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










Publication year range
1.
Antiviral Res ; 228: 105955, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964614

ABSTRACT

High levels of hepatitis B virus (HBV) surface antigen (HBsAg) in the blood of chronic HBV carriers are considered to drive the exhaustion of antigen-specific T and B lymphocytes and thus responsible for the persistence of infection. Accordingly, therapeutic elimination of HBsAg may facilitate the activation of adaptive antiviral immune responses against HBV and achieve a functional cure of chronic hepatitis B. We discovered recently that an amphipathic alpha helix spanning W156 to R169 of HBV small envelope (S) protein plays an essential role in the morphogenesis of subviral particles (SVPs) and metabolism of S protein. We thus hypothesized that pharmacological disruption of SVP morphogenesis may induce intracellular degradation of S protein and reduce HBsAg secretion. To identify inhibitors of SVP biogenesis, we screened 4417 bioactive compounds with a HepG2-derived cell line expressing HBV S protein and efficiently secreting small spherical SVPs. The screen identified 24 compounds that reduced intracellular SVPs and secreted HBsAg in a concentration-dependent manner. However, 18 of those compounds inhibited the secretion of HBsAg and HBeAg in HBV replicon transfected HepG2 cells at similar efficiency, suggesting each of those compounds may disrupt a common cellular function required for the synthesis and/or secretion of these viral proteins. Interestingly, lycorine more efficiently inhibited the secretion of HBsAg in HepG2 cells transfected with HBV replicons, HepG2.2.15 cells and HBV infected - HepG2 cells expressing sodium taurocholate cotransporting polypeptide (NTCP). The structure activity relationship and antiviral mechanism of lycorine against HBV have been determined.


Subject(s)
Antiviral Agents , Hepatitis B Surface Antigens , Hepatitis B virus , Humans , Hepatitis B virus/drug effects , Antiviral Agents/pharmacology , Hepatitis B Surface Antigens/metabolism , Hep G2 Cells , Virus Assembly/drug effects , Virion/drug effects , Drug Discovery , Virus Replication/drug effects , Small Molecule Libraries/pharmacology , Viral Envelope Proteins/metabolism , Hepatitis B e Antigens/metabolism
2.
Antiviral Res ; 221: 105782, 2024 01.
Article in English | MEDLINE | ID: mdl-38110058

ABSTRACT

Hepatitis B virus (HBV) chronically infects 296 million people worldwide and causes more than 820,000 deaths annually due to cirrhosis and hepatocellular carcinoma. Current standard-of-care medications for chronic hepatitis B (CHB) include nucleos(t)ide analogue (NA) viral DNA polymerase inhibitors and pegylated interferon alpha (PEG-IFN-α). NAs can efficiently suppress viral replication and improve liver pathology, but not eliminate or inactivate HBV covalently closed circular DNA (cccDNA). CCC DNA is the most stable HBV replication intermediate that exists as a minichromosome in the nucleus of infected hepatocyte to transcribe viral RNA and support viral protein translation and genome replication. Consequentially, a finite duration of NA therapy rarely achieves a sustained off-treatment suppression of viral replication and life-long NA treatment is most likely required. On the contrary, PEG-IFN-α has the benefit of finite treatment duration and achieves HBsAg seroclearance, the indication of durable immune control of HBV replication and functional cure of CHB, in approximately 5% of treated patients. However, the low antiviral efficacy and poor tolerability limit its use. Understanding how IFN-α suppresses HBV replication and regulates antiviral immune responses will help rational optimization of IFN therapy and development of novel immune modulators to improve the rate of functional cure. This review article highlights mechanistic insight on IFN control of HBV infection and recent progress in development of novel IFN regimens, small molecule IFN mimetics and combination therapy of PEG-IFN-α with new direct-acting antivirals and therapeutic vaccines to facilitate the functional cure of CHB.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Hepatitis C, Chronic , Liver Neoplasms , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Hepatitis B Surface Antigens , Hepatitis C, Chronic/drug therapy , Hepatitis B virus , Interferon-alpha/therapeutic use , Hepatitis B/drug therapy , DNA, Viral , Liver Neoplasms/drug therapy
3.
PLoS Pathog ; 18(1): e1010271, 2022 01.
Article in English | MEDLINE | ID: mdl-35061864

ABSTRACT

Flavivirus infection of cells induces massive rearrangements of the endoplasmic reticulum (ER) membrane to form viral replication organelles (ROs) which segregates viral RNA replication intermediates from the cytoplasmic RNA sensors. Among other viral nonstructural (NS) proteins, available evidence suggests for a prominent role of NS4B, an ER membrane protein with multiple transmembrane domains, in the formation of ROs and the evasion of the innate immune response. We previously reported a benzodiazepine compound, BDAA, which specifically inhibited yellow fever virus (YFV) replication in cultured cells and in vivo in hamsters, with resistant mutation mapped to P219 of NS4B protein. In the following mechanistic studies, we found that BDAA specifically enhances YFV induced inflammatory cytokine response in association with the induction of dramatic structural alteration of ROs and exposure of double-stranded RNA (dsRNA) in virus-infected cells. Interestingly, the BDAA-enhanced cytokine response in YFV-infected cells is attenuated in RIG-I or MAD5 knockout cells and completely abolished in MAVS knockout cells. However, BDAA inhibited YFV replication at a similar extent in the parent cells and cells deficient of RIG-I, MDA5 or MAVS. These results thus provided multiple lines of biological evidence to support a model that BDAA interaction with NS4B may impair the integrity of YFV ROs, which not only inhibits viral RNA replication, but also promotes the release of viral RNA from ROs, which consequentially activates RIG-I and MDA5. Although the innate immune enhancement activity of BDAA is not required for its antiviral activity in cultured cells, its dual antiviral mechanism is unique among all the reported antiviral agents thus far and warrants further investigation in animal models in future.


Subject(s)
Antiviral Agents/pharmacology , Benzodiazepines/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects , Yellow fever virus/drug effects , Cell Line , DEAD Box Protein 58/immunology , Humans , Immunity, Innate/immunology , Viral Nonstructural Proteins/drug effects , Yellow Fever/immunology , Yellow fever virus/immunology
4.
Animals (Basel) ; 11(8)2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34438618

ABSTRACT

The lymphoid organ is an essential part of the immune system involved in cellular and humoral immune responses in shrimp. However, its roles in the immune responses against different pathogens are still largely unclear. In the present study, transcriptomic analysis was applied to compare the differentially expressed genes (DEGs) in the lymphoid organ of shrimp after Vibrio or WSSV challenge. In total, 2127 DEGs were screened in the lymphoid organ of shrimp at 6 h post Vibrio parahaemolyticus injection, and 1569 DEGs were obtained at the same time after WSSV challenge. KEGG pathway enrichment analysis of these DEGs revealed that two significantly enriched pathways including "neuroactive ligand-receptor interaction" and "protein digestion and absorption" were responsive to both pathogens. In contrast, "lysosome" was the significantly enriched pathway only in Vibrio challenge whereas carbohydrate metabolism related pathways were the significantly enriched pathways only in WSSV challenge. Further analysis on immune-related DEGs showed that Vibrio challenge induced broad immune responses in the lymphoid organ including activation of several pattern recognition receptors, the proPO activating system, phagocytosis related genes, and immune effectors. In contrast, the immune responses seemed to be inhibited after WSSV infection. The data suggest that the shrimp lymphoid organ plays different functions in response to the infection of distinct pathogens at the early stage, which provides new insights into the immune functions of lymphoid organ in shrimp.

5.
Dev Comp Immunol ; 121: 104101, 2021 08.
Article in English | MEDLINE | ID: mdl-33862098

ABSTRACT

The cGAS-STING pathway plays essential roles in detecting cytosolic dsDNA and initiating antiviral and antibacterial responses in vertebrates. However, knowledge about its function in antiviral response of invertebrates is very limited. In the present study, a gene encoding a Mab21-containing protein, a cGAS homologue, was identified from a decapod crustacean Litopenaeus vannamei and designated as LvMab21cp. LvMab21cp was mainly distributed in intestine and hepatopancreas, showing similar expression profile with other genes in the cGAS-STING pathway, such as LvSTING and LvIRF. The expression levels of LvMab21cp, LvSTING and LvIRF were up-regulated in intestine and hepatopancreas of shrimp after white spot syndrome virus (WSSV) infection. Knockdown of LvMab21cp by dsRNA-mediated RNA interference could decrease the expression levels of its putative downstream genes, including LvSTING, LvIRF, LvVago4 and LvVago5, and enhance the in vivo propagation of WSSV in shrimp. Overexpression of LvMab21cp and LvSTING in HEK 293T cells activated the expression of mammalian IFNs upon simulation with interferon stimulatory DNA (ISD). These data suggest that LvMab21cp was a cGAS homologue, a member of the shrimp cGAS-STING pathway, and play an important role during WSSV infection. To our knowledge, this is the first report to show the role of the cGAS-STING pathway in the antiviral response of invertebrates, which will provide new insights into the innate immunity of invertebrates.


Subject(s)
Arthropod Proteins/metabolism , Immunity, Innate , Interferons/metabolism , Penaeidae/immunology , White spot syndrome virus 1/immunology , Animals , Arthropod Proteins/genetics , Gene Knockdown Techniques , HEK293 Cells , Hepatopancreas/immunology , Hepatopancreas/metabolism , Humans , Penaeidae/genetics , Penaeidae/virology , Signal Transduction/immunology
6.
Dev Comp Immunol ; 109: 103713, 2020 08.
Article in English | MEDLINE | ID: mdl-32304716

ABSTRACT

Leucine-rich repeat (LRR) is a vital structure in some pattern recognition receptors such as TLRs, NLRs and newly reported LRR-containing proteins. Apart from some limited reported LRR-containing proteins, most of LRR proteins, especially immune-related proteins, remain uncharacterized functionally. In the present study, a transmembrane protein containing several LRR motifs, designated as LvLRRm, was identified from the shrimp Litopenaeus vannamei. LvLRRm contained a long signal peptide, one LRRNT region, 12 LRR motifs, one LRRCT region and a transmembrane region. The transcripts of LvLRRm were widely distributed in all tested tissues of shrimp and they were responsive to Vibrio parahaemolyticus infection in several immune-related tissues including Oka, intestine, gill and hemocytes. Knockdown of LvLRRm by dsRNA interference led to a decreased survival rate of shrimp infected by Vibrio parahaemolyticus and an increased in vivo Vibrio propagation. Meanwhile, knockdown of LvLRRm also down-regulated the expression levels of genes involved in antibacterial immune signaling pathways, including the transcription factors LvDorsal and LvRelish, and several antimicrobial peptides. These data suggested that LvLRRm played important roles in shrimp against Vibrio infection, which was probably functioning through activation of antibacterial immune signaling pathways. The present study provided new evidence to elucidate the immune function of LRR-containing proteins in invertebrates.


Subject(s)
Arthropod Proteins/immunology , Membrane Proteins/immunology , Penaeidae/immunology , Proteins/immunology , Vibrio parahaemolyticus/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/genetics , Disease Resistance/genetics , Disease Resistance/immunology , Gene Expression Profiling , HEK293 Cells , Hemocytes/immunology , Hemocytes/metabolism , Hemocytes/microbiology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Leucine-Rich Repeat Proteins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Penaeidae/genetics , Penaeidae/microbiology , Phylogeny , Protein Binding , Protein Domains , Proteins/classification , Proteins/genetics , Sequence Homology, Amino Acid , Vibrio parahaemolyticus/physiology
7.
BMC Genomics ; 20(1): 247, 2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30922216

ABSTRACT

BACKGROUND: Functional communications between nervous, endocrine and immune systems are well established in both vertebrates and invertebrates. Circulating hemocytes act as fundamental players in this crosstalk, whose functions are conserved during the evolution of the main groups of metazoans. However, the roles of the neuroendocrine-immune (NEI) system in shrimp hemocytes during pathogen infection remain largely unknown. RESULTS: In this study, we sequenced six cDNA libraries prepared with hemocytes from Litopenaeus vannamei which were injected by WSSV (white spot syndrome virus) or PBS for 6 h using Illumina Hiseq 4000 platform. As a result, 3444 differentially expressed genes (DEGs), including 3240 up-regulated genes and 204 down-regulated genes, were identified from hemocytes after WSSV infection. Among these genes, 349 DEGs were correlated with innate immunity and categorized into seven groups based on their predictive function. Interestingly, 18 genes encoded putative neuropeptide precursors were induced significantly by WSSV infection. Furthermore, some genes were mapped to several typical processes in the NEI system, including proteolytic processing of prohormones, amino acid neurotransmitter pathways, biogenic amine biosynthesis and acetylcholine signaling pathway. CONCLUSIONS: The data suggested that WSSV infection triggers the activation of NEI in shrimp, which throws a light on the pivotal roles of NEI system mediated by hemocytes in shrimp antiviral immunity.


Subject(s)
Arthropod Proteins/genetics , Gene Expression Profiling/veterinary , Hemocytes/immunology , Penaeidae/virology , Animals , Gene Expression Regulation , High-Throughput Nucleotide Sequencing/veterinary , Molecular Sequence Annotation , Neurosecretory Systems/immunology , Penaeidae/genetics , Penaeidae/immunology , Sequence Analysis, RNA/veterinary , White spot syndrome virus 1/immunology , White spot syndrome virus 1/pathogenicity
8.
Dev Comp Immunol ; 67: 77-85, 2017 02.
Article in English | MEDLINE | ID: mdl-27840294

ABSTRACT

c-Jun N-terminal kinase (JNK) is a universal and essential subgroup of the mitogen-activated protein kinase (MAPK) superfamily, which is highly conserved from yeast to mammals and functions in a variety of physiological and pathological processes. In this study, we report the first oyster JNK gene homolog (ChJNK) and its biological functions in the Hong Kong oyster Crassostrea hongkongensis. The ChJNK protein consists of 383 amino acids and contains a conserved serine/threonine protein kinase (S_TKc) domain with a typical TPY motif. Phylogenetic analysis revealed that ChJNK shared a close evolutionary relationship with Crassostrea gigas JNK. Quantitative RT-PCR analyses revealed broad expression patterns of ChJNK mRNA in various adult tissues and different embryonic and larval stages of C. hongkongensis. When exposed to Vibrio alginolyticus or Staphylococcus haemolyticus, ChJNK mRNA expression levels were significantly up-regulated in the hemocytes and gills in a time-dependent manner. Additionally, subcellular localization studies that ChJNK is a cytoplasm-localized protein, and that its overexpression could significantly enhance the transcriptional activities of AP-1-Luc in HEK293T cells. In summary, this study provided the first experimental demonstration that oysters possess a functional JNK that participates in host defense against bacterial infection in C. hongkongensis.


Subject(s)
Crassostrea/immunology , Cytoplasm/metabolism , Hemocytes/immunology , MAP Kinase Kinase 4/metabolism , Staphylococcus hominis/immunology , Vibrio Infections/immunology , Vibrio alginolyticus/immunology , Animals , Bacterial Infections , HEK293 Cells , Hemocytes/microbiology , Humans , MAP Kinase Kinase 4/genetics , Phylogeny , Protein Transport , Transcriptome , Up-Regulation
9.
Fish Shellfish Immunol ; 59: 288-297, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27666188

ABSTRACT

Apoptosis plays an important role in homeostasis of the immune systems. The tumor necrosis factor receptors (TNFRs) play critical roles in the extrinsic apoptosis pathways and in determining cell fate. In this study, four death receptors (DR) named ChEDAR, ChTNFR27, ChTNFR5, and ChTNFR16 were identified from the oyster Crassostrea hongkongensis. These ChDRs proteins had 382, 396, 414 and 384 amino acids, respectively, with the typical domains of death receptors, such as the signal peptide (SP), transmembrane helix region (TM) and death domains. Phylogenetic analysis showed that the ChDR proteins clustered into three distinct groups, indicating that these subfamilies had common ancestors. mRNA expression of the ChDRs were detected in all 8 of the selected oyster tissues and at different stages of development. Furthermore, expression of all the genes was increased in the hemocytes of oysters challenged with pathogens or air stress. Fluorescence microscopy revealed that the full-length proteins of the ChDRs were located in the plasma membrane of HEK293T cells. Over-expression of the ChDRs activated the NF-κB-Luc reporter in HEK293T cells in a dose-dependent manner. These results indicate that the ChDRs may play important roles in the extrinsic apoptotic pathways in oysters.


Subject(s)
Crassostrea/genetics , Crassostrea/immunology , Gene Expression Regulation, Developmental , Immunity, Innate , Receptors, Tumor Necrosis Factor/genetics , Amino Acid Sequence , Animals , Apoptosis/immunology , Base Sequence , Cloning, Molecular , Crassostrea/classification , Crassostrea/microbiology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Evolution, Molecular , Organ Specificity , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/immunology , Saccharomyces cerevisiae/physiology , Sequence Alignment , Signal Transduction , Staphylococcus haemolyticus/physiology , Vibrio alginolyticus/physiology
10.
Fish Shellfish Immunol ; 56: 106-110, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27393236

ABSTRACT

TRIM proteins are a group of highly conserved proteins participating in a variety of biological processes such as regulation of development, apoptosis, and innate immunity. However, the functions of these proteins in the mollusk are still poorly understood. In the present study, a TRIM9 homolog (named ChTRIM9) was first identified from a transcript-ome library in the Hong Kong oyster Crassostrea hongkongensis. The full-length cDNA of ChTRIM9 is 2928 bp and has a predicted Open Reading Frame ORF) encoding 721 amino acids, encoding a putative 80.2 kDa protein. SMART analysis indicated that ChTRIM9 contains the three typical TRIM domains, a RING finger, two B-boxes, and a coiled-coil domain in the N-terminal region, whereas the C-terminal region contains a SPRY domain. qRT-PCR analysis revealed a ubiquitous presence of ChTRIM9, with the highest expression in the gills. Upon bacterial challenge in vivo, the ChTRIM9 transcripts in hemocytes were significantly down-regulated, indicating its involvement in signal transduction in immune response of oysters. Furthermore, ChTRIM9 was found to be localized mainly in the cytoplasm, and its over-expression inhibited the transcriptional activity of the NF-κB gene in HEK293T cells, demonstrating its negative role in regulating NF-κB signaling.


Subject(s)
Crassostrea/genetics , Crassostrea/immunology , Tripartite Motif Proteins/genetics , Animals , Cloning, Molecular , Crassostrea/metabolism , Crassostrea/microbiology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Immunity, Innate , NF-kappa B , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, Protein , Tripartite Motif Proteins/chemistry , Tripartite Motif Proteins/metabolism , Vibrio alginolyticus/physiology
11.
Fish Shellfish Immunol ; 47(2): 1015-23, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26549179

ABSTRACT

G-protein-coupled receptors (GPCRs) are the largest class of cell-surface receptors and play crucial roles in virtually every organ system. As one of the major downstream effectors of GPCRs, Akt can acquire information from the receptors and coordinate intracellular responses for many signaling pathways, through which the serine/threonine kinase masters numerous aspects of biological processes, such as cell survival, growth, proliferation, migration, angiogenesis, and metabolism. In the present study, we have characterized the first Akt1 ortholog in mollusks using the Hong Kong oyster, Crassostrea hongkongensis (designed ChAkt1). The full-length cDNA is 2223 bp and encodes a putative protein of 493 amino acids that contains an amino-terminal pleckstin homology (PH) domain, a central catalytic domain, and a carboxy-terminal regulatory domain. Quantitative real-time PCR analysis showed that ChAkt1 mRNA is broadly expressed in various tissues and during different stages of the oyster's embryonic and larval development. Upon exposure to two stressors (microbial infection and heat shock), the expression level of ChAkt1 mRNA increases significantly. Furthermore, ChAkt1 is located in the cytoplasm in HEK293T cells, where the over-expression of ChAkt1 regulates the transcriptional activities of NF-κB and p53 reporter genes. Taken together, our results indicate that ChAkt1 most likely plays a central role in response to various stimuli in oysters and has a particular response to microbial pathogens and high temperature.


Subject(s)
Crassostrea/physiology , Heat-Shock Response , Immunity, Innate , Proto-Oncogene Proteins c-akt/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Crassostrea/genetics , Crassostrea/immunology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Molecular Sequence Data , Organ Specificity , Phylogeny , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Sequence Alignment
12.
Dev Comp Immunol ; 53(1): 244-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26204814

ABSTRACT

Evolutionarily conserved signaling intermediate in Toll pathways (ECSIT) is a multifunctional adaptor protein that plays a key role in the regulation of the oxidative phosphorylation (OXPHOS) system, bone morphogenetic protein (BMP) pathway and Toll-like receptor (TLR) signaling pathway in mammals. However, the function of ECSIT homologs in mollusks, the second most diverse group of animals, is not well understood. In this study, we identified an ECSIT homolog in the Hong Kong oyster Crassostrea hongkongensis (ChECSIT) and investigated its biological functions. The full-length cDNA of ChECSIT is 1734 bp and includes an open reading frame (ORF) of 1074 bp that encodes a polypeptide of 451 amino acids. The predicted ChECSIT protein shares similar structural characteristics with other known ECSIT family proteins. Quantitative real-time PCR analysis revealed that ChECSIT mRNA is broadly expressed in all of the examined tissues and at different stages of embryonic development; its transcript level could be significantly up-regulated by challenge with microorganisms (Vibrio alginolyticus, Staphylococcus haemolyticus and Saccharomyces cerevisiae). In addition, ChECSIT was found to be located primarily in the cytoplasm, and its overexpression stimulated the transcriptional activity of an NF-κB reporter gene in HEK293T cells. These findings suggest that ChECSIT might be involved in embryogenesis processes and immune responses in C. hongkongensis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Crassostrea/immunology , Immunity, Innate/immunology , Toll-Like Receptors/metabolism , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Sequence , Animals , Base Sequence , Bone Morphogenetic Proteins/metabolism , Cell Line , Cloning, Molecular , Crassostrea/genetics , HEK293 Cells , Humans , Molecular Sequence Data , NF-kappa B/genetics , Open Reading Frames/genetics , Oxidative Phosphorylation , RNA, Messenger/genetics , Saccharomyces cerevisiae/immunology , Sequence Alignment , Sequence Analysis, DNA , Signal Transduction/immunology , Staphylococcus haemolyticus/immunology , Transcription, Genetic/genetics , Transcriptional Activation , Vibrio alginolyticus/immunology
13.
Dev Comp Immunol ; 51(1): 194-201, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25841657

ABSTRACT

The transcription factor Fos is a member of one of the best-studied AP-1 sub-families and has been implicated in a wide variety of biological processes, including the regulation of apoptosis, immune responses and cytokine production. In this report, a novel mollusk Fos (referred to as ChFos) gene was cloned and characterized from the Hong Kong oyster, Crassostrea hongkongensis. The deduced ChFos protein sequence comprised 333 amino acids and shared significant homology with invertebrate homologs. Phylogenetic analysis revealed that ChFos clusters with Fos from Crassostrea gigas and Crassostrea ariakensis. Quantitative real-time PCR analysis revealed that ChFos mRNA was broadly expressed in all tested tissues and during different stages of the oyster's embryonic and larval development. In addition, the expression of ChFos mRNA was significantly up-regulated under challenge with microorganisms (Vibrio alginolyticus, Staphylococcus haemolyticus and Saccharomyces cerevisiae) and pathogen-associated molecular patterns (PAMPs: LPS, PGN and polyI:C). Moreover, fluorescence microscopy showed that ChFos protein is localized in the nucleus in HEK293T cells. Reporter assays suggested that ChFos may act as an efficient transcription activator in the regulation of AP-1-responsive gene expression through interaction with ChJun. Overall, this study presents the first experimental evidence of the presence and functional characteristics of Fos in mollusks, which reveals its involvement in host protection against immune challenge in the oyster.


Subject(s)
Cell Nucleus/metabolism , Crassostrea/immunology , Infections/immunology , Oncogene Proteins v-fos/metabolism , Saccharomyces cerevisiae/immunology , Staphylococcus haemolyticus/immunology , Vibrio/immunology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Immunity , Molecular Sequence Data , Oncogene Proteins v-fos/genetics , Phylogeny , Sequence Homology, Amino Acid , Transcriptome , Up-Regulation
14.
Fish Shellfish Immunol ; 42(2): 316-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25463295

ABSTRACT

Prostaglandin E receptor 4 (PTGER4) is an essential receptor that can detect various physiological and pathological stimuli and has been implicated in a wide variety of biological processes, including the regulation of immune responses, cytokine production, and apoptosis. In this report, the first mollusk PTGER4, referred to as ChPTGER4, was cloned and characterized from the Hong Kong oyster Crassostrea hongkongensis. Its full-length cDNA is 1734 bp in length, including 5'- and 3'-untranslated region (UTRs) of 354 bp and 306 bp, respectively, and an open reading frame (ORF) of 1074 bp. ChPTGER4 comprises 357 amino acids and shares significant homology with its vertebrate homologs. The results of phylogenetic analysis revealed that ChPTGER4 clusters with PTGER4 from the Pacific oyster. In addition, quantitative real-time PCR analysis revealed that ChPTGER4 was constitutively expressed in all tissues examined and that its expression was significantly up-regulated in hemocytes and gills following challenge by pathogens (Vibrio alginolyticus, Staphylococcus haemolyticus and Saccharomyces cerevisiae) and pathogen-associated molecular patterns (PAMPs: lipopolysaccharide (LPS) and peptidoglycan (PGN). Moreover, fluorescence microscopy analysis revealed that ChPTGER4 localized to the membrane, and its overexpression significantly enhanced NF-κB reporter gene activation in the HEK293T cell line. In summary, this study provides the first experimental evidence of a functional PTGER4 in mollusks, which suggests its involvement in the innate immune response in oyster.


Subject(s)
Crassostrea/immunology , Crassostrea/microbiology , Immunity, Innate , Receptors, Prostaglandin E, EP4 Subtype/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Crassostrea/genetics , Crassostrea/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Lipopolysaccharides/pharmacology , Molecular Sequence Data , Peptidoglycan/pharmacology , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Saccharomyces cerevisiae/physiology , Sequence Alignment , Staphylococcus haemolyticus/physiology , Vibrio alginolyticus/physiology
15.
Fish Shellfish Immunol ; 42(2): 225-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25463302

ABSTRACT

Apoptosis has been primarily investigated in mammals, and little is known about apoptosis in mollusks. The proteins Bax and Bak play critical roles in the mitochondrial apoptosis pathway and in determining cell fate. In this study, ChBax and ChBak, homologs of the well-known Bax and Bak proteins, were identified from the oyster Crassostrea hongkongensis. The ChBax/Bak proteins consist of 207/232 amino acids with the typical domains found in BCL-2 family members. ChBax and ChBak mRNA expression were detected in all 8 of the selected oyster tissues and at the different stages of development. Fluorescence microscopy revealed that the full-length proteins of ChBax/Bak were located in the cytoplasm and mitochondrial outer membrane, of HEK293T cells, respectively. Furthermore, both of the genes' expression levels were found to increase in the hemocytes of oysters challenged with pathogens. The over-expression of ChBax or ChBak activates the p53-Luc reporter gene in HEK293T cells in a dose-dependent manner. These results indicate that ChBax and ChBak may play important roles in the mitochondrial apoptotic pathway in oysters.


Subject(s)
Apoptosis/genetics , Crassostrea/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Crassostrea/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Genes, Reporter , HEK293 Cells , Hemocytes/metabolism , Humans , Mitochondria/physiology , Molecular Sequence Data , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
16.
Fish Shellfish Immunol ; 40(2): 514-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25120216

ABSTRACT

Ficolins are a group of soluble animal proteins with multiple roles in innate immunity. These proteins recognize and bind carbohydrates in pathogens and activate the complement system, leading to opsonization, leukocyte activation, and direct pathogen killing, which have been reported in many animal species but might not be present in the shellfish lineage. In the present study, we identified the first fibrinogen-related protein from the oyster, Crassostrea hongkongensis. This novel ficolin-like protein contains a typical signal peptide and a fibrinogen-related domain (designated ChFCN) at the N and C termini, respectively, but does not contain the additional collagen-like domain of ficolins. The full-length cDNA of ChFCN is 1105 bp, encoding a putative protein of 297 amino acids with the molecular weight of 35.5 kD. ChFCN is ubiquitously expressed in selected tissues, with the highest expression level observed in the gills. The temporal expression of ChFCN following microbe infection shows that the expression of ChFCN in hemocytes increases at 3 h post-challenge. The ChFCN protein expression was also examined, and fluorescence microscopy revealed that deChFCN (truncated signal peptide) is located in the cytoplasm of HeLa cells. Full-length ChFCN was detected in the medium supernatant by western blot analysis. Recombinant ChFCN proteins with the molecular weight about 50 kD bind Saccharomyces cerevisiae, Staphylococcus haemolyticus or Escherichia coli K-12, but not those from Vibrio alginolyticus. Furthermore, the rChFCN protein could agglutinate Gram-negative bacteria E. coli K-12 and enhance the phagocytosis of C. hongkongensis hemocytes in vitro. These results indicate that ChFCN might play an important role in the immunity response of oysters.


Subject(s)
Crassostrea/genetics , Gene Expression Regulation , Immunity, Innate , Lectins/genetics , Amino Acid Sequence , Animals , Bacterial Physiological Phenomena , Base Sequence , Crassostrea/classification , Crassostrea/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Lectins/chemistry , Lectins/metabolism , Molecular Sequence Data , Phylogeny , Sequence Alignment , Ficolins
SELECTION OF CITATIONS
SEARCH DETAIL