Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Virol ; 98(2): e0180123, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38193691

ABSTRACT

In mammals, NLRX1 is a unique member of the nucleotide-binding domain and leucine-rich repeat (NLR) family showing an ability to negatively regulate IFN antiviral immunity. Intron-containing genes, including NLRX1, have more than one transcript due to alternative splicing; however, little is known about the function of its splicing variants. Here, we identified a transcript variant of NLRX1 in zebrafish (Danio rerio), termed NLRX1-tv4, as a negative regulator of fish IFN response. Zebrafish NLRX1-tv4 was slightly induced by viral infection, with an expression pattern similar to the full-length NLRX1. Despite the lack of an N-terminal domain that exists in the full-length NLRX1, overexpression of NLRX1-tv4 still impaired fish IFN antiviral response and promoted viral replication in fish cells, similar to the full-length NLRX1. Mechanistically, NLRX1-tv4 targeted STING for proteasome-dependent protein degradation by recruiting an E3 ubiquitin ligase RNF5 to drive the K48-linked ubiquitination, eventually downregulating the IFN antiviral response. Mapping of NLRX1-tv4 domains showed that its N-terminal and C-terminal regions exhibited a similar potential to inhibit STING-mediated IFN antiviral response. Our findings reveal that like the full-length NLRX1, zebrafish NLRX-tv4 functions as an inhibitor to shape fish IFN antiviral response.IMPORTANCEIn this study, we demonstrate that a transcript variant of zebrafish NLRX1, termed NLRX1-tv4, downregulates fish IFN response and promotes virus replication by targeting STING for protein degradation and impairing the interaction of STING and TBK1 and that its N- and C-terminus exhibit a similar inhibitory potential. Our results are helpful in clarifying the current contradictory understanding of structure and function of vertebrate NLRX1s.


Subject(s)
Membrane Proteins , Mitochondrial Proteins , Zebrafish Proteins , Animals , Immunity, Innate , Protein Domains , Protein Isoforms/genetics , Ubiquitin-Protein Ligases , Ubiquitination , Zebrafish/immunology , Zebrafish/metabolism , Mitochondrial Proteins/metabolism , Zebrafish Proteins/metabolism , Membrane Proteins/metabolism , Interferons/metabolism
2.
J Immunol ; 209(7): 1335-1347, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36165180

ABSTRACT

Tripartite motif (TRIM) family proteins have come forth as important modulators of innate signaling dependent on of E3 ligase activity. Recently, several human TRIM proteins have been identified as unorthodox RNA-binding proteins by RNA interactome analyses; however, their targets and functions remain largely unknown. FTRCA1 is a crucian carp (Carassius auratus)-specific finTRIM (fish novel TRIM) member and negatively regulates the IFN antiviral response by targeting two retinoic acid-inducible gene-I (RIG-I)-like receptor (RLR) pathway molecules, that is, TANK-binding kinase 1 (TBK1) and IFN regulatory factor 7 (IRF7). In this study, we identify FTRCA1 as an RNA-binding E3 ligase and characterize the contribution of its RNA-binding activity and E3 ligase activity to fish IFN response. Besides targeting TBK1 and IRF7, FTRCA1 downregulates fish IFN response also by targeting stimulator of IFN response cGAMP interactor 1 (STING1). E3 ligase activity is required for full inhibition on the TBK1- and IRF7-mediated IFN response, but partial inhibition on the STING1-mediated IFN response. However, FTRCA1 has a general binding potential to mRNAs in vitro, it selectively binds STING1 and IRF7 mRNAs in vivo to attenuate mRNA levels, and it directly interacts with TBK1 protein to target protein degradation for downregulating the IFN response. Our results present an interesting example of a fish species-specific finTRIM protein that has acquired RNA-binding activity and E3 ligase activity to fine-tune fish IFN response.


Subject(s)
Factor VII , RNA , Animals , Antiviral Agents , Fish Proteins/genetics , Humans , Immunity, Innate , RNA, Messenger , Tretinoin , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
3.
J Immunol ; 208(5): 1189-1203, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35101889

ABSTRACT

The small HERC family currently comprises four members (HERC3-6) involved in the regulation of various physiological activities. Little is known about the role of HERCs in IFN response. In this study, we identify a novel fish HERC member, named crucian carp HERC7, as a negative regulator of fish IFN response. Genome-wide search of homologs and comprehensive phylogenetic analyses reveal that the small HERC family, apart from HERC3-6 that have been well-characterized in mammals, contains a novel HERC7 subfamily exclusively in nonmammalian vertebrates. Lineage-specific and even species-specific expansion of HERC7 subfamily in fish indicates that crucian carp HERC7 might be species-specific. In virally infected fish cells, HERC7 is induced by IFN and selectively targets three retinoic acid-inducible gene-I-like receptor signaling factors for degradation to attenuate IFN response by two distinct strategies. Mechanistically, HERC7 delivers mediator of IFN regulatory factor 3 activator and mitochondrial antiviral signaling protein for proteasome-dependent degradation at the protein level and facilitates IFN regulatory factor 7 transcript decay at the mRNA level, thus abrogating cellular IFN induction to promote virus replication. Whereas HERC7 is a putative E3 ligase, the E3 ligase activity is not required for its negative regulatory function. These results demonstrate that the ongoing expansion of the small HERC family generates a novel HERC7 to fine-tune fish IFN antiviral response.


Subject(s)
Interferon Regulatory Factor-7/metabolism , Interferons/immunology , Reoviridae/immunology , Rhabdoviridae/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Carps , Cell Line , Fish Proteins/genetics , HEK293 Cells , Humans , Interferon Regulatory Factor-7/genetics , Membrane Proteins/metabolism , RNA Stability/genetics , RNA, Messenger/genetics , Signal Transduction/immunology , Trans-Activators/genetics
4.
PLoS Pathog ; 17(1): e1009220, 2021 01.
Article in English | MEDLINE | ID: mdl-33476326

ABSTRACT

The eponymous member of the interferon regulatory factor (IRF) family, IRF1, was originally identified as a nuclear factor that binds and activates the promoters of type I interferon genes. However, subsequent studies using genetic knockouts or RNAi-mediated depletion of IRF1 provide a much broader view, linking IRF1 to a wide range of functions in protection against invading pathogens. Conserved throughout vertebrate evolution, IRF1 has been shown in recent years to mediate constitutive as well as inducible host defenses against a variety of viruses. Fine-tuning of these ancient IRF1-mediated host defenses, and countering strategies by pathogens to disarm IRF1, play crucial roles in pathogenesis and determining the outcome of infection.


Subject(s)
Communicable Diseases/immunology , Communicable Diseases/therapy , Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Interferon Regulatory Factor-1/metabolism , Animals , Communicable Diseases/metabolism , Humans , Interferon Regulatory Factor-1/immunology
5.
Int J Mol Sci ; 24(5)2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36902023

ABSTRACT

In humans, four small HERCs (HERC3-6) exhibit differential degrees of antiviral activity toward HIV-1. Recently we revealed a novel member HERC7 of small HERCs exclusively in non-mammalian vertebrates and varied copies of herc7 genes in distinct fish species, raising a question of what is the exact role for a certain fish herc7 gene. Here, a total of four herc7 genes (named HERC7a-d sequentially) are identified in the zebrafish genome. They are transcriptionally induced by a viral infection, and detailed promoter analyses indicate that zebrafish herc7c is a typical interferon (IFN)-stimulated gene. Overexpression of zebrafish HERC7c promotes SVCV (spring viremia of carp virus) replication in fish cells and concomitantly downregulates cellular IFN response. Mechanistically, zebrafish HERC7c targets STING, MAVS, and IRF7 for protein degradation, thus impairing cellular IFN response. Whereas the recently-identified crucian carp HERC7 has an E3 ligase activity for the conjugation of both ubiquitin and ISG15, zebrafish HERC7c only displays the potential to transfer ubiquitin. Considering the necessity for timely regulation of IFN expression during viral infection, these results together suggest that zebrafish HERC7c is a negative regulator of fish IFN antiviral response.


Subject(s)
Fish Diseases , Rhabdoviridae Infections , Animals , Humans , Zebrafish/genetics , Interferons/metabolism , Zebrafish Proteins/metabolism , Antiviral Agents , Ubiquitins
6.
J Immunol ; 205(1): 237-250, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32471880

ABSTRACT

In mammals, transcription factors of IFN-regulatory factors (IRFs) family translate viral recognition into IFN antiviral responses through translocating to nucleus and subsequently binding to the promoters of IFN and IFN-stimulated genes (ISGs). In addition to IRF1-9 conserved across vertebrates and IRF10 in teleost fish and bird, teleost fish has another novel member, IRF11; however, little is known about its role in IFN response. In this study, we provide evidence that IRF11 is present only in Osteichthyes (bony fish) but lost in tetrapods and subsequently characterize the stimulatory potential of zebrafish IRF11 to IFN antiviral response relevant to its subcellular localization and promoter binding. Overexpression of zebrafish IRF11 restricts virus replication through induction of IFN and ISGs. Zebrafish IRF11 is constitutively localized to nucleus, which is driven by a tripartite NLS motif, consisting of three interdependent basic clusters, two in DNA binding domain (DBD) and one in the region immediately C-terminal to DBD. Nuclear IRF11 binds to the IRF-binding element/IFN-stimulated response element motifs of zebrafish IFN promoters depending on the two conserved amino acids (K78, R82) within DBD helix α3. K78 and R82 also benefit zebrafish IRF11 nuclear import as two key residues positioned at the first basic cluster of the tripartite NLS motif. Such features enable zebrafish IRF11 to function as a positive transcription factor for fish IFN antiviral response. Our results identify a unique tripartite NLS motif that integrates DNA-binding activity and nuclear import ability, allowing zebrafish IRF11 to initiate IFN and ISG expression.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferons/genetics , Rhabdoviridae Infections/veterinary , Transcription Factors, TFII/metabolism , Zebrafish Proteins/metabolism , Zebrafish/immunology , Amino Acid Motifs/genetics , Amino Acid Sequence/genetics , Animals , Cell Line , Cell Nucleus/metabolism , Conserved Sequence/genetics , Gene Expression Regulation/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Interferon Regulatory Factor-1/genetics , Interferons/metabolism , Promoter Regions, Genetic/genetics , Protein Domains/genetics , Response Elements , Rhabdoviridae/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology , Signal Transduction/genetics , Transcription Factors, TFII/genetics , Virus Replication/immunology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/virology
7.
J Immunol ; 202(8): 2407-2420, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30850476

ABSTRACT

In mammals, tripartite motif (TRIM) proteins have emerged as pivotal players endowed with, directly, antiviral effects and, indirectly, modulatory capacity of the innate immune response. An unprecedented expansion of TRIM family has occurred in fish; however, the functional role of fish TRIM family members remains largely unknown. In this study, we identify a species-specific TRIM gene from crucian carp Carassius auratus, named FTRCA1, phylogenetically similar to the members of finTRIM, a subfamily of TRIM exclusively in teleost fish. FTRCA1 is induced by IFN and IFN stimuli as a typical IFN-stimulated gene. Overexpression of FTRCA1 negatively regulates IFN antiviral response by inhibition of IRF3 phosphorylation; consistently, knockdown of FTRCA1 results in enhanced levels of IRF3 phosphorylation and also IFN expression following poly(I:C) transfection. Whereas FTRCA1 is associated with several pivotal signaling molecules of RIG-I-like receptor pathway, its association with TBK1 results in autophage-lysosomal degradation of TBK1, thus abrogating the downstream IFN induction. Interestingly, FTRCA1 is phosphorylated by TBK1, but this phosphorylation is not required for downregulation of TBK1 protein. Transfection assays indicate that FTRCA1 is likely an E3 ligase with the requirement of RING finger domain, and deletion of N-terminal RING domain or mutation of seven conservative sites abolishes the negative regulatory function of FTRCA1. Collectively, these results illuminate a novel finTRIM-mediated innate immune modulatory pathway, thus providing insights into species-specific regulation of fish IFN response.


Subject(s)
Autophagosomes/immunology , Fish Proteins/immunology , Goldfish/immunology , Interferons/immunology , Lysosomes/immunology , Protein Serine-Threonine Kinases/immunology , Proteolysis , Tripartite Motif Proteins/immunology , Animals , Fish Proteins/genetics , Goldfish/genetics , HEK293 Cells , Humans , Interferons/genetics , Lysosomes/genetics , Protein Serine-Threonine Kinases/genetics , Tripartite Motif Proteins/genetics
8.
J Immunol ; 200(2): 688-703, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29203516

ABSTRACT

In mammals, RIG-I like receptors (RLRs) RIG-I and melanoma differentiation-associated gene 5 (MDA5) sense cytosolic viral RNA, leading to IFN antiviral response; however, LGP2 exhibits controversial functions. The same happens to fish LGP2. In this study we report that three zebrafish LGP2 splicing transcripts, a full-length LGP2, and two truncating variants, LGP2v1 and LGP2v2, play distinct roles during IFN antiviral response. Overexpression of the full-length LGP2 not only potentiates IFN response through the RLR pathway, in the absence or presence of poly(I:C) at limited concentrations, but also inhibits IFN response by relative high concentrations of poly(I:C) through functional attenuation of signaling factors involved in the RLR pathway; however, LGP2v1 and LGP2v2 only retain the inhibitory role. Consistently, LGP2 but not LGP2v1 and LGP2v2 confers protection on fish cells against spring viremia of carp virus (SVCV) infection and at limited expression levels, LGP2 exerts more significant protection than either RIG-I or MDA5. Further data suggest that in the early phase of SVCV infection, LGP2 functions as a positive regulator but along with SVCV replicating in cells up to a certain titer, which leads to a far more robust expression of IFN, LGP2 switches to a negative role. These in vitro results suggest an ingenious mechanism where the three zebrafish LGP2 splicing transcripts work cooperatively to shape IFN antiviral responses.


Subject(s)
Alternative Splicing , Antiviral Agents/metabolism , Disease Resistance/genetics , Fish Proteins/genetics , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Interferons/biosynthesis , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Line , Disease Resistance/immunology , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/virology , Host-Pathogen Interactions/immunology , Poly I-C/immunology , Polylysine/immunology , RNA Helicases/genetics , RNA Helicases/metabolism , Signal Transduction , Virus Replication/immunology , Zebrafish/virology
9.
Fish Shellfish Immunol ; 90: 180-187, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31048035

ABSTRACT

In mammals and fish, emerging evidence highlights that TRIM family members play important roles in the interferon (IFN) antiviral immune response. Fish TRIM family has undergone an unprecedented expansion leading to generation of finTRIM subfamily, which is exclusively specific to fish. Our recent results have shown that FTRCA1 (finTRIM C. auratus 1) is likely a fish species-specific finTRIM member in crucian carp C. auratus and acts as a negative modulator to downregulate fish IFN response by autophage-lysosomal degradation of protein kinase TBK1. In the present study, we found that FTRCA1 also impedes the activation of crucian carp IFN promoter by IRF7 but not by IRF3. Mechanistically, FTRCA1 attenuates IRF7 transcription levels likely due to enhanced decay of IRF7 mRNA, leading to reduced IRF7 protein levels and subsequently reduced fish IFN expression. E3 ligase activity is required for FTRCA1 to negatively regulate IRF7-mediated IFN response, because ligase-inactive mutants and the RING-deleted mutant of FTRCA1 lose the ability to block the activation of crucian carp IFN promoter by IRF7. These results together indicate that FTRCA1 is a multifaceted modulator to target different signaling factors for shaping fish IFN response in crucian carp.


Subject(s)
Carps/genetics , Carps/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Interferon Regulatory Factor-7/metabolism , Interferons/metabolism , Transcription, Genetic , Animals , Carps/metabolism , Fish Proteins/metabolism
10.
Fish Shellfish Immunol ; 86: 1058-1063, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30593899

ABSTRACT

In mammals, virus infection of host cells triggers innate immune response, characterized by induction of interferon (IFN) and downstream IFN-stimulated genes (ISGs). The initiation of IFN antiviral response is dependent on host recognition of virus infection. In fish, similar IFN antiviral response is induced in response to RNA or DNA virus infection; however, the detailed mechanisms underlying recognition of a given virus and activation of downstream signaling remain largely unexplored. Using an infection model with Epithelioma papulosum cyprini (EPC) cells and spring viremia of carp virus (SVCV), a negative sense single-stranded RNA virus, we reported that fish RLR signaling pathway was involved in SVCV-triggered fish IFN response. IFN response was significantly initiated in EPC cells when infected with SVCV, as evidenced by activation of fish IFN promoters, upregulation of IFN and ISGs at mRNA and protein levels. However, function blockade of RIG-I and MDA5, two cytosolic receptors of fish RLR family, significantly attenuated the activation of fish IFN promoters and also the induction of fish IFN and ISGs by SVCV infection. Consistently, SVCV infection-triggered IFN response were blocked in EPC cells when transfected with the dominant negative mutants of pivotal RLR signaling factors, including MAVS, MITA, TBK1, IRF3 and IRF7. These results together shed light on the conservation of RLR-mediated IFN signaling that contributes to fish cells responding to RNA virus infection.


Subject(s)
Fish Diseases/immunology , Rhabdoviridae Infections/veterinary , Rhabdoviridae/physiology , Signal Transduction , Animals , Cell Line, Tumor , Cyprinidae/immunology , DEAD Box Protein 58/metabolism , Fish Diseases/virology , Immunity, Innate , Interferons/immunology , Promoter Regions, Genetic , Rhabdoviridae Infections/immunology
11.
J Immunol ; 197(5): 1893-904, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27496972

ABSTRACT

In mammals, IFN regulatory factor (IRF)1, IRF3, and IRF7 are three critical transcription factors that are pivotal for cooperative regulation of the type I IFN response. In this study, we explored the relative contribution of zebrafish (Danio rerio) IRF1 (DrIRF1), IRF3 (DrIRF3), and IRF7 (DrIRF7) (DrIRF1/3/7) to zebrafish IFNΦ1 (DrIFNΦ1) and IFNΦ3 (DrIFNΦ3) (DrIFNΦ1/3) activation. Following spring viremia of carp virus infection, DrIFNΦ1/3 and DrIRF1/3/7 transcripts are significantly induced in zebrafish tissues, which correlates with the replication of spring viremia of carp virus. DrIRF1/3/7 selectively bind to the IRF-binding element/IFN-stimulated regulatory element sites of DrIFNΦ1/3 promoters, with the exception that DrIRF3 has no preference for two IRF-binding element/IFN-stimulated regulatory element motifs within the DrIFNΦ3 promoter. Consistently, DrIRF3 alone activates DrIFNΦ1, but not DrIFNΦ3; DrIRF7 predominantly stimulates DrIFNΦ3; and DrIRF1 has similar potential to DrIFNΦ1 and DrIFNΦ3. Strikingly, DrIRF3 facilitates the binding of DrIRF1 and DrIRF7 to both zebrafish IFN promoters, and so does DrIRF7 for the binding of DrIRF1, particularly to the DrIFNΦ3 promoter. These binding properties correlate with differential responses of DrIFNΦ1 and DrIFNΦ3 to the combinatory stimulation of DrIRF1/3/7, depending on their relative amounts. Similar to the dual roles of human IRF3 in regulating IRF7-activated IFNα genes, DrIRF3 exerts dual effects on DrIRF1-mediated DrIFNΦ3 gene expression: an inhibitory effect at lower concentrations and a synergistic effect at higher concentrations. These data provide evidence that fish and mammals have evolved a similar IRF-dependent regulatory mechanism fine-tuning IFN gene activation.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , Interferons/genetics , Rhabdoviridae Infections/immunology , Animals , Binding Sites , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/immunology , Interferons/immunology , Promoter Regions, Genetic , Protein Binding , Rhabdoviridae , Rhabdoviridae Infections/metabolism , Signal Transduction , Transcription, Genetic , Transcriptional Activation , Zebrafish/genetics , Zebrafish/immunology
12.
Neuropsychol Rev ; 27(1): 46-61, 2017 03.
Article in English | MEDLINE | ID: mdl-28102493

ABSTRACT

Autism spectrum disorders (ASD) are characterized by executive dysfunction, and working memory (WM) comprises one core component of executive function. Many studies have investigated WM impairments in individuals with ASD, however, a conclusive agreement has not been reached. The present study provided a meta-analytic review of WM impairments in individuals with ASD and evaluated potential moderating variables of this problem. Twenty-eight studies were included in this study, and the participants comprised 819 individuals with ASD and 875 healthy controls. A significant WM impairment (Cohen's d = -0.61) was identified in the individuals with ASD, however, this impairment was not associated with age. Results of moderation analyses showed that (a) spatial WM was more severely impaired than verbal WM and (b) the component of cognitive processing (maintenance vs. maintenance plus manipulation) did not affect the severity of WM impairments. These findings suggest that WM is impaired in individuals with ASD and may have implications for interventions related to WM impairments in these individuals.


Subject(s)
Autism Spectrum Disorder/complications , Memory Disorders/etiology , Memory, Short-Term/physiology , Humans , Memory Disorders/diagnosis , Neuropsychological Tests
13.
J Immunol ; 194(3): 1225-38, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25535281

ABSTRACT

In mammals, type I IFNs (mainly IFN-α/ß) are primarily regulated by transcription factors of the IFN regulatory factor (IRF) family. Fish IFNs do not show a one-to-one orthologous relationship with mammalian type I IFN homologues. Using a bacterial one-hybrid reporter screening system and an overexpression approach to explore the molecular mechanism underlying fish IFN induction, we identified zebrafish Danio rerio IRF (DrIRF)1 as a positive regulator of the fish IFN antiviral response. Among 12 zebrafish IRF family genes, DrIRF1 is most abundant in zebrafish immune tissues, including head kidney and spleen; upon virus infection, it is one of most significantly induced genes. Overexpression of DrIRF1 induces the expression of IFN and IFN-stimulated genes, hence protecting epithelioma papulosum cyprini cells against spring viremia of carp virus infection. As a transcription factor with constitutively nuclear retention, DrIRF1 directly binds to the IFN-stimulated regulatory element/IRF-binding element sites of zebrafish IFN promoters, which are dependent on four conserved amino acids of the N-terminal DNA-binding domain helix α3 motif. Mutation of either residue reveals a differential requirement for DrIRF1-mediated activation of zebrafish IFNϕ1 and IFNϕ3 promoters. Notably, C-terminal phosphorylation of DrIRF1 is observed and is not required for in vitro binding of DrIRF1 to fish IFN promoters. Unlike DrIRF3 and DrIRF7, which are responsible for differential expression of zebrafish IFNϕ1 and IFNϕ3 through the retinoic acid-inducible gene I-like receptor pathway, DrIRF1 works in concert with MyD88 to activate zebrafish IFNϕ3 but not IFNϕ1. These results provide insights into the evolving function of IRF1 as a positive IFN regulator.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factor-1/metabolism , Interferons/genetics , Interferons/metabolism , Myeloid Differentiation Factor 88/metabolism , Promoter Regions, Genetic , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cluster Analysis , Gene Expression Profiling , Gene Order , Interferon Regulatory Factor-1/chemistry , Interferon Regulatory Factor-1/genetics , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Sequence Alignment , Transcription Factors/metabolism , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Zebrafish
14.
J Immunol ; 193(3): 1100-9, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24958903

ABSTRACT

IFN regulatory factor (IRF) 10 belongs to the IRF family and exists exclusively in birds and fish. Most IRFs have been identified as critical regulators in the IFN responses in both fish and mammals; however, the role of IRF10 is unclear. In this study, we identified IRF10 in zebrafish (Danio rerio) and found that it serves as a negative regulator to balance the innate antiviral immune responses. Zebrafish IRF10 (DrIRF10) was induced by intracellular polyinosinic:polycytidylic acid in ZF4 (zebrafish embryo fibroblast-like) cells. DrIRF10 inhibited the activation of zebrafish IFN1 (DrIFN1) and DrIFN3 promoters in epithelioma papulosum cyprinid cells in the presence or absence of polyinosinic:polycytidylic acid stimulation through direct interaction with the IFN promoters, and this inhibition was also shown to block IFN signaling. Overexpression of DrIRF10 was able to abolish the induction of DrIFN1 and DrIFN3 mediated by the retinoic acid-inducible gene I-like receptors. In addition, functional domain analysis of DrIRF10 showed that either the DNA binding domain or the IRF association domain is sufficient for its inhibitory activity for IFN signaling. Lastly, overexpression of DrIRF10 decreased the transcription level of several IFN-stimulated genes, resulting in the susceptibility of host cells to spring viremia of carp virus infection. Collectively, these data suggest that DrIRF10 inhibits the expression of DrIFN1 and DrIFN3 to avoid an excessive immune response, a unique regulation mechanism of the IFN responses in lower vertebrates.


Subject(s)
Down-Regulation/immunology , Fish Proteins/antagonists & inhibitors , Interferon Regulatory Factors/physiology , Interferons/antagonists & inhibitors , Zebrafish/immunology , Animals , Carcinoma/genetics , Carcinoma/immunology , Carcinoma/pathology , Down-Regulation/genetics , Fish Proteins/biosynthesis , Fish Proteins/genetics , HEK293 Cells , Humans , Interferon Regulatory Factors/genetics , Interferons/biosynthesis , Interferons/genetics , Zebrafish/genetics
15.
J BUON ; 21(5): 1195-1202, 2016.
Article in English | MEDLINE | ID: mdl-27837623

ABSTRACT

PURPOSE: The main purpose of this study was to demonstrate the anticancer effects of 4-terpineol against Hep-G2 hepatocellular carcinoma (HCC) cells by evaluating its effect on apoptosis induction, cell migration, DNA fragmentation and cell cycle phase distribution. METHODS: MTT assay was used to evaluate the cytotoxic effect of 4-terpineol on Hep-G2 cells, while fluorescence microscopy and flow cytometry were used to study apoptosis induction. Wound healing assay was used to study the effects of 4-terpineol on cell migration, while gel electrophoresis was performed to evaluate the effects on DNA fragmentation. Flow cytometry using propidium iodide (PI) as a probe was used to evaluate the effects on cell cycle arrest. Cells treated with dimethylsulfoxide (DMSO) only served as controls. BALB/c nude mice weighing about 35 g each were used for in vivo studies using 10 and 20 mg/kg of 4-terpineol dose. RESULTS: 4-terpineol induced dose-dependent cytotoxicity in Hep-G2 hepatocellular carcinoma cells. Gel electrophoresis indicated that DNA fragmentation was associated with increasing dose of 4-terpineol. It was also observed that a wound scratch in the vehicle-treated control cells was practically entirely closed after 48 hrs of incubation. However, treatment with 0, 25, 50 and 100 µM dose of 4-terpineol resulted in inhibition of wound healing in a dose-dependent manner. The percentage of apoptotic cells increased from 2.5% in the control cells to 10.3, 64.6 and 78.9% in cells treated with 25, 50 and 100 µM of 4-terpineol respectively. 4-terpineol-treated cells exhibited increased percentage of cells in sub-G1 phase of the cell cycle. The in vivo mouse results indicated that 10 and 20 mg/kg of 4-terpineol decreased the tumor weight and tumor volume in a dose-dependent manner. CONCLUSION: The results of this study showed that 4-terpineol exhibits anticancer effects in Hep-G2 cells by inducing apoptosis, DNA fragmentation, inhibition of cell migration and sub-G1 cell cycle arrest.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cell Movement/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Liver Neoplasms/drug therapy , Menthol/analogs & derivatives , Animals , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Menthol/pharmacology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(3): 409-12, 2015 May.
Article in Zh | MEDLINE | ID: mdl-26121863

ABSTRACT

OBJECTIVE: To investigate the protein expression of the p16 gene and the methylation of its promoter in breast cancer, and to analyze the correlation between the p16 DNA methylation and the clinicopathological features. METHODS: Immuno-histochemistry technique (SP method) and methylation-specific-PCR (MSP) were used to detect p16 protein expression and the methylation of the p16 promoter in 47 breast cancer samples as well as in 20 hyperplasia samples of mammary glands. Results The p16 protein expression in breast cancer samples significantly lower when compared with those of hyperplasia samples (48. 9% vs. 70. 0%) and p16 methylation was more frequent in breast-tumor tissues when compared with those of hyperplasia samples (38. 3% vs. 20. 0%), but the statistical significance wasn't found (P> 0. 05). Down-regulation of p16 protein was negatively correlation with p16 gene hypermethylation (r= -0. 33, P =0. 02). Meanwhile, p16 methylation in breast cancer tissues correlated with histological type, lymph node metastasis, but not correlated with the age, tumor diameter, TNM stage, expression of estrogen receptor (ER) and progesterone receptor (PR) gene status. CONCLUSION: The downregulation of p16 protein induced by promoter methylation of p16 gene may not contribute to early cancinogenesis, but may contribute to progression of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Genes, p16 , Promoter Regions, Genetic , Breast/pathology , Carcinogenesis , Disease Progression , Down-Regulation , Female , Humans , Hyperplasia , Immunohistochemistry , Lymphatic Metastasis
17.
Fish Shellfish Immunol ; 39(2): 168-77, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24825429

ABSTRACT

Human viperin is known as an interferon (IFN)-inducible antiviral protein and localizes to endoplasmic reticulum (ER) via its N-terminal amphipathic α-helix. Little is known about subcellular localization of fish viperin. Herein, we characterized subcellular localization of a fish viperin from crucian carp Carassius auratus. Crucian carp viperin is nearly identical to the other viperin proteins in sequence, with the exception of the first N-terminal 70 amino acids that are defined as N-terminal variable domain including an amphipathic α-helix. In addition to N-terminal variable domain, crucian carp viperin protein harbors a conserved middle radical SAM domain and a conserved C-terminal domain. Subcellular localization analyses indicate that crucian carp viperin is a cytoplasmic protein associated with ER. Sequence analyses reveal that amino acids 1-74 forms an amphipathic α-helix domain that drives ER-localization of crucian carp viperin. In addition, Coimmunoprecipitation assays show that crucian carp viperin proteins are able to self-associate. These results together indicate that similar to mammalian homologs, fish viperins likely play important roles in IFN response.


Subject(s)
Carps/genetics , Fish Proteins/genetics , Animals , Base Sequence , Conserved Sequence/genetics , Endoplasmic Reticulum/metabolism , Immunoprecipitation , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors , Protein Structure, Tertiary , Proteins/genetics , Sequence Analysis, DNA , Species Specificity
18.
Fish Shellfish Immunol ; 41(2): 222-30, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25219369

ABSTRACT

Mammalian mitochondrial antiviral signaling protein (MAVS) is an essential adapter involved in retinoic acid-inducible gene-I (RIG-I)-like receptor (RLR)-triggered interferon (IFN) antiviral immunity. Fish MAVS homologues have been identified in several fish species; however, the signaling pathway involving fish MAVS-mediated IFN response remains to be investigated. In the present study, we report identification of a fish MAVS orthologue from crucian carp Carassius auratus blastulae embryonic (CAB) cells and its function role in fish RLR signaling. Crucian carp MAVS is constitutively expressed in CAB cells and is not transcriptionally induced by cytosolic poly (I:C) and IFN. Overexpression of crucian carp MAVS results in activation of fish IFN promoter and ISRE-containing promoter as well as transcriptional expression of IFN and ISGs including PKR and Mx1, which is impaired by functional blockade of signaling molecules TBK1 and IRF3/7. Either cytosolic poly (I:C)-induced or RIG-I-induced IFN response is attenuated by functional blockade of crucian carp MAVS. These results together indicate that fish MAVS contributes to IFN antiviral immunity downstream of cytosolic poly (I:C) and RIG-I and upstream of TBK1 and IRF3/7. Moreover, we provide evidence that apart from crucian carp MAVS, crucian carp MITA is also involved in cytosolic poly (I:C)- and RIG-I-induced IFN response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carps/immunology , Mitochondrial Proteins/metabolism , Signal Transduction/immunology , Animals , Blastula/metabolism , Cloning, Molecular , DNA Primers/genetics , Interferons/immunology , Luciferases , Poly I-C/immunology , Real-Time Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/immunology
19.
STAR Protoc ; 5(3): 103156, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-38941183

ABSTRACT

A gene-rescue experiment under a mutant background is essential to clarify gene function and the resulting biological potential in vivo. Here, we present a protocol for determining the change in interferon response by microinjecting plasmids into one-cell-stage zebrafish embryos. We describe steps for comparing the resistance potential to virus infection in wild-type and knockout zebrafish larvae following plasmid microinjection. We then detail how to link the enhanced interferon immunity to the improved resistance in knockout zebrafish larvae by gene-rescue experiments. For complete details on the use and execution of this protocol, please refer to Qu et al.1.


Subject(s)
Gene Knockout Techniques , Interferons , Zebrafish , Animals , Zebrafish/genetics , Gene Knockout Techniques/methods , Interferons/genetics , Interferons/metabolism , Interferons/immunology , Microinjections/methods , Disease Resistance/genetics , Disease Resistance/immunology , Larva/virology , Larva/immunology , Larva/genetics , Plasmids/genetics
20.
World J Gastrointest Oncol ; 16(5): 1947-1964, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38764850

ABSTRACT

BACKGROUND: Gastric cancer (GC) has a high mortality rate worldwide. Despite significant progress in GC diagnosis and treatment, the prognosis for affected patients still remains unfavorable. AIM: To identify important candidate genes related to the development of GC and identify potential pathogenic mechanisms through comprehensive bioinformatics analysis. METHODS: The Gene Expression Omnibus database was used to obtain the GSE183136 dataset, which includes a total of 135 GC samples. The limma package in R software was employed to identify differentially expressed genes (DEGs). Thereafter, enrichment analyses of Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were performed for the gene modules using the clusterProfile package in R software. The protein-protein interaction (PPI) networks of target genes were constructed using STRING and visualized by Cytoscape software. The common hub genes that emerged in the cohort of DEGs that was retrieved from the GEPIA database were then screened using a Venn Diagram. The expression levels of these overlapping genes in stomach adenocarcinoma samples and non-tumor samples and their association with prognosis in GC patients were also obtained from the GEPIA database and Kaplan-Meier curves. Moreover, real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting were performed to determine the mRNA and protein levels of glutamic-pyruvic transaminase (GPT) in GC and normal immortalized cell lines. In addition, cell viability, cell cycle distribution, migration and invasion were evaluated by cell counting kit-8, flow cytometry and transwell assays. Furthermore, we also conducted a retrospective analysis on 70 GC patients diagnosed and surgically treated in Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University between January 2017 to December 2020. The tumor and adjacent normal samples were collected from the patients to determine the potential association between the expression level of GPT and the clinical as well as pathological features of GC patients. RESULTS: We selected 19214 genes from the GSE183136 dataset, among which there were 250 downregulated genes and 401 upregulated genes in the tumor samples of stage III-IV in comparison to those in tumor samples of stage I-II with a P-value < 0.05. In addition, GO and KEGG results revealed that the various upregulated DEGs were mainly enriched in plasma membrane and neuroactive ligand-receptor interaction, whereas the downregulated DEGs were primarily enriched in cytosol and pancreatic secretion, vascular smooth muscle contraction and biosynthesis of the different cofactors. Furthermore, PPI networks were constructed based on the various upregulated and downregulated genes, and there were a total 15 upregulated and 10 downregulated hub genes. After a comprehensive analysis, several hub genes, including runt-related transcription factor 2 (RUNX2), salmonella pathogenicity island 1 (SPI1), lysyl oxidase (LOX), fibrillin 1 (FBN1) and GPT, displayed prognostic values. Interestingly, it was observed that GPT was downregulated in GC cells and its upregulation could suppress the malignant phenotypes of GC cells. Furthermore, the expression level of GPT was found to be associated with age, lymph node metastasis, pathological staging and distant metastasis (P < 0.05). CONCLUSION: RUNX2, SPI1, LOX, FBN1 and GPT were identified key hub genes in GC by bioinformatics analysis. GPT was significantly associated with the prognosis of GC, and its upregulation can effectively inhibit the proliferative, migrative and invasive capabilities of GC cells.

SELECTION OF CITATIONS
SEARCH DETAIL