Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Viruses ; 14(5)2022 04 29.
Article in English | MEDLINE | ID: mdl-35632672

ABSTRACT

Long noncoding RNAs (lncRNAs) function as micro regulators to impact gene expression after multiple pathogen infections, which have been largely studied in the last few years. Although lncRNA studies on lower vertebrates have received less attention than those on mammals, current studies suggest that lncRNA plays an important role in the immune response of fish to pathogen infections. Here, we studied the effect of MIR122HG as the host gene of miR-122 and indirectly negatively regulate MAVS-mediated antiviral immune responses in miiuy croaker (Miichthysmiiuy). We found that poly(I:C) significantly increases the host MIR122HG expression. The increased MIR122HG expression inhibited the production of the antiviral immune-related genes IFN-1, ISG15 and Viperin upon SCRV treatment. In addition, MIR122HG can act as a pivotally negative regulator involved in the MAVS-mediated NF-κB and IRF3 signaling pathways, which can effectively avoid an excessive immune response. Additionally, we found that MIR122HG can promote the replication of SCRV. Our study provides evidence about the involvement of lncRNAs in the antiviral immune response of fish and broadens the understanding of the function of lncRNAs as a precursor miRNA in teleost fish.


Subject(s)
MicroRNAs , Perciformes , RNA, Long Noncoding , Animals , Antiviral Agents/metabolism , Fish Proteins/genetics , Fish Proteins/metabolism , Immunity , Mammals , MicroRNAs/metabolism , Perciformes/genetics , Perciformes/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
2.
Fish Shellfish Immunol ; 124: 21-27, 2022 May.
Article in English | MEDLINE | ID: mdl-35367373

ABSTRACT

Interferon-mediated innate immune response is the first line of defense against foreign pathogen infection. Overexpression of MITA can activate the expression of interferon and promote the innate immune response of the body to the virus. These innate immune responses are tightly controlled to prevent the host from over-immunizing itself. In this study, we reported that structurally highly conserved PCNA negatively regulates MITA. PCNA overexpression can promote MITA degradation and block the expression of interferon, while the autophagy inhibitor 3-MA significantly inhibits MITA degradation, indicating that PCNA can degrade MITA through the autophagy pathway. PCNA inhibits interferon production by targeting MITA and avoids excessive immune response. In summary, our results indicate that PCNA is involved in the immune response by degrading MITA through the autophagy pathway, which will provide new ideas for further studies on the regulatory mechanism of immune signaling pathways in lower vertebrates.


Subject(s)
Perciformes , Animals , Autophagy , Immunity, Innate/genetics , Interferons/genetics , Perciformes/genetics , Proliferating Cell Nuclear Antigen
3.
Infect Immun ; 90(5): e0012022, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35416706

ABSTRACT

The innate immune response is the first line of defense against pathogen infection. Eradication of pathogen infection requires appropriate immune and inflammatory responses, but excessive inflammation may cause inflammatory and autoimmune diseases. MicroRNAs (miRNAs) are a group of small noncoding RNAs, and accumulating evidence has shown that in mammals, they can act as negative regulators that participate in the regulation of inflammation and immune responses. However, the miRNA-mediated immune regulation networks in the inflammatory responses of lower vertebrates are largely unknown. In this study, we report an miRNA, miR-132, identified from miiuy croaker, that acts as a negative regulator in the host's bacterium-induced inflammatory response. We found that miR-132 expression was dramatically increased upon infection by the Gram-negative bacterium Vibrio harveyi and lipopolysaccharide (LPS). Inducible miR-132 inhibits the production of inflammatory cytokines by targeting tumor necrosis factor receptor-associated factor 6 (TRAF6), transforming growth factor-activated protein kinase 1 (TAK1), and TAK1 binding protein 1 (TAB1), thus avoiding an excessive inflammatory response. Furthermore, we demonstrate that miR-132 modulates the inflammatory response through a TRAF6-, TAK1-, and TAB1-mediated NF-κB signaling pathway. These results collectively reveal that miR-132 plays a negative regulatory role in the host antibacterial immune response, which will help to gain insight into the intricate network of host resistance to pathogen infection in lower vertebrates.


Subject(s)
MicroRNAs , TNF Receptor-Associated Factor 6 , Animals , Cytokines/metabolism , Fishes/genetics , Fishes/metabolism , Inflammation , Mammals , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism
4.
Dev Comp Immunol ; 132: 104401, 2022 07.
Article in English | MEDLINE | ID: mdl-35339534

ABSTRACT

MyD88 is a typical street protein of the TLRs signaling pathway and is a central player in innate immune signaling, which can regulate the NF-κB signaling pathway and promote downstream inflammatory factors. However, studies on the molecular mechanisms of the MyD88-mediated NF-κB signaling pathway in teleosts have been poorly reported. In this study, we report that Zw10 targets MyD88 to inhibit NF-κB activation. Zw10 inhibits cell proliferation and MyD88-mediated innate immunity in fish. Zw10 interacts with MyD88, and its Δ2 domain is very critical for MyD88 degradation. In addition, we found that Zw10 degrade MyD88 by autophagy, thereby negatively regulating the MyD88-mediated NF-κB signaling pathway. This study not only enriches the research on the innate immunity of teleost fish, but also provides insights for the regulating mechanism for mammals.


Subject(s)
Myeloid Differentiation Factor 88 , NF-kappa B , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy , Fish Proteins/metabolism , Fishes/metabolism , Mammals , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Signal Transduction
5.
Fish Shellfish Immunol ; 123: 94-101, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35240295

ABSTRACT

Accumulating evidence has demonstrated that microRNAs (miRNAs) regulate various physiological and pathological processes at the transcriptional level, thus called novel regulators in immune response. In this study, we used bioinformatics and functional experiments to determine the role of miR-103 and miR-190 in the regulation of IL-1R1 gene involved in the immune and inflammatory responses in miiuy croakers. First, we predicted the target genes of miR-103 and miR-190 through bioinformatics and found that IL-1R1 is a direct target gene of miR-103 and miR-190. This was further confirmed by the dual-luciferase reporter assay that the over-expression of miR-103, miR-190 mimics and the pre-miR-103, pre-miR-190 plasmids inhibit the luciferase levels of the wild-type of IL-1R1 3'UTR. miR-103 and miR-190 inhibitors increase the luciferase levels of IL-1R1-3'UTR. Additionally, we found that miR-103 and miR-190 could negatively regulate the mRNA expression of IL-1R1. Importantly, we demonstrated that miR-103 and miR-190 significantly inhibit the NF-κB signaling pathway by targeting IL-1R1 upon LPS stimulation. Collectively, these results provide strong evidence for an important regulatory mechanism of miR-103 and miR-190 targeting the IL-1R1 gene, thereby preventing excessive inflammatory immune responses from causing autoimmunity.


Subject(s)
MicroRNAs , Perciformes , 3' Untranslated Regions , Animals , Gene Expression Regulation , Immunity , MicroRNAs/metabolism , NF-kappa B/metabolism
6.
Dev Comp Immunol ; 126: 104259, 2022 01.
Article in English | MEDLINE | ID: mdl-34536468

ABSTRACT

MDA5 is a member of retinoic acid-inducible gene I (RIG-I)-like receptors (RLR receptors), which may play a crucial role in the immune regulation process. Recently, microRNAs (miRNAs) have been shown to act as an important regulator in the RLRs signaling pathway. Additionally, the MDA5 gene, as a significant cytosolic pathogen recognition receptor (PRR), its characteristics and functions have been extensively investigated, while less research has been done on the mechanisms of MDA5-miRNA mediated gene regulation. In this study, the evolution and functional characterization of MDA5 from miiuy croaker (mmiMDA5) were characterized. Comparative genomic analysis demonstrated that the ascidiacea and superclass do not have the MDA5 gene in the process of evolution. MDA5 contains four structural domains: CARD, ResIII, Helicase C, and RIG-I C-RD. The MDA5 was ubiquitously expressed in all tested miiuy croaker tissues. Moreover, the expressions were significantly up-regulated after stimulation with poly (I: C), which indicated that MDA5 might be involved in the antiviral immune response. The bioinformatics predicted programs have indicated that miR-203 has a direct negative regulatory effect on MDA5 in miiuy croaker. Furthermore, the dual-luciferase reporter assay have showed that miR-203 was the direct negative regulator of MDA5 in miiuy croaker. This study is the first to demonstrate that miRNA can suppress cytokines by regulating the RLR signaling pathway in teleost fish, providing some new ideas for studying miRNA-mediated regulation of immune responses in mammals.


Subject(s)
MicroRNAs , Perciformes , Animals , Fish Proteins/metabolism , Mammals/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Poly I-C , Signal Transduction/genetics
7.
Drug Chem Toxicol ; 45(3): 1427-1431, 2022 May.
Article in English | MEDLINE | ID: mdl-34711124

ABSTRACT

The present study explored the effects of combination of Tinospora cordifolia and Arabinogalactan on surface membrane dynamics and programmed cell deaths in rat model of lung cancer. The rats were divided into different groups namely normal control, benzo(a)pyrene (BP) treated, BP + Tinospora cordifolia (TC)-treated, BP + Arabinogalactan (A)-treated and BP + TC + A-treated groups. Significant changes were observed in the membrane dynamics of rats treated with BP. The carcinogen treatment demonstrated a marked decrease in membrane microviscosity. Also, excimer/monomer ratio and fluidity parameters of BP treated rats showed significant rise. On the other hand, combination of Tinospora cordifolia and Arabinogalactan improvised surface membrane dynamics. Moreover, micronuclei formation along with protein expression of bcl-2 showed significant increase in the lungs of BP treated rats. The combined treatment of Tinospora cordifolia and Arabinogalactan moderated the micronuclei formation in BP treated rats. Also, the combined treatment regulated the protein expressions of bcl-2 in BP-treated rats. As a result, marked improvement was noticed in apoptosis of BP treated cells treated with combination treatment. This study concludes that the Tinospora cordifolia and Arabinogalactan in combination improve the surface membrane dynamics and apoptosis in BP-treated rats.


Subject(s)
Tinospora , Animals , Benzo(a)pyrene/toxicity , Carcinogenesis , Galactans , Lung , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-bcl-2 , Rats
8.
Infect Immun ; 90(1): e0058521, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34748368

ABSTRACT

Upon recognition of pathogen components by pattern recognition receptors, cells could be activated to produce inflammatory cytokines and type I interferons. The inflammation is tightly modulated by the host to prevent inappropriate inflammatory responses. MicroRNAs (miRNAs) are noncoding small RNAs that can inhibit gene expression and participate in various biological functions, including maintaining a balanced immune response in the host. To maintain the balance of the immune response, these pathways are closely regulated by the host to prevent inappropriate reactions of the cells. However, in lower vertebrates, the miRNA-mediated inflammatory response regulatory networks remain largely unknown. Here, we report that two miRNAs, i.e., miR-20-1 and miR-101a, were identified as negative regulators in teleost inflammatory responses. Initially, we found that both miR-20-1 and miR-101a dramatically increased after lipopolysaccharide (LPS) stimulation and Vibrio harveyi infection. Upregulated miR-20-1 and miR-101a inhibited LPS-induced inflammatory cytokine production by targeting tumor necrosis factor receptor-associated factor 6 (TRAF6), thus avoiding excessive inflammation. Moreover, miR-20-1 and miR-101a regulate the inflammatory responses through the TRAF6-mediated NF-κB signaling pathway. Collectively, these data indicate that miR-20-1 and miR-101a act as negative regulators by regulating the TRAF6-mediated NF-κB signaling pathway and participate in host antibacterial immune responses, which will provide new insights into the intricate networks of the host-pathogen interactions in the lower vertebrates.


Subject(s)
Fish Diseases/etiology , Fish Diseases/metabolism , Inflammation/veterinary , MicroRNAs/genetics , NF-kappa B/metabolism , Perciformes/genetics , Perciformes/metabolism , TNF Receptor-Associated Factor 6/genetics , Animals , Cytokines/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/adverse effects , Models, Biological , RNA Interference , Signal Transduction
9.
Front Immunol ; 12: 734520, 2021.
Article in English | MEDLINE | ID: mdl-34956174

ABSTRACT

Upon recognition of bacterial or viral components by Toll-like receptors (TLRs), cells could be activated to induce a series of reactions to produce inflammatory cytokines, type I interferon (IFN), and IFN stimulating genes (ISG). MicroRNAs (miRNAs) are an important regulatory molecules that are widely involved in the regulatory networks of mammalian inflammation and immune responses; however, in lower vertebrates, the regulatory network of miRNA-mediated immune responses is poorly understood. Here, we report two miRNAs form Miichthys miiuy, namely, miR-181b-2 and miR-21-1, that play a negative role in host antiviral and antibacterial immunity. We found that miR-181b-2 and miR-21-1 are abundantly expressed in gram-negative bacteria, as well as RNA rhabdovirus infection. Inducible miR-181b-2 and miR-21-1 suppress the production of inflammatory cytokines and type I IFN by targeting TRIF, thereby avoiding excessive inflammation. We further revealed that miR-181b-2 and miR-21-1 modulate antibacterial and antiviral immunity through the TRIF-mediated NF-κB and IRF3 signaling pathways. The overall results indicate that miR-181b-2 and miR-21-1 act as negative feedback regulators and participate in host antibacterial and antiviral immune responses; this finding could provide information for a deeper understanding of the resistance of lower vertebrates to the invasion of pathogens and to avoidance of excessive immunity.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Interferon Regulatory Factor-3/immunology , MicroRNAs/immunology , NF-kappa B/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation/immunology , MicroRNAs/genetics , Perciformes , RNA, Messenger/genetics , RNA, Messenger/immunology
10.
Fish Shellfish Immunol ; 118: 66-71, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34474149

ABSTRACT

The inflammatory response is a protective process to clear detrimental stimuli, constitutes the defense against infectious pathogens. Clearing pathogen infection requires appropriate immune and inflammatory response, but excessive inflammatory response can lead to uncontrolled inflammation, autoimmune disease, or pathogen transmission. Accumulating evidences show that miRNAs are important and multifunctional regulators of innate immunity and inflammation. However, in the inflammatory response of lower vertebrates, the miRNAs regulatory networks are largely unknown. In this study, a combination of bioinformatics and experimental techniques were used to investigate the functions of miR-148. IL-1ß is a hypothetical target gene of miR-148 predicted by bioinformatics. In addition, dual-luciferase reporter gene experiment was used to verify the targeting effect of miR-148 on IL-1ß-3'UTR. miR-148 inhibits IL-1ß expression in a dose-dependent manner at protein and mRNA levels. It is important that miR-148 participates in regulation of LPS-induced the NF-κB signaling pathway by inhibiting IL-1ß. These results will improve our understanding of the regulation of miRNAs in fish on the immune response.


Subject(s)
MicroRNAs , Perciformes , Animals , Inflammation , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Perciformes/immunology , Signal Transduction
11.
Dev Comp Immunol ; 117: 103976, 2021 04.
Article in English | MEDLINE | ID: mdl-33347907

ABSTRACT

The inflammatory response is a self-defense process that fights the pathogen invasion by eliminating harmful stimuli. However, excessive inflammation may disrupt immune homeostasis, even causing chronic inflammation or autoimmune diseases. MicroRNAs (miRNAs) are a crucial regulator that can negatively regulate gene expression and participate in multiple biological processes of growth, development, and immune response in organisms. However, the miRNA-mediated modulation networks of inflammatory responses remain largely unclear in lower vertebrates. In this study, miR-128 was identified as a negative regulator to participate in the NF-κB signaling pathway by targeting TAB2 in miiuy croaker. First, we predicted target genes of miR-128 through the bioinformatics software programs and found that TAB2 is a direct target of miR-128. We also found that miR-128 can inhibit TAB2 expression at the mRNA and protein levels. Besides, upon LPS stimulation, miR-128 inhibits the expression of inflammatory cytokines by targeting TAB2 to avoid excessive inflammation. Particularly, we found that miR-128 can regulate TAB2-mediated NF-κB signaling pathways. In summary, our results indicate that miR-128 plays a critical role in suppressing inflammatory responses by regulating the TAB2-mediated NF-κB signaling pathway in miiuy croaker.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Fish Proteins/immunology , Immunity/immunology , Inflammation/immunology , MicroRNAs/immunology , Perciformes/immunology , 3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Blotting, Western , Cell Line , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Regulation/immunology , Immunity/genetics , Inflammation/genetics , Inflammation/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Perciformes/genetics , Perciformes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Signal Transduction/immunology
12.
Front Genet ; 10: 779, 2019.
Article in English | MEDLINE | ID: mdl-31552091

ABSTRACT

Cold acclimation and vegetative/reproductive transition are two important evolutionary adaptive mechanisms for winter wheat surviving the freezing temperature in winter and successful seeds setting in the next year. MicroRNA (miRNA) is a class of regulatory small RNAs (sRNAs), which plays critical roles in the growth and development of plants. However, the regulation mechanism of miRNAs during cold acclimation and vegetative/reproductive transition of winter wheat is not much understood. In this study, four sRNA libraries from leaves of winter wheat grown in the field at the three-leaf stage, winter dormancy stage, spring green-up stage, and jointing stage were analyzed to identify known and novel miRNAs and to understand their potential roles in the growth and development of winter wheat. We examined miRNA expression using a high-throughput sequencing technique. A total of 373 known, 55 novel, and 27 putative novel miRNAs were identified. Ninety-one miRNAs were found to be differentially expressed at the four stages. Among them, the expression of six known and eight novel miRNAs was significantly suppressed at the winter dormancy stage, whereas the expression levels of seven known and eight novel miRNAs were induced at this stage; three known miRNAs and three novel miRNAs were significantly induced at the spring green-up stage; six known miRNAs were induced at the spring green-up stage and reached the highest expression level at the jointing stage; and 20 known miRNAs and 10 novel miRNAs were significantly induced at the jointing stage. Expression of a number of representative differentially expressed miRNAs was verified using quantitative real-time polymerase chain reaction (qRT-PCR). Potential target genes for known and novel miRNAs were predicted. Moreover, six novel target genes for four Pooideae species-specific miRNAs and two novel miRNAs were verified using the RNA ligase-mediated 5'-rapid amplification of cDNA ends (RLM-5'RACE) technique. These results indicate that miRNAs are key non-coding regulatory factors modulating the growth and development of wheat. Our study provides valuable information for in-depth understanding of the regulatory mechanism of miRNAs in cold acclimation and vegetative/reproductive transition of winter wheat grown in the field.

13.
Dev Comp Immunol ; 96: 47-50, 2019 07.
Article in English | MEDLINE | ID: mdl-30826381

ABSTRACT

microRNAs have been demonstrated to be critical regulators of the immune responses. While, the miRNA-mediate the detail regulatory mechanism response is still not clear in fish species. In this research, the regulation of miRNA to the NF-κB signaling through decreasing the target gene mRNAs was discussed in miiuy croaker. We first used the bioinformatics predicted miR-144 has a direct negative regulatory affect on IL1ß in miiuy croaker, further the luciferase assays were used to probe the functions of miR-144. The overexpression of miR-144 mimics and pre-miR-144 plasmid all showed the dose-dependent pattern on IL1ß. Moreover, the inhibition of luciferase activity was attenuated after co-transfected with miR-144 inhibitors. In addition, we observed that the miR-144 could negative regulate to the nuclear factor kappaB (NF-κB) signaling in miiuy croaker by targeting IL1ß. In conclusion, our studies on miR-144 will enlarge knowledge of its functions in regulation of immune response, further provide a new insight to research on the immune regulation mechanism in teleost fish.


Subject(s)
Fish Proteins/genetics , Interleukin-1beta/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Perciformes/immunology , Animals , Fish Proteins/immunology , Fish Proteins/metabolism , Interleukin-1beta/immunology , MicroRNAs/immunology , NF-kappa B/immunology , Perciformes/genetics , Perciformes/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
14.
J Immunol ; 201(3): 982-994, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29967101

ABSTRACT

Viral infection induces type I IFN production, which plays critical roles in orchestrating the antiviral defense by inducing direct antiviral activities. To establish a persistent infection, viruses have evolved numerous strategies to specifically interfere with IFN production or its downstream mediators, thereby evading the immune responses. MicroRNAs (miRNAs) are a family of small noncoding RNAs that posttranscriptionally regulate the expressions of specific target genes. Although accumulating evidence demonstrates that miRNAs play vital roles in regulating viral infection, miRNAs that target intracellular sensors and adaptors of innate immunity have not been fully uncovered. In this paper, we identify fish miR-210 as a robust regulator involved in regulating virus-host interactions. We found that rhabdovirus significantly upregulated the expression of fish miR-210. Inducible miR-210 modulates virus-triggered type I IFN and inflammatory cytokine production by targeting stimulator of IFN genes (STING), thereby promoting viral replication. Furthermore, we demonstrated that miR-210 regulates innate immune response through NF-κB, IFN regulatory factor 3, and JAK/STAT signaling pathways. The collective findings indicate that inducible miR-210 plays a regulatory role in virus-host interactions through STING-mediated singling pathway by targeting STING.


Subject(s)
Fishes/immunology , Immunity, Innate/immunology , Membrane Proteins/metabolism , MicroRNAs/immunology , Rhabdoviridae/immunology , Signal Transduction/immunology , Animals , Cell Line , Cell Line, Tumor , Gene Targeting/methods , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Up-Regulation/immunology , Virus Replication/immunology
15.
Front Immunol ; 9: 1413, 2018.
Article in English | MEDLINE | ID: mdl-30013548

ABSTRACT

Lipopolysaccharide (LPS) is the major component of the outer membrane of Gram-negative bacteria. This molecule can induce strong immune response and various biological effects. In mammals, TLR4 can recognize LPS and induce inflammatory response. However, the innate receptor in fish for recognizing LPS remains ambiguous. LPS can invade the cytoplasm via outer membrane vesicles produced by Gram-negative bacteria and could be detected by intracellular receptor caspase-11 in mammals, so, there may also exist the intracellular receptors that can recognize LPS in fish. NOD1 is a member of NOD-like receptors family and can recognize the iE-DAP in the cytoplasm in mammals. In fish, NOD1 can also respond to infection of Gram-negative bacteria and may play an important role in the identification of bacterial components. In this study, to study whether NOD1 is a recognition receptor for LPS, we detected the expression of NOD1 and several cytokines at transcript levels to determine whether LPS can induce inflammatory response in teleost fish and NOD1 can respond to LPS. Then, we perform the binding analysis between NOD1 and ultrapure LPS by using Streptavidin pulldown assay and enzyme-linked immunosorbent assay to prove that NOD1 can be combined with LPS, and using dual luciferase reporter gene assay to verify the signal pathways activated by NOD1. Next, through cell viability analysis, we proved that LPS-induced cytotoxicity can be mediated by NOD1 in fish. The results showed that NOD1 can identify LPS and activate the NF-κB signal pathway by recruiting RIPK2 and then promoting the expression of inflammatory cytokines to induce the resistance of organism against bacterial infection.

16.
Dev Comp Immunol ; 87: 171-175, 2018 10.
Article in English | MEDLINE | ID: mdl-29935287

ABSTRACT

Pattern recognition receptors can recognize pathogens, and then cells are induced to produce pro-inflammatory cytokines and interferon by multiple signaling pathways. Nevertheless, excessive inflammation disrupts immune homeostasis, thereby inducing autoimmune and inflammatory diseases. Thus, the regulation of immune responses is extremely important for host to keep homeostasis. In this study, we found that miR-19a plays a negative regulator in MyD88-mediated NF-κB signaling pathway by targeting MyD88 in miiuy croaker. Furthermore, over-expression of miR-19a in macrophages suppresses the expression of MyD88 and its downstream signaling genes of IRAK1, IRAK4 and TRAF6, whereas, the inhibitor of miR-19a has opposite effect. This study can increase our knowledge and help us to furthermore understand miRNAs regulatory mechanism in teleost fish.


Subject(s)
Fish Proteins/genetics , Gene Expression Regulation , MicroRNAs/genetics , Myeloid Differentiation Factor 88/genetics , Perciformes/genetics , Signal Transduction/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cells, Cultured , HEK293 Cells , Humans , Macrophages/metabolism , NF-kappa B/metabolism , Sequence Homology, Nucleic Acid
17.
Infect Immun ; 86(6)2018 06.
Article in English | MEDLINE | ID: mdl-29632247

ABSTRACT

Inflammation is the host self-protection mechanism to eliminate pathogen invasion. The excessive inflammatory response can result in uncontrolled inflammation, autoimmune diseases, or pathogen dissemination. Recent studies have widely shown that microRNAs (miRNAs) contribute to the regulation of inflammation in mammals by repressing gene expression at the posttranscriptional level. However, the miRNA-mediated mechanism in the inflammatory response in fish remains hazy. In the present study, the regulatory mechanism of the miR-216a-mediated inflammatory response in teleost fish was addressed. We found that the expression of miR-216a could be significantly upregulated in the miiuy croaker after challenge with Vibrio anguillarum and lipopolysaccharide. Bioinformatics predictions demonstrated a potential binding site of miR-216a in the 3' untranslated region of the p65 gene, and the result was further confirmed by luciferase assay. Moreover, both the mRNA and protein levels of p65 in macrophages were downregulated by miR-216a. Deletion mutant analysis of the miR-216a promoter showed that the Ap1 and Sp1 transcription factor binding sites are indispensable for the transcription of miR-216a. Further study revealed that overexpression of miR-216a suppresses inflammatory cytokine expression and negatively regulates NF-κB signaling, which inhibit an excessive inflammatory response. The collective results indicate that miR-216a plays a role as a negative regulator involved in modulating the bacterium-induced inflammatory response.


Subject(s)
Cytokines/metabolism , Inflammation/veterinary , MicroRNAs/metabolism , NF-kappa B/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Perciformes , Animals , Cytokines/genetics , Fish Diseases/metabolism , Gene Expression Regulation/immunology , HEK293 Cells , HeLa Cells , Humans , Inflammation/metabolism , Lipopolysaccharides/toxicity , MicroRNAs/genetics , NF-kappa B/genetics , Nucleocytoplasmic Transport Proteins/genetics , RNA Interference
18.
J Biol Chem ; 293(4): 1386-1396, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29242191

ABSTRACT

Innate immune responses are the first defense against pathogenic invaders. Activation and termination of these immune responses are regulated by several mechanisms. MicroRNAs (miRNAs), a group of small non-coding RNAs, have been implicated in the regulation of a spectrum of both physiological and pathological conditions, including immune responses. Although the immune regulatory miRNA networks in higher vertebrates have been well described, regulation of these responses in fish species is poorly understood. In the present study, we investigated the role of the miRNA miR-203 involved in inflammatory responses in miiuy croaker (Miichthys miiuy). We found that the Gram-negative bacterium Vibrio anguillarum and lipopolysaccharide significantly up-regulated host miR-203 expression. The increased miR-203 expression suppressed the production of inflammatory cytokines and thereby prevented mounting of a full immune response. Mechanistically, we identified and validated IL-1 receptor-associated kinase 4 (IRAK4) as a target of miR-203. We observed that miR-203 post-transcriptionally controls IRAK4 expression and thereby inhibits the activation of nuclear factor κB (NF-κB) signaling. In summary, our findings reveal that miR-203 in fish is a critical suppressor of innate immune responses to bacterial infection by suppressing a feedback to IRAK4-NF-κB-mediated signaling.


Subject(s)
Fish Proteins/immunology , Interleukin-1 Receptor-Associated Kinases/immunology , MicroRNAs/immunology , Perciformes/immunology , Signal Transduction/immunology , Vibrio Infections/immunology , Vibrio/immunology , Animals , Perciformes/microbiology
19.
BMC Genomics ; 19(Suppl 10): 935, 2018 Dec 31.
Article in English | MEDLINE | ID: mdl-30598105

ABSTRACT

BACKGROUND: Soil salinity is one of the primary causes of yield decline in rice. Pokkali (Pok) is a highly salt-tolerant landrace, whereas IR29 is a salt-sensitive but widely cultivated genotype. Comparative analysis of these genotypes may offer a better understanding of the salinity tolerance mechanisms in rice. Although most stress-responsive genes are regulated at the transcriptional level, in many cases, changes at the transcriptional level are not always accompanied with the changes in protein abundance, which suggests that the transcriptome needs to be studied in conjunction with the proteome to link the phenotype of stress tolerance or sensitivity. Published reports have largely underscored the importance of transcriptional regulation during salt stress in these genotypes, but the regulation at the translational level has been rarely studied. Using RNA-Seq, we simultaneously analyzed the transcriptome and translatome from control and salt-exposed Pok and IR29 seedlings to unravel molecular insights into gene regulatory mechanisms that differ between these genotypes. RESULTS: Clear differences were evident at both transcriptional and translational levels between the two genotypes even under the control condition. In response to salt stress, 57 differentially expressed genes (DEGs) were commonly upregulated at both transcriptional and translational levels in both genotypes; the overall number of up/downregulated DEGs in IR29 was comparable at both transcriptional and translational levels, whereas in Pok, the number of upregulated DEGs was considerably higher at the translational level (544 DEGs) than at the transcriptional level (219 DEGs); in contrast, the number of downregulated DEGs (58) was significantly less at the translational level than at the transcriptional level (397 DEGs). These results imply that Pok stabilizes mRNAs and also efficiently loads mRNAs onto polysomes for translation during salt stress. CONCLUSION: Under salt stress, Pok is more efficient in maintaining cell wall integrity, detoxifying reactive oxygen species (ROS), translocating molecules and maintaining photosynthesis. The present study confirmed the known salt stress-associated genes and also identified a number of putative new salt-responsive genes. Most importantly, the study revealed that the translational regulation under salinity plays an important role in salt-tolerant Pok, but such regulation was less evident in the salt-sensitive IR29.


Subject(s)
Gene Expression Profiling , Genotype , Oryza/genetics , Oryza/physiology , Protein Biosynthesis , Salt Tolerance/genetics , Gene Ontology , Oryza/metabolism , Plant Proteins/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism
20.
J Virol ; 92(2)2018 01 15.
Article in English | MEDLINE | ID: mdl-29093090

ABSTRACT

Effectively recognizing invading viruses and subsequently inducing innate antiviral immunity are essential for host antiviral defense. Although these processes are closely regulated by the host to maintain immune balance, viruses have evolved the ability to downregulate or upregulate these processes for their survival. MicroRNAs (miRNAs) are a family of small noncoding RNAs that play vital roles in modulating host immune response. Accumulating evidence demonstrates that host miRNAs as mediators are involved in regulating viral replication and host antiviral immunity in mammals. However, the underlying regulatory mechanisms in fish species are still poorly understood. Here, we found that rhabdovirus infection significantly upregulated host miR-3570 expression in miiuy croaker macrophages. Induced miR-3570 negatively modulated RNA virus-triggered type I interferon (IFN) and antiviral gene production, thus facilitating viral replication. Furthermore, miR-3570 was found to target and posttranscriptionally downregulate mitochondrial antiviral signaling protein (MAVS), which functions as a platform for innate antiviral signal transduction. Moreover, we demonstrated that miR-3570 suppressed the expression of MAVS, thereby inhibiting MAVS-mediated NF-κB and IRF3 signaling. The collective results demonstrated a novel regulation mechanism of MAVS-mediated immunity during RNA viral infection by miRNA.IMPORTANCE RNA viral infection could upregulate host miR-3570 expression in miiuy croaker macrophages. Induced miR-3570 negatively modulates RNA virus-triggered type I IFN and antiviral gene production, thus facilitating viral replication. Remarkably, miR-3570 could target and inhibit MAVS expression, which thus modulates MAVS-mediated NF-κB and IRF3 signaling. The collective results of this study suggest a novel regulation mechanism of MAVS-mediated immunity during RNA viral infection by miR-3570. Thus, a novel mechanism for virus evasion in fish is proposed.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , MicroRNAs/genetics , RNA Interference , Rhabdoviridae/immunology , 3' Untranslated Regions , Animals , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/virology , Gene Knockdown Techniques , Humans , Models, Biological , RNA Processing, Post-Transcriptional , Rhabdoviridae Infections/veterinary , Signal Transduction , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL