Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 692263, 2021.
Article in English | MEDLINE | ID: mdl-34497603

ABSTRACT

IFNL3/IFNL4 polymorphisms are inversely associated with the risk of chronic hepatitis C virus (HCV) infection and cirrhosis, two major risk factors for developing hepatocellular carcinoma (HCC). To further explore these inverse associations and their molecular underpinnings, we analyzed IFNL3/IFNL4 polymorphisms represented by the IFNL4 genotype (presence of rs368234815-dG or rs12979860-T alleles) in HCV patients: 2969 from Japan and 2931 from Taiwan. IFNL4 genotype was associated with an increased risk of HCV-related HCC (OR=1.28, 95%CI=1.07-1.52, P=0.0058) in the general population of Japanese patients, but not in Taiwanese patients who achieved treatment-induced viral clearance. IFNL4 genotype was also associated with a decreased risk of cirrhosis (OR=0.66, 95%CI=0.46-0.93, P=0.018, in Taiwanese patients). We then engineered HepG2 cells to inducibly express IFN-λ4 in the presence or absence of interferon lambda receptor 1 (IFNLR1). Induction of IFN-λ4 resulted in its intracellular accumulation, mainly in lysosomes and late endosomes, and increased ER stress, leading to apoptosis and reduced proliferation. We identified the very-low-density lipoprotein receptor (VLDLR), which facilitates HCV entry into hepatocytes, as a transcript induced by IFN-λ4 but not IFN-λ3. Our results suggest that the molecular mechanisms underlying the anti-cirrhotic but pro-HCV associations observed for IFNL3/IFNL4 polymorphisms are, at least in part, contributed by intracellular accumulation of IFN-λ4 causing ER stress in hepatic cells.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Endoplasmic Reticulum Stress , Hepatitis C/metabolism , Interleukins/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Adult , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Case-Control Studies , Cell Proliferation , Databases, Factual , Female , Genetic Predisposition to Disease , Hep G2 Cells , Hepatitis C/genetics , Hepatitis C/virology , Humans , Interferons/genetics , Interleukins/genetics , Japan , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/prevention & control , Liver Cirrhosis/virology , Liver Neoplasms/genetics , Liver Neoplasms/virology , Male , Middle Aged , Phenotype , Polymorphism, Genetic , Protective Factors , Risk Assessment , Risk Factors , Taiwan
2.
medRxiv ; 2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34282422

ABSTRACT

Genomic regions have been associated with COVID-19 susceptibility and outcomes, including the chr12q24.13 locus encoding antiviral proteins OAS1-3. Here, we report genetic, functional, and clinical insights into genetic associations within this locus. In Europeans, the risk of hospitalized vs. non-hospitalized COVID-19 was associated with a single 19Kb-haplotype comprised of 76 OAS1 variants included in a 95% credible set within a large genomic fragment introgressed from Neandertals. The risk haplotype was also associated with impaired spontaneous but not treatment-induced SARS-CoV-2 clearance in a clinical trial with pegIFN-λ1. We demonstrate that two exonic variants, rs10774671 and rs1131454, affect splicing and nonsense-mediated decay of OAS1 . We suggest that genetically-regulated loss of OAS1 expression contributes to impaired spontaneous clearance of SARS-CoV-2 and elevated risk of hospitalization for COVID-19. Our results provide the rationale for further clinical studies using interferons to compensate for impaired spontaneous SARS-CoV-2 clearance, particularly in carriers of the OAS1 risk haplotypes.

3.
Commun Biol ; 4(1): 386, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33753867

ABSTRACT

APOBEC3A (A3A) and APOBEC3B (A3B) enzymes drive APOBEC-mediated mutagenesis. Identification of factors affecting the activity of these enzymes could help modulate mutagenesis and associated clinical outcomes. Here, we show that canonical and alternatively spliced A3A and A3B isoforms produce corresponding mutagenic and non-mutagenic enzymes. Increased expression of the mutagenic A3B isoform predicted shorter progression-free survival in bladder cancer. We demonstrate that the production of mutagenic vs. non-mutagenic A3B protein isoforms was considerably affected by inclusion/skipping of exon 5 in A3B. Furthermore, exon 5 skipping, resulting in lower levels of mutagenic A3B enzyme, could be increased in vitro. Specifically, we showed the effects of treatment with an SF3B1 inhibitor affecting spliceosome interaction with a branch point site in intron 4, or with splice-switching oligonucleotides targeting exon 5 of A3B. Our results underscore the clinical role of A3B and implicate alternative splicing of A3B as a mechanism that could be targeted to restrict APOBEC-mediated mutagenesis.


Subject(s)
Alternative Splicing , Biomarkers, Tumor/genetics , Cytidine Deaminase/genetics , Minor Histocompatibility Antigens/genetics , Mutagenesis , Proteins/genetics , Urinary Bladder Neoplasms/genetics , Biomarkers, Tumor/metabolism , Cytidine Deaminase/metabolism , Epoxy Compounds/pharmacology , Exons , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Isoenzymes , Macrolides/pharmacology , Minor Histocompatibility Antigens/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Progression-Free Survival , Proteins/metabolism , RNA Splicing Factors/antagonists & inhibitors , RNA Splicing Factors/metabolism , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/therapy
4.
Nat Genet ; 52(12): 1283-1293, 2020 12.
Article in English | MEDLINE | ID: mdl-33077916

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19, utilizes angiotensin-converting enzyme 2 (ACE2) for entry into target cells. ACE2 has been proposed as an interferon-stimulated gene (ISG). Thus, interferon-induced variability in ACE2 expression levels could be important for susceptibility to COVID-19 or its outcomes. Here, we report the discovery of a novel, transcriptionally independent truncated isoform of ACE2, which we designate as deltaACE2 (dACE2). We demonstrate that dACE2, but not ACE2, is an ISG. In The Cancer Genome Atlas, the expression of dACE2 was enriched in squamous tumors of the respiratory, gastrointestinal and urogenital tracts. In vitro, dACE2, which lacks 356 amino-terminal amino acids, was non-functional in binding the SARS-CoV-2 spike protein and as a carboxypeptidase. Our results suggest that the ISG-type induction of dACE2 in IFN-high conditions created by treatments, an inflammatory tumor microenvironment or viral co-infections is unlikely to increase the cellular entry of SARS-CoV-2 and promote infection.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Interferons/metabolism , RNA Viruses/physiology , Receptors, Coronavirus/metabolism , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Cell Line , Enzyme Induction , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Receptors, Coronavirus/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Spike Glycoprotein, Coronavirus/metabolism
5.
bioRxiv ; 2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32743577

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which causes COVID-19, utilizes angiotensin-converting enzyme 2 (ACE2) for entry into target cells. ACE2 has been proposed as an interferon-stimulated gene (ISG). Thus, interferon-induced variability in ACE2 expression levels could be important for susceptibility to COVID-19 or its outcomes. Here, we report the discovery of a novel, primate-specific isoform of ACE2, which we designate as deltaACE2 (dACE2). We demonstrate that dACE2, but not ACE2, is an ISG. In vitro, dACE2, which lacks 356 N-terminal amino acids, was non-functional in binding the SARS-CoV-2 spike protein and as a carboxypeptidase. Our results reconcile current knowledge on ACE2 expression and suggest that the ISG-type induction of dACE2 in IFN-high conditions created by treatments, inflammatory tumor microenvironment, or viral co-infections is unlikely to affect the cellular entry of SARS-CoV-2 and promote infection.

6.
Mol Carcinog ; 57(9): 1130-1143, 2018 09.
Article in English | MEDLINE | ID: mdl-29683208

ABSTRACT

Pancreatic Cancer (PC) is a deadly disease in need of new therapeutic options. We recently developed a novel tricarbonylmethane agent (CMC2.24) as a therapeutic agent for PC, and evaluated its efficacy in preclinical models of PC. CMC2.24 inhibited the growth of various human PC cell lines in a concentration and time-dependent manner. Normal human pancreatic epithelial cells were resistant to CMC2.24, indicating selectivity. CMC2.24 reduced the growth of subcutaneous and orthotopic PC xenografts in mice by up to 65% (P < 0.02), and the growth of a human patient-derived tumor xenograft by 47.5% (P < 0.03 vs vehicle control). Mechanistically, CMC2.24 inhibited the Ras-RAF-MEK-ERK pathway. Based on Ras Pull-Down Assays, CMC2.24 inhibited Ras-GTP, the active form of Ras, in MIA PaCa-2 cells and in pancreatic acinar explants isolated from Kras mutant mice, by 90.3% and 89.1%, respectively (P < 0.01, for both). The inhibition of active Ras led to an inhibition of c-RAF, MEK, and ERK phosphorylation by 93%, 91%, and 87%, respectively (P < 0.02, for all) in PC xenografts. Furthermore, c-RAF overexpression partially rescued MIA PaCa-2 cells from the cell growth inhibition by CMC2.24. In addition, downstream of ERK, CMC2.24 inhibited STAT3 phosphorylation levels at the serine 727 residue, enhanced the levels of superoxide anion in mitochondria, and induced intrinsic apoptosis as shown by the release of cytochrome c from the mitochondria to the cytosol and the further cleavage of caspase 9 in PC cells. In conclusion, CMC2.24, a potential Ras inhibitor, is an efficacious agent for PC treatment in preclinical models, deserving further evaluation.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Curcumin/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Signal Transduction/drug effects , ras Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Curcumin/pharmacology , Curcumin/therapeutic use , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
7.
J Immunol ; 199(11): 3808-3820, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29070670

ABSTRACT

Type III IFNs are important mediators of antiviral immunity. IFN-λ4 is a unique type III IFN because it is produced only in individuals who carry a dG allele of a genetic variant rs368234815-dG/TT. Counterintuitively, those individuals who can produce IFN-λ4, an antiviral cytokine, are also less likely to clear hepatitis C virus infection. In this study, we searched for unique functional properties of IFN-λ4 that might explain its negative effect on hepatitis C virus clearance. We used fresh primary human hepatocytes (PHHs) treated with recombinant type III IFNs or infected with Sendai virus to model acute viral infection and subsequently validated our findings in HepG2 cell line models. Endogenous IFN-λ4 protein was detectable only in Sendai virus-infected PHHs from individuals with the dG allele, where it was poorly secreted but highly functional, even at concentrations < 50 pg/ml. IFN-λ4 acted faster than other type III IFNs in inducing antiviral genes, as well as negative regulators of the IFN response, such as USP18 and SOCS1 Transient treatment of PHHs with IFN-λ4, but not IFN-λ3, caused a strong and sustained induction of SOCS1 and refractoriness to further stimulation with IFN-λ3. Our results suggest unique functional properties of IFN-λ4 that can be important in viral clearance and other clinical conditions.


Subject(s)
Alleles , Hepatocytes/immunology , Interferons/genetics , Interleukins/genetics , Respirovirus Infections/immunology , Sendai virus/immunology , Adolescent , Adult , Aged , Endopeptidases/genetics , Female , Hep G2 Cells , Hepacivirus/immunology , Hepatitis C/genetics , Hepatitis C/immunology , Hepatocytes/virology , Humans , Immunity , Interferons/metabolism , Interleukins/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide , Suppressor of Cytokine Signaling 1 Protein/genetics , Ubiquitin Thiolesterase , Up-Regulation , Viral Load , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...