRNAs) between PiZ and control mice and found that miR-34b/c was up-regulated and its levels correlated with intrahepatic ATZ. Furthermore, in PiZ mouse livers, we found that Forkhead Box O3 (FOXO3) driving microRNA-34b/c (miR-34b/c) expression was activated and miR-34b/c expression was dependent upon c-Jun N-terminal kinase (JNK) phosphorylation on Ser574 Deletion of miR-34b/c in PiZ mice resulted in early development of liver fibrosis and increased signaling of platelet-derived growth factor (PDGF), a target of miR-34b/c. Activation of FOXO3 and increased miR-34c were confirmed in livers of humans with AAT deficiency. In addition, JNK-activated FOXO3 and miR-34b/c up-regulation were detected in several mouse models of liver fibrosis. This study reveals a pathway involved in liver fibrosis and potentially implicated in both genetic and acquired causes of hepatic fibrosis.


Subject(s)
Forkhead Box Protein O3/metabolism , Liver Cirrhosis , MAP Kinase Kinase 4/metabolism , Up-Regulation , Animals , Disease Models, Animal , Forkhead Box Protein O3/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/prevention & control , MAP Kinase Kinase 4/genetics , Male , Mice , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism
8.
J Hepatol ; 79(3): 666-676, 2023 09.
Article in English | MEDLINE | ID: mdl-37290592

ABSTRACT

BACKGROUND & AIMS: Liver injury after COVID-19 vaccination is very rare and shows clinical and histomorphological similarities with autoimmune hepatitis (AIH). Little is known about the pathophysiology of COVID-19 vaccine-induced liver injury (VILI) and its relationship to AIH. Therefore, we compared VILI with AIH. METHODS: Formalin-fixed and paraffin-embedded liver biopsy samples from patients with VILI (n = 6) and from patients with an initial diagnosis of AIH (n = 9) were included. Both cohorts were compared by histomorphological evaluation, whole-transcriptome and spatial transcriptome sequencing, multiplex immunofluorescence, and immune repertoire sequencing. RESULTS: Histomorphology was similar in both cohorts but showed more pronounced centrilobular necrosis in VILI. Gene expression profiling showed that mitochondrial metabolism and oxidative stress-related pathways were more and interferon response pathways were less enriched in VILI. Multiplex analysis revealed that inflammation in VILI was dominated by CD8+ effector T cells, similar to drug-induced autoimmune-like hepatitis. In contrast, AIH showed a dominance of CD4+ effector T cells and CD79a+ B and plasma cells. T-cell receptor (TCR) and B-cell receptor sequencing showed that T and B cell clones were more dominant in VILI than in AIH. In addition, many T cell clones detected in the liver were also found in the blood. Interestingly, analysis of TCR beta chain and Ig heavy chain variable-joining gene usage further showed that TRBV6-1, TRBV5-1, TRBV7-6, and IgHV1-24 genes are used differently in VILI than in AIH. CONCLUSIONS: Our analyses support that SARS-CoV-2 VILI is related to AIH but also shows distinct differences from AIH in histomorphology, pathway activation, cellular immune infiltrates, and TCR usage. Therefore, VILI may be a separate entity, which is distinct from AIH and more closely related to drug-induced autoimmune-like hepatitis. IMPACT AND IMPLICATIONS: Little is known about the pathophysiology of COVID-19 vaccine-induced liver injury (VILI). Our analysis shows that COVID-19 VILI shares some similarities with autoimmune hepatitis, but also has distinct differences such as increased activation of metabolic pathways, a more prominent CD8+ T cell infiltrate, and an oligoclonal T and B cell response. Our findings suggest that VILI is a distinct disease entity. Therefore, there is a good chance that many patients with COVID-19 VILI will recover completely and will not develop long-term autoimmune hepatitis.


Subject(s)
COVID-19 , Chemical and Drug Induced Liver Injury, Chronic , Hepatitis, Autoimmune , Humans , COVID-19 Vaccines/adverse effects , SARS-CoV-2 , COVID-19/prevention & control , Liver/pathology , Receptors, Antigen, T-Cell , Vaccination
9.
Pathobiology ; 90(3): 166-175, 2023.
Article in English | MEDLINE | ID: mdl-36202073

ABSTRACT

INTRODUCTION: Colorectal carcinoma (CRC) is among the most common carcinomas in women and men. In the advanced stage, patients are treated based on the RAS status. Recent studies indicate that in the future, in addition to KRAS and NRAS, alterations in other genes, such as PIK3CA or TP53, will be considered for therapy. Therefore, it is important to know the mutational landscape of routinely diagnosed CRC. METHOD: We report the molecular profile of 512 Swiss CRC patients analyzed by targeted next-generation sequencing as part of routine diagnostics at our institute. RESULTS: Pathogenic and likely pathogenic variants were found in 462 (90%) CRC patients. Variants were detected in TP53 (54.3%), KRAS (48.2%), PIK3CA (15.6%), BRAF (13.5%), SMAD4 (10.5%), FBXW7 (7.8%), NRAS (3.5%), PTEN (2.7%), ERBB2 (1.6%), AKT1 (1.5%), and CTNNB1 (0.9%). The remaining pathogenic alterations were found in the genes ATM(n= 1), MAP2K1(n= 1), and IDH2(n= 1). DISCUSSION/CONCLUSIONS: Our analysis revealed the prevalence of potential predictive markers in a large cohort of CRC patients obtained during routine diagnostic analysis. Furthermore, our study is the first of this size to uncover the molecular landscape of CRC in Switzerland.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins p21(ras) , Male , Humans , Female , Prevalence , Proto-Oncogene Proteins p21(ras)/genetics , Switzerland/epidemiology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Mutation , Class I Phosphatidylinositol 3-Kinases/genetics , High-Throughput Nucleotide Sequencing
10.
Nucleic Acids Res ; 49(15): 8488-8504, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34313788

ABSTRACT

Systematic perturbation screens provide comprehensive resources for the elucidation of cancer driver genes. The perturbation of many genes in relatively few cell lines in such functional screens necessitates the development of specialized computational tools with sufficient statistical power. Here we developed APSiC (Analysis of Perturbation Screens for identifying novel Cancer genes) to identify genetic drivers and effectors in perturbation screens even with few samples. Applying APSiC to the shRNA screen Project DRIVE, APSiC identified well-known and novel putative mutational and amplified cancer genes across all cancer types and in specific cancer types. Additionally, APSiC discovered tumor-promoting and tumor-suppressive effectors, respectively, for individual cancer types, including genes involved in cell cycle control, Wnt/ß-catenin and hippo signalling pathways. We functionally demonstrated that LRRC4B, a putative novel tumor-suppressive effector, suppresses proliferation by delaying cell cycle and modulates apoptosis in breast cancer. We demonstrate APSiC is a robust statistical framework for discovery of novel cancer genes through analysis of large-scale perturbation screens. The analysis of DRIVE using APSiC is provided as a web portal and represents a valuable resource for the discovery of novel cancer genes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, Neoplasm/genetics , Genomics , Neoplasms/genetics , Apoptosis/genetics , Cell Line, Tumor , Gene Amplification/genetics , Humans , Neoplasms/pathology , RNA, Small Interfering/genetics , Signal Transduction/genetics
11.
BMC Genomics ; 22(1): 592, 2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34348664

ABSTRACT

BACKGROUND: Genetic aberrations in hepatocellular carcinoma (HCC) are well known, but the functional consequences of such aberrations remain poorly understood. RESULTS: Here, we explored the effect of defined genetic changes on the transcriptome, proteome and phosphoproteome in twelve tumors from an mTOR-driven hepatocellular carcinoma mouse model. Using Network-based Integration of multi-omiCS data (NetICS), we detected 74 'mediators' that relay via molecular interactions the effects of genetic and miRNA expression changes. The detected mediators account for the effects of oncogenic mTOR signaling on the transcriptome, proteome and phosphoproteome. We confirmed the dysregulation of the mediators YAP1, GRB2, SIRT1, HDAC4 and LIS1 in human HCC. CONCLUSIONS: This study suggests that targeting pathways such as YAP1 or GRB2 signaling and pathways regulating global histone acetylation could be beneficial in treating HCC with hyperactive mTOR signaling.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Pharmaceutical Preparations , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Transcriptome
12.
J Hepatol ; 75(4): 840-847, 2021 10.
Article in English | MEDLINE | ID: mdl-34004216

ABSTRACT

BACKGROUND & AIMS: Loss of serum HBsAg is a hallmark of spontaneous and therapy induced resolution of HBV infection, since it generally reflects a profound decrease in viral replication. However, integrated HBV DNA can contribute to HBsAg expression independent of viral replication. The relative contributions of these sources of HBsAg are not well understood. Specifically, it is not known whether actively transcribed HBV integration could spread throughout the entire liver. METHODS: The relative distribution of HBsAg and HBV RNA in liver biopsy tissue from HBeAg-negative (HBe-) patients was analyzed by immunohistochemistry and in situ hybridization (ISH), respectively. Frozen biopsy tissue was used for molecular analysis of intrahepatic viral RNA, virus-host chimeric transcripts and viral DNA. RESULTS: Immunohistochemistry and ISH analysis revealed HBsAg and HBV RNA positivity in virtually all hepatocytes in the liver of some HBe- patients despite very low viremia. Reverse transcription quantitative PCR and RNA-sequencing analysis confirmed high expression levels of HBV envelope-encoding RNAs. However, the amount of viral transcriptional template (covalently closed circular (ccc)DNA) was too low to support this ubiquitous HBV RNA expression. In contrast, levels of total cellular HBV DNA were consistent with ubiquitous HBV integration. Finally, RNA-sequencing revealed the presence of many HBV-host chimeric transcripts with the potential for HBsAg expression. CONCLUSIONS: Transcriptionally active HBV integration can extend to the entire liver in some HBe- patients. This can lead to ubiquitous HBsAg expression independent of HBV replication. In such patients, HBsAg is probably not a clinically useful surrogate marker for viral resolution or functional cure. LAY SUMMARY: Loss of serum hepatitis B surface antigen (HBsAg) indicates resolution of HBV infection. However, integrated HBV DNA can contribute to HBsAg production independently of viral replication. We investigated the extent of HBsAg-producing viral integration in the livers of patients with low serum viral loads. Our findings suggest that transcriptionally active HBV integration can extend to the entire liver in some patients, questioning the clinical utility of HBsAg as a surrogate marker for viral replication.


Subject(s)
DNA, Viral/analysis , Hepatitis B Antibodies/analysis , Hepatitis B/blood , Viral Load/statistics & numerical data , Adult , DNA, Viral/blood , Female , Hepatitis B/physiopathology , Hepatitis B/virology , Hepatitis B Antibodies/blood , Hepatitis B virus/genetics , Humans , Male , Middle Aged , Viral Load/methods
13.
Ther Umsch ; 78(10): 565-573, 2021.
Article in German | MEDLINE | ID: mdl-34844436

ABSTRACT

Pathology of hepato-pancreato-biliary tumors: An overview. Abstract. Malignant tumors of the hepato-pancreato-biliary organs are usually conventional adenocarcinomas, except for the liver, where they are, based on their characteristic morphology, called hepatocellular neoplasms. In addition to these pure morphological approaches, molecular and especially morphomolecular investigations have yielded many new findings in recent years, always with the aim of providing patients the best possible therapy. For example, focal nodular hyperplasia (FNH) of the liver is now considered a reactive change that does not require further therapy. On the one hand, molecular stratification of hepatocellular adenomas allows to identify high-risk tumors, and which should be surgically treated. As a further example, tumors of the extra- and intrahepatic bile ducts are differentiated today into small-duct-type and large-duct-type, which have different, therapeutically relevant, molecular profiles. The present review provides an overview of the most important and common tumors of the hepato-pancreato-biliary organs.


Subject(s)
Liver Neoplasms , Humans
14.
Lab Invest ; 100(11): 1475-1484, 2020 11.
Article in English | MEDLINE | ID: mdl-32616816

ABSTRACT

Liquid biopsy, the analysis of circulating tumor DNA (ctDNA), is a promising tool in oncology, especially in personalized medicine. Although its main applications currently focus on selection and adjustment of therapy, ctDNA may also be used to monitor residual disease, establish prognosis, detect relapses, and possibly screen at-risk individuals. CtDNA represents a small and variable proportion of circulating cell-free DNA (ccfDNA) which is itself present at a low concentration in normal individuals and so analyzing ctDNA is technically challenging. Various commercial systems have recently appeared on the market, but it remains difficult for practitioners to compare their performance and to determine whether they yield comparable results. As a first step toward establishing national guidelines for ctDNA analyses, four laboratories in Switzerland joined a comparative exercise to assess ccfDNA extraction and ctDNA analysis by sequencing. Extraction was performed using six distinct methods and yielded ccfDNA of equally high quality, suitable for sequencing. Sequencing of synthetic samples containing predefined amounts of eight mutations was performed on three different systems, with similar results. In all four laboratories, mutations were easily identified down to 1% allele frequency, whereas detection at 0.1% proved challenging. Linearity was excellent in all cases and while molecular yield was superior with one system this did not impact on sensitivity. This study also led to several additional conclusions: First, national guidelines should concentrate on principles of good laboratory practice rather than recommend a particular system. Second, it is essential that laboratories thoroughly validate every aspect of extraction and sequencing, in particular with respect to initial amount of DNA and average sequencing depth. Finally, as software proved critical for mutation detection, laboratories should validate the performance of variant callers and underlying algorithms with respect to various types of mutations.


Subject(s)
Circulating Tumor DNA/isolation & purification , DNA Mutational Analysis , Liquid Biopsy/statistics & numerical data , Humans , Laboratories/statistics & numerical data
15.
Pathobiology ; 87(3): 171-178, 2020.
Article in English | MEDLINE | ID: mdl-32079019

ABSTRACT

INTRODUCTION: Gastrointestinal stromal tumor (GIST) is the most common mesenchymal neoplasm of the gastrointestinal tract. It has distinct molecular features and primarily affects the KIT and PDGFRA genes. OBJECTIVE: We wanted to assess the molecular profile of 68 GIST patients who were sequenced consecutively between 2014 and 2019 at our institute of pathology. METHODS: Our cohort comprised 60 primary and 8 metastatic GIST patients; 43 and 57% of the cases, respectively, were analyzed by Sanger sequencing or next-generation sequencing (NGS). RESULTS: Of the 60 primary GIST patients, 47 (78%) showed a KIT mutation; 2 cases showed a double KIT mutation, and 1 of these was a therapy-naive GIST. Nine (15%) patients harbored a PDGFRA mutation, 2 (3%) had a BRAF mutation, 1 (2%) had a PIK3CA mutation, and 1 (2%) did not show any mutation. One BRAF and the PIK3CA mutation have not been described in GIST before. All metastatic GIST harbored exclusively KIT mutations. CONCLUSION: A retrospective analysis of GIST sequenced at our institute revealed incidences of KIT and PDGFRA mutations comparable to those in other cohorts from Europe. Interestingly, we found 2 previously undescribed mutations in the BRAF and PIK3CA genes as well as 1 treatment-naive case with a double KIT mutation in exon 11.


Subject(s)
DNA Mutational Analysis , Gastrointestinal Neoplasms/genetics , Gastrointestinal Stromal Tumors/genetics , Mutation , Aged , Class I Phosphatidylinositol 3-Kinases/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Paraffin Embedding , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Retrospective Studies , Switzerland
16.
Int J Colorectal Dis ; 35(2): 259-268, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31838579

ABSTRACT

PURPOSE: Evaluation of tumor budding in colorectal cancer (CRC) may help to predict the tumors' metastatic potential and patients with an aggressive tumor, although not yet metastasized at time of surgery might benefit from adjuvant therapy. METHODS: The degree of intratumoral tumor budding (ITB) was classified as low, intermediate, and high grade according to the recommendations of the International Tumor Budding Consensus Conference (ITBCC) 2016 on H&E and pankeratin-stained TMA sections from 1262 CRC, no special type (NST), including 655 stage II CRC and was correlated to clinicopathological data and overall survival. RESULTS: Results show that higher ITB rates are significantly linked to higher tumor grade and stage, positive nodal status, lymphovascular invasion (P < 0.0001 each), absence of peritumoral lymphocytes, infiltrating type invasive tumor margin, left-sided cancer localization, and mismatch-repair proficient cancers (P < 0.05 each). In a cohort of 655 stage II CRC, ITB was associated with lymphovascular invasion (P = 0.0459) and adverse clinical outcome (P < 0.0001). In a multivariate analysis including tumor stage, tumor grade, lymphovascular invasion, ITB, and tumor localization, only low tumor stage (P = 0.0022) and absence of lymphovascular invasion (P = 0.0043) showed independent prognostic significance. CONCLUSION: In conclusion, our findings argue towards a clinical utility of ITB as a prognostic biomarker in stage II colorectal cancer to define patients who might benefit from adjuvant therapy. ITB might be used as additional or surrogate marker in CRC in which peritumoral tumor budding is difficult to assess.


Subject(s)
Blood Vessels/pathology , Cell Movement , Colorectal Neoplasms/pathology , Lymphatic Vessels/pathology , Aged , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Female , Humans , Lymphatic Metastasis , Male , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Predictive Value of Tests , Retrospective Studies , Risk Factors , Treatment Outcome
17.
Liver Int ; 39(11): 2124-2135, 2019 11.
Article in English | MEDLINE | ID: mdl-31033124

ABSTRACT

BACKGROUND AND AIMS: Primary biliary cholangitis is an autoimmune biliary disease characterized by injury of bile ducts, eventually leading to cirrhosis and death. In most cases, anti-mitochondrial antibodies and persistently elevated serum alkaline phosphatase are the basis for the serological diagnosis. Anti-nuclear antibodies are also useful and may indicate a more aggressive diseases course. In patients in which anti-mitochondrial antibodies are not detected, an accurate diagnosis requires liver histology. This study aims at identifying specific biomarkers for the serological diagnosis of primary biliary cholangitis. METHODS: Sera from patients affected by primary biliary cholangitis, primary sclerosing cholangitis, hepatitis C virus (with and without cryoglobulinemia), hepatocarcinoma and healthy donors were tested on a protein array representing 1658 human proteins. The most reactive autoantigens were confirmed by DELFIA analysis on expanded cohorts of the same mentioned serum classes, and on autoimmune hepatitis sera, using anti-PDC-E2 as reference biomarker. RESULTS: Two autoantigens, SPATA31A3 and GARP, showed high reactivity with primary biliary cholangitis sera, containing or not anti-mitochondrial antibodies. Their combination with PDC-E2 allowed to discriminate primary biliary cholangitis from all tested control classes with high sensitivity and specificity. We found that GARP expression is upregulated upon exposure to biliary salts in human cholangiocytes, an event involving EGFR and insulin pathways. GARP expression was also detected in biliary duct cells of PBC patients. CONCLUSIONS: This study highlighted SPATA31A3 and GARP as new biomarkers for primary biliary cholangitis and unravelled molecular stimuli underlying GARP expression in human cholangiocytes.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Liver Cirrhosis, Biliary/diagnosis , Membrane Proteins/immunology , Mitochondria/immunology , Adult , Aged , Aged, 80 and over , Biomarkers , Female , Humans , Liver Cirrhosis, Biliary/blood , Liver Cirrhosis, Biliary/immunology , Male , Middle Aged , Young Adult
18.
Gut ; 67(11): 1984-1994, 2018 11.
Article in English | MEDLINE | ID: mdl-29437871

ABSTRACT

OBJECTIVE: Tumour-infiltrating lymphocytes (TILs) favour survival in human colorectal cancer (CRC). Chemotactic factors underlying their recruitment remain undefined. We investigated chemokines attracting T cells into human CRCs, their cellular sources and microenvironmental triggers. DESIGN: Expression of genes encoding immune cell markers, chemokines and bacterial 16S ribosomal RNA (16SrRNA) was assessed by quantitative reverse transcription-PCR in fresh CRC samples and corresponding tumour-free tissues. Chemokine receptor expression on TILs was evaluated by flow cytometry on cell suspensions from digested tissues. Chemokine production by CRC cells was evaluated in vitro and in vivo, on generation of intraperitoneal or intracecal tumour xenografts in immune-deficient mice. T cell trafficking was assessed on adoptive transfer of human TILs into tumour-bearing mice. Gut flora composition was analysed by 16SrRNA sequencing. RESULTS: CRC infiltration by distinct T cell subsets was associated with defined chemokine gene signatures, including CCL5, CXCL9 and CXCL10 for cytotoxic T lymphocytes and T-helper (Th)1 cells; CCL17, CCL22 and CXCL12 for Th1 and regulatory T cells; CXCL13 for follicular Th cells; and CCL20 and CCL17 for interleukin (IL)-17-producing Th cells. These chemokines were expressed by tumour cells on exposure to gut bacteria in vitro and in vivo. Their expression was significantly higher in intracecal than in intraperitoneal xenografts and was dramatically reduced by antibiotic treatment of tumour-bearing mice. In clinical samples, abundance of defined bacteria correlated with high chemokine expression, enhanced T cell infiltration and improved survival. CONCLUSIONS: Gut microbiota stimulate chemokine production by CRC cells, thus favouring recruitment of beneficial T cells into tumour tissues.


Subject(s)
Chemokines/metabolism , Colorectal Neoplasms/immunology , Gastrointestinal Microbiome/immunology , Lymphocytes, Tumor-Infiltrating/microbiology , Adult , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Situ Hybridization , Male , Mice , Middle Aged , RNA, Ribosomal, 16S/metabolism , Real-Time Polymerase Chain Reaction
19.
Lab Invest ; 98(1): 95-105, 2018 01.
Article in English | MEDLINE | ID: mdl-29035381

ABSTRACT

Hepatocellular carcinoma (HCC) represents the fifth and ninth cause of mortality among male and female cancer patients, respectively and typically arises on a background of a cirrhotic liver. HCC develops in a multi-step process, often encompassing chronic liver injury, steatosis and cirrhosis eventually leading to the malignant transformation of hepatocytes. Aberrant expression of the class I homeobox gene family (HOX), a group of genes crucial in embryogenesis, has been reported in a variety of malignancies including solid tumors. Among HOX genes, HOXA13 is most overexpressed in HCC and is known to be directly regulated by the long non-coding RNA HOTTIP. In this study, taking advantage of a tissue microarray containing 305 tissue specimens, we found that HOXA13 protein expression increased monotonically from normal liver to cirrhotic liver to HCC and that HOXA13-positive HCCs were preferentially poorly differentiated and had fewer E-cadherin-positive cells. In two independent cohorts, patients with HOXA13-positive HCC had worse overall survival than those with HOXA13-negative HCC. Using HOXA13 immunohistochemistry and HOTTIP RNA in situ hybridization on consecutive sections of 16 resected HCCs, we demonstrated that HOXA13 and HOTTIP were expressed in the same neoplastic hepatocyte populations. Stable overexpression of HOXA13 in liver cancer cell lines resulted in increased colony formation on soft agar and migration potential as well as reduced sensitivity to sorafenib in vitro. Our results provide compelling evidence of a role for HOXA13 in HCC development and highlight for the first time its ability to modulate response to sorafenib.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Liver/drug effects , Sorafenib/pharmacology , Binding Sites , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Cohort Studies , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Liver/metabolism , Liver/pathology , Liver/surgery , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Survival Analysis , Tissue Array Analysis
20.
Hepatology ; 66(1): 124-135, 2017 07.
Article in English | MEDLINE | ID: mdl-28295475

ABSTRACT

α1 -Antitrypsin (AAT) deficiency is one of the most common genetic disorders and the liver disease due to the Z mutant of AAT (ATZ) is a prototype of conformational disorder due to protein misfolding with consequent aberrant intermolecular protein aggregation. In the present study, we found that livers of PiZ transgenic mice expressing human ATZ have altered expression of a network of hepatocyte transcriptional factors, including hepatocyte nuclear factor-4α, that is early down-regulated and induces a transcriptional repression of ATZ expression. Reduced hepatocyte nuclear factor-4α was associated with activation of ß-catenin, which regulates liver zonation. Livers of PiZ mice and human patients with AAT deficiency were both found to have a severe perturbation of liver zonation. Functionally, PiZ mice showed a severe defect of ureagenesis, as shown by increased baseline ammonia, and reduced urea production and survival after an ammonia challenge. Down-regulation of hepatocyte nuclear factor-4α expression and defective zonation in livers have not been recognized so far as features of the liver disease caused by ATZ and are likely involved in metabolic disturbances and in the increased risk of hepatocellular carcinoma in patients with AAT deficiency. CONCLUSION: The findings of this study are consistent with the concept that abnormal AAT protein conformation and intrahepatic accumulation have broad effects on metabolic liver functions. (Hepatology 2017;66:124-135).


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 4/genetics , Liver Neoplasms/pathology , alpha 1-Antitrypsin Deficiency/genetics , Aging/genetics , Analysis of Variance , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Down-Regulation , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Random Allocation , Statistics, Nonparametric , alpha 1-Antitrypsin Deficiency/pathology
SELECTION OF CITATIONS
SEARCH DETAIL