1
|
Niu J, Li G, Liu Y, Wan C, Li Y, Dai Y, Hu H, Peng L, Fang R, Ye C. The important role of TLR2/MyD88/JNK in regulating the pathogenesis and inflammation induced by pseudorabies virus in mice. Vet Microbiol 2025; 304:110496. [PMID: 40156971 DOI: 10.1016/j.vetmic.2025.110496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
The host innate immune response plays a critical role in regulating and controlling viral infections by releasing inflammatory cytokines. Pseudorabies virus (PRV), a swine alphaherpesvirus, can cause severe encephalitis in piglets and various non-natural hosts. Previous studies demonstrated that PRV infection induced the significant elevation of pro-inflammatory cytokines levels and lethal inflammatory response in the mouse model. However, the underlying mechanisms responsible for activation and production of pro-inflammatory cytokines during PRV infection remain to be fully elucidated. In this study, we confirmed that PRV induced significant inflammatory response in C57BL/6 mice during its acute infection. Furthermore, TLR2/MyD88 axis was shown to be associated with the pathogenesis of PRV in mice. Specifically, TLR2/MyD88 axis was required for PRV-induced activation of NF-κB pathway and the subsequent pro-inflammatory cytokines expression. Meanwhile, MAPK/JNK and PI3K/Akt signaling pathways were also activated by TLR2/MyD88 axis and involved in regulating pro-inflammatory cytokines expression induced by PRV infection, respectively. Notably, administration of the JNK inhibitor (SP600125) could reduce clinical symptoms, alleviate pathological damage and prolong survival time of mice infected by PRV. Overall, this study strengthens our understanding upon the mechanism of host inflammatory response induced by PRV, and suggests that JNK signaling may act as a therapeutic target in controlling of PRV infection.
Collapse
Affiliation(s)
- Jingyi Niu
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Gang Li
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Yiyu Liu
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Chao Wan
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Yixuan Li
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Yu Dai
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Haixia Hu
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Lianci Peng
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Rendong Fang
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China.
| | - Chao Ye
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China.
| |
Collapse
|
2
|
Gu R, Zhao F, Li Q, Hou L, Liu G, Sun X, Du J, Fang B. Hesperetin Inhibits Porcine Reproductive and Respiratory Syndrome Virus Replication by Downregulating the P38/JUN/FOS Pathway In Vitro. Microorganisms 2025; 13:450. [PMID: 40005815 PMCID: PMC11858564 DOI: 10.3390/microorganisms13020450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/02/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a persistent pathogen that causes significant economic losses to the global swine industry. Commercial vaccines provide only partial protection, and no effective therapeutic treatments are currently available. In this study, we demonstrate that hesperetin (Hst), a flavonoid glycoside derived from orange and tangerine peels, inhibits PRRSV replication in a dose-dependent manner in Marc-145 and PAMs cells. Transcriptome analysis further reveals that the anti-PRRSV effects of Hst are associated with the suppression of the P38 MAPK pathway, as Hst significantly downregulates key genes, including NRA41, JUN, FOS, and DUSP1. Subsequent investigations show that Hst inhibits PRRSV replication by downregulating the P38/JUN/FOS signaling cascade. These findings offer valuable insights for the development of novel preventive and therapeutic strategies against PRRSV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Binghu Fang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (R.G.); (F.Z.); (Q.L.); (L.H.); (G.L.); (X.S.); (J.D.)
| |
Collapse
|
3
|
Du K, Xia Y, Wu Q, Yin M, Zhao H, Chen XW. Analysis of whole transcriptome reveals the immune response to porcine reproductive and respiratory syndrome virus infection and tylvalosin tartrate treatment in the porcine alveolar macrophages. Front Immunol 2025; 15:1506371. [PMID: 39872536 PMCID: PMC11769836 DOI: 10.3389/fimmu.2024.1506371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Porcine reproductive and respiratory syndrome virus (PRRSV) is a major pathogen that has caused severe economic losses in the swine industry. Screening key host immune-related genetic factors in the porcine alveolar macrophages (PAMs) is critical to improve the anti-virial ability in pigs. Methods In this study, an in vivo model was set to evaluate the anti-PRRSV effect of tylvalosin tartrates. Then, strand-specific RNA-sequencing (ssRNA-seq) and miRNA-sequencing (miRNA-seq) were carried out to profile the whole transcriptome of PAMs in the negative control, PRRSV-infected, and tylvalosin tartrates-treatment group. Results The ssRNA-seq identified 11740 long non-coding RNAs in PAMs. Based on our attention mechanism-improved graph convolutional network, 41.07% and 28.59% lncRNAs were predicted to be located in the nucleus and cytoplasm, respectively. The miRNA-seq revealed that tylvalosin tartrates-enhanced miRNAs might play roles in regulating angiogenesis and innate immune-related functions, and it rescued the expression of three anti-inflammation miRNAs (ssc-miR-30a-5p, ssc-miR-218-5p, and ssc-miR-218) that were downregulated due to PRRSV infection. The cytoplasmic lncRNAs enhanced by tylvalosin tartrates might form ceRNA networks with miRNAs to regulate PAM chemotaxis. While cytoplasmic lncRNAs that were rescued by tylvalosin tartrates might protect PAMs via efferocytosis-related ceRNA networks. On the other hand, the tylvalosin tartrates-rescued nuclear lncRNAs might negatively regulate T cell apoptosis and bind to key anti-inflammation factor IL37 to protect the lungs by cis- and trans-regulation. Conclusions Our data provides a catalog of key non-coding RNAs in response to PRRSV and tylvalosin tartrates and might enrich the genetic basis for future PRRSV prevention and control.
Collapse
Affiliation(s)
| | | | | | | | | | - Xi-wen Chen
- Animal Disease Prevention and Control and Healthy Breeding Engineering Technology Research Centre, Mianyang Normal University, Mianyang, China
| |
Collapse
|
4
|
Chen D, Xu S, Jiang R, Guo Y, Yang X, Zhang Y, Zhou L, Ge X, Han J, Guo X, Yang H. IL-1β induced by PRRSV co-infection inhibited CSFV C-strain proliferation via the TLR4/NF-κB/MAPK pathways and the NLRP3 inflammasome. Vet Microbiol 2022; 273:109513. [DOI: 10.1016/j.vetmic.2022.109513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 12/24/2022]
|
5
|
Trypsin enhances SARS-CoV-2 infection by facilitating viral entry. Arch Virol 2022; 167:441-458. [PMID: 35079901 PMCID: PMC8789370 DOI: 10.1007/s00705-021-05343-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/10/2021] [Indexed: 12/28/2022]
Abstract
Coronaviruses infect cells by cytoplasmic or endosomal membrane fusion, driven by the spike (S) protein, which must be primed by proteolytic cleavage at the S1/S2 furin cleavage site (FCS) and the S2′ site by cellular proteases. Exogenous trypsin as a medium additive facilitates isolation and propagation of several coronaviruses in vitro. Here, we show that trypsin enhances severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in cultured cells and that SARS-CoV-2 enters cells via either a non-endosomal or an endosomal fusion pathway, depending on the presence of trypsin. Interestingly, trypsin enabled viral entry at the cell surface and led to more efficient infection than trypsin-independent endosomal entry, suggesting that trypsin production in the target organs may trigger a high level of replication of SARS-CoV-2 and cause severe tissue injury. Extensive syncytium formation and enhanced growth kinetics were observed only in the presence of exogenous trypsin when cell-adapted SARS-CoV-2 strains were tested. During 50 serial passages without the addition of trypsin, a specific R685S mutation occurred in the S1/S2 FCS (681PRRAR685) that was completely conserved but accompanied by several mutations in the S2 fusion subunit in the presence of trypsin. These findings demonstrate that the S1/S2 FCS is essential for proteolytic priming of the S protein and fusion activity for SARS-CoV-2 entry but not for viral replication. Our data can potentially contribute to the improvement of SARS-CoV-2 production for the development of vaccines or antivirals and motivate further investigations into the explicit functions of cell-adaptation-related genetic drift in SARS-CoV-2 pathogenesis.
Collapse
|
6
|
The Roles of c-Jun N-Terminal Kinase (JNK) in Infectious Diseases. Int J Mol Sci 2021; 22:ijms22179640. [PMID: 34502556 PMCID: PMC8431791 DOI: 10.3390/ijms22179640] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 01/12/2023] Open
Abstract
c-Jun N-terminal kinases (JNKs) are among the most crucial mitogen-activated protein kinases (MAPKs) and regulate various cellular processes, including cell proliferation, apoptosis, autophagy, and inflammation. Microbes heavily rely on cellular signaling pathways for their effective replication; hence, JNKs may play important roles in infectious diseases. In this review, we describe the basic signaling properties of MAPKs and JNKs in apoptosis, autophagy, and inflammasome activation. Furthermore, we discuss the roles of JNKs in various infectious diseases induced by viruses, bacteria, fungi, and parasites, as well as their potential to serve as targets for the development of therapeutic agents for infectious diseases. We expect this review to expand our understanding of the JNK signaling pathway’s role in infectious diseases and provide important clues for the prevention and treatment of infectious diseases.
Collapse
|
7
|
Jeon JH, Lee C. Stress-activated protein kinases are involved in the replication of porcine deltacoronavirus. Virology 2021; 559:196-209. [PMID: 33964685 DOI: 10.1016/j.virol.2021.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 11/30/2022]
Abstract
This study was conducted to examine the role of stress-activated protein kinases (SAPKs), including c-Jun NH2-terminal kinases (JNK1/2) and p38 mitogen-activated protein kinase (MAPK), in porcine deltacoronavirus (PDCoV) infection. Results demonstrated the activation of JNK1/2 and p38 MAPK in PDCoV-infected cells, which occurred concomitant with viral biosynthesis and irrespective of cell type. Pharmacological inhibition or knockdown of either SAPK significantly attenuated PDCoV replication, whereas addition of a signaling activator augmented virus infectivity. Moreover, pharmacological inhibition of JNK1/2 or p38 MAPK activation was innocuous to viral entry but significantly detrimental to post uncoating stages of the replication cycle. Remarkably, cytokine gene expression in PDCoV-infected IPEC-J2 cells was modified by inhibiting the activation of either SAPK. Collectively, these data indicate that JNK1/2 and p38 MAPK signaling pathways contribute to viral biosynthesis and regulate immune responses, thereby favoring the replication of PDCoV.
Collapse
Affiliation(s)
- Ji Hyun Jeon
- Animal Virology Laboratory, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 702-701, Republic of Korea
| | - Changhee Lee
- Animal Virology Laboratory, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 702-701, Republic of Korea.
| |
Collapse
|
8
|
Molecular and Cellular Mechanisms for PRRSV Pathogenesis and Host Response to Infection. Virus Res 2020; 286:197980. [PMID: 32311386 PMCID: PMC7165118 DOI: 10.1016/j.virusres.2020.197980] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022]
Abstract
PRRSV has evolved to arm with various strategies to modify host antiviral response. Viral modulation of homeostatic cellular processes provides favorable conditions for PRRSV survival during infection. PRRSV modulation of cellular processes includes pathways for interferons, apoptosis, microRNAs, cytokines, autophagy, and viral genome recombination.
Porcine reproductive and respiratory syndrome virus (PRRSV) has caused tremendous amounts of economic losses to the swine industry for more than three decades, but its control is still unsatisfactory. A significant amount of information is available for host cell-virus interactions during infection, and it is evident that PRRSV has evolved to equip various strategies to disrupt the host antiviral system and provide favorable conditions for survival. The current study reviews viral strategies for modulations of cellular processes including innate immunity, apoptosis, microRNAs, inflammatory cytokines, and other cellular pathways.
Collapse
|
9
|
Fan L. Signaling pathways involved in regulating apoptosis induction in host cells upon PRRSV infection. Virus Genes 2019; 55:433-439. [PMID: 31004277 DOI: 10.1007/s11262-019-01665-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/14/2019] [Indexed: 12/11/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is the etiologic agent of porcine reproductive and respiratory syndrome (PRRS), a devastating disease of swine that poses a serious threat to the swine industry worldwide. The induction of apoptosis in host cells is suggested to be the key cellular mechanism that contributes to the pathogenesis of PRRS. Various signaling pathways have been identified to be involved in regulating PRRSV-induced apoptosis. In this review, we summarize the potential signaling pathways that contribute to PRRSV-induced apoptosis, and propose the issues that need to be addressed in future studies for a better understanding of the molecular basis underlying the pathogenesis of PRRS.
Collapse
Affiliation(s)
- Lihong Fan
- Department of Preventive Medicine, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China.
| |
Collapse
|
10
|
Transcriptional profiles of PBMCs from pigs infected with three genetically diverse porcine reproductive and respiratory syndrome virus strains. Mol Biol Rep 2018; 45:675-688. [PMID: 29882085 PMCID: PMC6156768 DOI: 10.1007/s11033-018-4204-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 05/31/2018] [Indexed: 01/23/2023]
Abstract
Porcine reproductive and respiratory syndrome virus is the cause of reproductive failure in sows and respiratory disease in young pigs, which has been considered as one of the most costly diseases to the worldwide pig industry for almost 30 years. This study used microarray-based transcriptomic analysis of PBMCs from experimentally infected pigs to explore the patterns of immune dysregulation after infection with two East European PRRSV strains from subtype 2 (BOR and ILI) in comparison to a Danish subtype 1 strain (DAN). Transcriptional profiles were determined at day 7 post infection in three tested groups of pigs and analysed in comparison with the expression profile of control group. Microarray analysis revealed differential regulation (> 1.5-fold change) of 4253 and 7335 genes in groups infected with BOR and ILI strains, respectively, and of 12518 genes in pigs infected with Danish strain. Subtype 2 PRRSV strains showed greater induction of many genes, especially those involved in innate immunity, such as interferon stimulated antiviral genes and inflammatory markers. Functional analysis of the microarray data revealed a significant up-regulation of genes involved in processes such as acute phase response, granulocyte and agranulocyte adhesion and diapedesis, as well as down-regulation of genes enrolled in pathways engaged in protein synthesis, cell division, as well as B and T cell signaling. This study provided an insight into the host response to three different PRRSV strains at a molecular level and demonstrated variability between strains of different pathogenicity level.
Collapse
|
11
|
Cellular cholesterol is required for porcine nidovirus infection. Arch Virol 2017; 162:3753-3767. [PMID: 28884395 PMCID: PMC7086867 DOI: 10.1007/s00705-017-3545-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/12/2017] [Indexed: 12/14/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) and porcine epidemic diarrhea virus (PEDV) are porcine nidoviruses that are considered emerging and re-emerging viral pathogens of pigs that pose a significant economic threat to the global pork industry. Although cholesterol is known to affect the replication of a broad range of viruses in vitro, its significance and role in porcine nidovirus infection remains to be elucidated. Therefore, the present study was conducted to determine whether cellular or/and viral cholesterol levels play a role in porcine nidovirus infection. Our results showed that depletion of cellular cholesterol by treating cells with methyl-β-cyclodextrin (MβCD) dose-dependently suppressed the replication of both nidoviruses. Conversely, cholesterol depletion from the viral envelope had no inhibitory effect on porcine nidovirus production. The addition of exogenous cholesterol to MβCD-treated cells moderately restored the infectivity of porcine nidoviruses, indicating that the presence of cholesterol in the target cell membrane is critical for viral replication. The antiviral activity of MβCD on porcine nidovirus infection was found to be predominantly exerted when used as a treatment pre-infection or prior to the viral entry process. Furthermore, pharmacological sequestration of cellular cholesterol efficiently blocked both virus attachment and internalization and, accordingly, markedly affected subsequent post-entry steps of the replication cycle, including viral RNA and protein biosynthesis and progeny virus production. Taken together, our data indicate that cell membrane cholesterol is required for porcine nidovirus entry into cells, and pharmacological drugs that hamper cholesterol-dependent virus entry may have antiviral potential against porcine nidoviruses.
Collapse
|
12
|
Porcine reproductive and respiratory syndrome virus (PRRSV) up-regulates IL-8 expression through TAK-1/JNK/AP-1 pathways. Virology 2017; 506:64-72. [PMID: 28347884 PMCID: PMC7111726 DOI: 10.1016/j.virol.2017.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/08/2017] [Accepted: 03/21/2017] [Indexed: 12/21/2022]
Abstract
The acute phase of respiratory distress caused by porcine reproductive and respiratory syndrome virus (PRRSV) is likely a consequence of the release of inflammatory cytokines in the lung. IL-8, the main chemokine and activator of neutrophils, might be related to the lung injury upon PRRSV infection. In this study, we showed that PRRSV induced IL-8 expression in vivo and in vitro. Subsequently, we demonstrated that JNK and NF-κB pathways were activated upon PRRSV infection and required for the enhancement of IL-8 expression. We further verified that PRRSV-activated TAK-1 was essential for the activation of JNK and NF-κB pathways and IL-8 expression. Moreover, we revealed an AP-1 binding motif in the cloned porcine IL-8 (pIL-8) promoter, and deletion of this motif abolished the pIL-8 promoter activity. Finally, we found that the JNK-activated AP-1 subunit c-Jun was critical for the up-regulation of IL-8 expression by PRRSV. These data suggest that PRRSV-induced IL-8 production is likely through the TAK-1/JNK/AP-1 pathways. PRRSV infection induces IL-8 expression in vitro and in vivo. PRRSV up-regulates IL-8 expression through TAK-1/JNK/AP-1 pathways. AP-1 element in porcine IL-8 promoter is essential for PRRSV induced IL-8 expression.
Collapse
|
13
|
Porcine Reproductive and Respiratory Syndrome Virus nsp1α Inhibits NF-κB Activation by Targeting the Linear Ubiquitin Chain Assembly Complex. J Virol 2017; 91:JVI.01911-16. [PMID: 27881655 DOI: 10.1128/jvi.01911-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/17/2016] [Indexed: 01/18/2023] Open
Abstract
Linear ubiquitination, a newly discovered posttranslational modification, is catalyzed by the linear ubiquitin chain assembly complex (LUBAC), which is composed of three subunits: one catalytic subunit HOIP and two accessory molecules, HOIL-1L and SHARPIN. Accumulating evidence suggests that linear ubiquitination plays a crucial role in innate immune signaling and especially in the activation of the NF-κB pathway by conjugating linear polyubiquitin chains to NF-κB essential modulator (NEMO, also called IKKγ), the regulatory subunit of the IKK complex. Porcine reproductive and respiratory syndrome virus (PRRSV), an Arterivirus that has devastated the swine industry worldwide, is an ideal model to study the host's disordered inflammatory responses after viral infection. Here, we found that LUBAC-induced NF-κB and proinflammatory cytokine expression can be inhibited in the early phase of PRRSV infection. Screening the PRRSV-encoded proteins showed that nonstructural protein 1α (nsp1α) suppresses LUBAC-mediated NF-κB activation and its CTE domain is required for the inhibition. Mechanistically, nsp1α binds to HOIP/HOIL-1L and impairs the interaction between HOIP and SHARPIN, thus reducing the LUBAC-dependent linear ubiquitination of NEMO. Moreover, PRRSV infection also blocks LUBAC complex formation and NEMO linear-ubiquitination, the important step for transducing NF-κB signaling. This unexpected finding demonstrates a previously unrecognized role of PRRSV nsp1α in modulating LUBAC signaling and explains an additional mechanism of immune modulation by PRRSV. IMPORTANCE Porcine reproductive and respiratory syndrome (PRRS) is one of the most important veterinary infectious diseases in countries with intensive swine industries. PRRS virus (PRRSV) infection usually suppresses proinflammatory cytokine expression in the early stage of infection, whereas it induces an inflammatory storm in the late stage. However, precisely how the virus is capable of doing so remains obscure. In this study, we found that by blocking the interaction of its catalytic subunit HOIP and accessory molecule SHARPIN, PRRSV can suppress NF-κB signal transduction in the early stage of infection. Our findings not only reveal a novel mechanism evolved by PRRSV to regulate inflammatory responses but also highlight the important role of linear ubiquitination modification during virus infection.
Collapse
|
14
|
Diseases Primarily Affecting the Reproductive System. Vet Med (Auckl) 2017. [PMCID: PMC7150237 DOI: 10.1016/b978-0-7020-5246-0.00018-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Lee C, Kim Y, Jeon JH. JNK and p38 mitogen-activated protein kinase pathways contribute to porcine epidemic diarrhea virus infection. Virus Res 2016; 222:1-12. [PMID: 27215486 PMCID: PMC7114560 DOI: 10.1016/j.virusres.2016.05.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/16/2022]
Abstract
PEDV infection activated p38 MAPK and JNK1/2 in vitro. UV-inactivated virus failed to induce p38 MAPK and JNK1/2 activation. Pharmacological inhibition of p38 MAPK or JNK activation impaired PEDV replication. SAPK cascades do not affect the apoptosis pathway during PEDV infection. PEDV exploits the p38 MAPK and JNK signaling pathways for optimal replication.
The mitogen-activated protein kinase (MAPK) pathways, which are central building blocks in the intracellular signaling network, are often manipulated by viruses of diverse families to favor their replication. Among the MAPK family, the extracellular signal-regulated kinase (ERK) pathway is known to be modulated during the infection with porcine epidemic diarrhea virus (PEDV); however, involvement of stress-activated protein kinases (SAPKs) comprising p38 MAPK and c-Jun NH2-terminal kinase (JNK) remains to be determined. Therefore, in the present study, we investigated whether activation of p38 MAPK and JNK cascades is required for PEDV replication. Our results showed that PEDV activates p38 MAPK and JNK1/2 up to 24 h post-infection, whereas, thereafter their phosphorylation levels recede to baseline levels or even fall below them. Notably, UV-irradiated inactivated PEDV, which can enter cells but cannot replicate inside them, failed to induce phosphorylation of p38 MAPK and JNK1/2 suggesting that viral biosynthesis is essential for activation of these kinases. Treatment of cells with selective p38 or JNK inhibitors markedly impaired PEDV replication in a dose-dependent manner and these antiviral effects were found to be maximal during the early times of the infection. Furthermore, direct pharmacological inhibition of p38 MAPK or JNK1/2 activation resulted in a significant reduction of viral RNA synthesis, viral protein expression, and progeny release. However, independent treatments with either SAPK inhibitor did not inhibit PEDV-induced apoptotic cell death mediated by activation of mitochondrial apoptosis-inducing factor (AIF) suggesting that SAPKs are irrelevant to the apoptosis pathway during PEDV infection. In summary, our data demonstrated critical roles of the p38 and JNK1/2 signaling pathways in facilitating successful viral infection during the post-entry steps of the PEDV life cycle.
Collapse
Affiliation(s)
- Changhee Lee
- Animal Virology Laboratory, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Republic of Korea.
| | - Youngnam Kim
- Animal Virology Laboratory, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Ji Hyun Jeon
- Animal Virology Laboratory, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Republic of Korea
| |
Collapse
|
16
|
Chen R, Xi L, Huang X, Ma T, Ren H, Ji G. Effect of Jun N-terminal kinase 1 and 2 on the replication of Penicillium marneffei in human macrophages. Microb Pathog 2015; 82:1-6. [PMID: 25792289 DOI: 10.1016/j.micpath.2015.03.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 11/15/2022]
Abstract
Penicillium marneffei (P. marneffei) is a human pathogen which persists in macrophages and threatens the immunocompromised patients. To clarify the mechanisms involved, we evaluated the effect of c-Jun N-terminal kinase 1 and 2 (JNK1/2) on cytokine expression, phagosomal maturation and multiplication of P. marneffei in P. marneffei-stimulated human macrophages. P. marneffei induced the rapid phosphorylation of JNK1/2. Using the specific inhibitor of JNK1/2 (SP600125), we found that the inhibition of JNK1/2 suppressed P. marneffei-induced tumor necrosis factor-α and IL-10 production. In addition, the presence of SP600125 increased phagosomal acidification and maturation and decreased intracellular replication. These data suggest that JNK1/2 may play an important role in promoting the replication of P. marneffei. Our findings further indicate that the pathogen through the JNK1/2 pathway may attenuate the immune response and macrophage antifungal function.
Collapse
Affiliation(s)
- Renqiong Chen
- Department of Dermatology, Lianyungang First People's Hospital, Lianyungang 222002, China
| | - Liyan Xi
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaowen Huang
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tuan Ma
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hong Ren
- Department of Dermatology, Lianyungang First People's Hospital, Lianyungang 222002, China
| | - Guangquan Ji
- Department of Dermatology, Lianyungang First People's Hospital, Lianyungang 222002, China.
| |
Collapse
|
17
|
Han M, Yoo D. Modulation of innate immune signaling by nonstructural protein 1 (nsp1) in the family Arteriviridae. Virus Res 2014; 194:100-9. [PMID: 25262851 PMCID: PMC7114407 DOI: 10.1016/j.virusres.2014.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 12/24/2022]
Abstract
Arteriviruses infect immune cells and may cause persistence in infected hosts. Inefficient induction of pro-inflammatory cytokines and type I IFNs are observed during infection of this group of viruses, suggesting that they may have evolved to escape the host immune surveillance for efficient survival. Recent studies have identified viral proteins regulating the innate immune signaling, and among these, nsp1 (nonstructural protein 1) is the most potent IFN antagonist. For porcine reproductive and respiratory syndrome virus (PRRSV), individual subunits (nsp1α and nsp1β) of nsp1 suppress type I IFN production. In particular, PRRSV-nsp1α degrades CREB (cyclic AMP responsive element binding)-binding protein (CBP), a key component of the IFN enhanceosome, whereas PRRSV-nsp1β degrades karyopherin-α1 which is known to mediate the nuclear import of ISGF3 (interferon-stimulated gene factor 3). All individual subunits of nsp1 of PRRSV, equine arteritis virus (EAV), lactate dehydrogenase-elevating virus (LDV), and simian hemorrhagic fever virus (SHFV) appear to contain IFN suppressive activities. As with PRRSV-nsp1α, CBP degradation is evident by LDV-nsp1α and partly by SHFV-nsp1γ. This review summarizes the biogenesis and the role of individual subunits of nsp1 of arteriviruses for innate immune modulation.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Dongwan Yoo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA.
| |
Collapse
|
18
|
Zhao H, Guo XK, Bi Y, Zhu Y, Feng WH. PKCδ is required for porcine reproductive and respiratory syndrome virus replication. Virology 2014; 468-470:96-103. [PMID: 25155198 DOI: 10.1016/j.virol.2014.07.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 06/17/2014] [Accepted: 07/21/2014] [Indexed: 01/27/2023]
Abstract
Protein kinase C (PKC) that transduces signals to modulate a wide range of cellular functions has been shown to regulate a number of viral infections. Herein, we showed that inhibition of PKC with the PKC inhibitor GF109203X significantly impaired porcine reproductive and respiratory syndrome virus (PRRSV) replication. Inhibition of PKC led to virus yield reduction, which was associated with decreased viral RNA synthesis and lowered virus protein expression. And this inhibitory effect by PKC inhibitor was shown to occur at the early stage of PRRSV infection. Subsequently, we found that PRRSV infection activated PKCδ in PAMs and knockdown of PKCδ by small interfering RNA (siRNA) suppressed PRRSV replication, suggesting that novel PKCδ may play an important factor in PRRSV replication. Taken together, these data imply that PKC is involved in PRRSV infection and beneficial to PRRSV replication, extending our understanding of PRRSV replication.
Collapse
Affiliation(s)
- Haiyan Zhao
- State Key Laboratories of Agrobiotechnology, China; Department of Microbiology and Immunology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Xue-Kun Guo
- State Key Laboratories of Agrobiotechnology, China; Department of Microbiology and Immunology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Yanmin Bi
- State Key Laboratories of Agrobiotechnology, China; Department of Microbiology and Immunology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Yihui Zhu
- College of Biological Science, Hunan Normal University, Changsha 410006, China
| | - Wen-Hai Feng
- State Key Laboratories of Agrobiotechnology, China; Department of Microbiology and Immunology, College of Biological Science, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
19
|
Porcine reproductive and respiratory syndrome virus induces IL-1β production depending on TLR4/MyD88 pathway and NLRP3 inflammasome in primary porcine alveolar macrophages. Mediators Inflamm 2014; 2014:403515. [PMID: 24966466 PMCID: PMC4055429 DOI: 10.1155/2014/403515] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/23/2014] [Accepted: 04/23/2014] [Indexed: 12/17/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is an Arterivirus that has been devastating the swine industry worldwide since the late 1980s. Previous studies have reported that PRRSV infection induced the production of IL-1β. However, the cellular sensors and signaling pathways involved in this process have not been elucidated yet. Here, we studied the mechanisms responsible for the production of IL-1β in response to highly pathogenic PRRSV. Upon PRRSV infection of primary porcine alveolar macrophages, both mRNA expression and secretion of IL-1β were significantly increased in a time- and dose-dependent manner. We also investigated the role of several pattern-recognition receptors and adaptor molecules in this response and showed that the TLR4/MyD88 pathway and its downstream signaling molecules, NF-κB, ERK1/2, and p38 MAPKs, were involved in IL-1β production during PRRSV infection. Treatment with specific inhibitors or siRNA knockdown assays demonstrated that components of the NLRP3 inflammasome were crucial for IL-1β secretion but not for IL-1β mRNA expression. Furthermore, TLR4/MyD88/NF-κB signaling pathway was involved in PRRSV-induced expression of NLRP3 inflammasome components. Together, our results deciphered the pathways leading from recognition of PRRSV to the production and release of IL-1β, providing a deeper knowledge of the mechanisms of PRRSV-induced inflammation responses.
Collapse
|
20
|
Jing H, Fang L, Wang D, Ding Z, Luo R, Chen H, Xiao S. Porcine reproductive and respiratory syndrome virus infection activates NOD2-RIP2 signal pathway in MARC-145 cells. Virology 2014; 458-459:162-71. [PMID: 24928048 DOI: 10.1016/j.virol.2014.04.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 02/12/2014] [Accepted: 04/22/2014] [Indexed: 12/21/2022]
Abstract
Nucleotide-binding oligomerization domains (NOD)-like receptors (NLRs) evolve as a group of germline-encoded receptors that detect cytosolic pathogen-associated molecular patterns. Porcine reproductive and respiratory syndrome virus (PRRSV) is an Arterivirus that has been devastating the swine industry worldwide. By examining the expression kinetics of ten selected NLRs, NOD2 and NLRP3 were found to be continuously up-regulated in PRRSV-infected MARC-145 cells during 48 h of post-infection. Further study revealed that PRRSV infection enhanced the expression and phosphorylation of RIP2. Knockdown of NOD2 and RIP2 by siRNA significantly decreased PRRSV-induced phosphorylation of NF-κB subunit p65, JNK, Erk and p38 MAPK, as well as the expression of IL-6, IL-8, TNF-α, and RANTES in MARC-145 cells. Moreover, increased expression of NOD2 and RIP2 mRNA were observed in alveolar macrophages isolated from PRRSV-challenged piglets at 3, 7 and 10 day post-challenge. Collectively, our results revealed that PRRSV infection activates NOD2-RIP2 signaling pathway to induce pro-inflammatory response.
Collapse
Affiliation(s)
- Huiyuan Jing
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Liurong Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Dang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhen Ding
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shaobo Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
21
|
Johnson JC, Martinez O, Honko AN, Hensley LE, Olinger GG, Basler CF. Pyridinyl imidazole inhibitors of p38 MAP kinase impair viral entry and reduce cytokine induction by Zaire ebolavirus in human dendritic cells. Antiviral Res 2014; 107:102-9. [PMID: 24815087 DOI: 10.1016/j.antiviral.2014.04.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 01/03/2023]
Abstract
Antigen presenting cells (APCs), including macrophages and dendritic cells, are early and sustained targets of Ebola virus (EBOV) infection in vivo. Because EBOV activates mitogen-activated protein kinase (MAPK) signaling upon infection of APCs, we evaluated the effect of pyridinyl imidazole inhibitors of p38 MAPK on EBOV infection of human APCs and EBOV mediated cytokine production from human DCs. The p38 MAPK inhibitors reduced viral replication in PMA-differentiated macrophage-like human THP-1 cells with an IC50 of 4.73μM (SB202190), 8.26μM (p38kinhIII) and 8.21μM (SB203580) and primary human monocyte-derived dendritic cells (MDDCs) with an IC50 of 2.67μM (SB202190). Furthermore, cytokine production from EBOV-treated MDDCs was inhibited in a dose-dependent manner. A control pyridinyl imidazole compound failed to inhibit either EBOV infection or cytokine induction. Using an established EBOV virus-like particle (VLP) entry assay, we demonstrate that inhibitor pretreatment blocked VLP entry suggesting that the inhibitors blocked infection and replication at least in part by blocking EBOV entry. Taken together, our results indicate that pyridinyl imidazole p38 MAPK inhibitors may serve as leads for the development of therapeutics to treat EBOV infection.
Collapse
Affiliation(s)
- Joshua C Johnson
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Osvaldo Martinez
- Dept. of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Anna N Honko
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Lisa E Hensley
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Gene G Olinger
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Christopher F Basler
- Dept. of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
22
|
Differential host cell gene expression and regulation of cell cycle progression by nonstructural protein 11 of porcine reproductive and respiratory syndrome virus. BIOMED RESEARCH INTERNATIONAL 2014; 2014:430508. [PMID: 24719865 PMCID: PMC3955671 DOI: 10.1155/2014/430508] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/07/2014] [Indexed: 02/08/2023]
Abstract
Nonstructural protein 11 (nsp11) of porcine reproductive and respiratory syndrome virus (PRRSV) is a viral endoribonuclease with an unknown function. The regulation of cellular gene expression by nsp11 was examined by RNA microarrays using MARC-nsp11 cells constitutively expressing nsp11. In these cells, the interferon-β, interferon regulatory factor 3, and nuclear factor-κB activities were suppressed compared to those of parental cells, suggesting that nsp11 might serve as a viral interferon antagonist. Differential cellular transcriptome was examined using Affymetrix exon chips representing 28,536 transcripts, and after statistical analyses 66 cellular genes were shown to be upregulated and 104 genes were downregulated by nsp11. These genes were grouped into 5 major signaling pathways according to their functional relations: histone-related, cell cycle and DNA replication, mitogen activated protein kinase signaling, complement, and ubiquitin-proteasome pathways. Of these, the modulation of cell cycle by nsp11 was further investigated since many of the regulated genes fell in this particular pathway. Flow cytometry showed that nsp11 caused the delay of cell cycle progression at the S phase and the BrdU staining confirmed the cell cycle arrest in nsp11-expressing cells. The study provides insights into the understanding of specific cellular responses to nsp11 during PRRSV infection.
Collapse
|
23
|
Luo R, Fang L, Jin H, Wang D, An K, Xu N, Chen H, Xiao S. Label-Free Quantitative Phosphoproteomic Analysis Reveals Differentially Regulated Proteins and Pathway in PRRSV-Infected Pulmonary Alveolar Macrophages. J Proteome Res 2014; 13:1270-80. [DOI: 10.1021/pr400852d] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Rui Luo
- Division
of Animal Infectious Diseases, State Key Laboratory of Agricultural
Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No. 1 Shi-zi-shan Street, Wuhan, Hubei 430070, China
| | - Liurong Fang
- Division
of Animal Infectious Diseases, State Key Laboratory of Agricultural
Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No. 1 Shi-zi-shan Street, Wuhan, Hubei 430070, China
| | - Hui Jin
- Division
of Animal Infectious Diseases, State Key Laboratory of Agricultural
Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No. 1 Shi-zi-shan Street, Wuhan, Hubei 430070, China
| | - Dang Wang
- Division
of Animal Infectious Diseases, State Key Laboratory of Agricultural
Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No. 1 Shi-zi-shan Street, Wuhan, Hubei 430070, China
| | - Kang An
- Division
of Animal Infectious Diseases, State Key Laboratory of Agricultural
Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No. 1 Shi-zi-shan Street, Wuhan, Hubei 430070, China
| | - Ningzhi Xu
- Laboratory
of Cell and Molecular Biology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dong
Dan San Tiao, Beijing 100005, China
| | - Huanchun Chen
- Division
of Animal Infectious Diseases, State Key Laboratory of Agricultural
Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No. 1 Shi-zi-shan Street, Wuhan, Hubei 430070, China
| | - Shaobo Xiao
- Division
of Animal Infectious Diseases, State Key Laboratory of Agricultural
Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No. 1 Shi-zi-shan Street, Wuhan, Hubei 430070, China
| |
Collapse
|
24
|
Wang LL, Huang H, Zhang CR, Xia J, Liu SS, Wang XW. Cloning and functional characterization of c-Jun NH2-terminal kinase from the Mediterranean species of the Whitefly Bemisia tabaci complex. Int J Mol Sci 2013; 14:13433-46. [PMID: 23807503 PMCID: PMC3742195 DOI: 10.3390/ijms140713433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 05/29/2013] [Accepted: 06/18/2013] [Indexed: 12/19/2022] Open
Abstract
c-Jun NH2-terminal kinase (JNK) signaling is a highly conserved pathway that controls gene transcription in response to a wide variety of biological and environmental stresses. In this study, a JNK from the invasive Mediterranean (MED) species of the whitefly Bemisia tabaci complex was cloned and characterized. The full-length JNK cDNA of MED consists of 1565 bp, with an 1176 bp open reading frame encoding 392 amino acids. Comparison of JNK amino acid sequences among different species showed that the sequences of JNKs are highly conserved. To reveal its biological function, the gene expression and functional activation of JNK were analyzed during various stress conditions. Quantitative RT-PCR analysis showed that the relative expression level of JNK remained hardly unchanged when the insects were transferred from cotton (a suitable host plant) to tobacco (an unsuitable host plant), infected with bacteria and treated with high and low temperatures. However, the mRNA level of JNK significantly increased when treated with fungal pathogens. Furthermore, we found that the amount of phosphorylated JNK increased significantly after fungal infection, while there is no obvious change for phosphorylated p38 and ERK. Our results indicate that the whitefly JNK plays an important role in whitefly’s immune responses to fungal infection.
Collapse
Affiliation(s)
- Lan-Lan Wang
- Ministry of Agriculture Key Laboratory of Agricultural Entomology, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China.
| | | | | | | | | | | |
Collapse
|
25
|
Cholleti H, Paidikondala M, Munir M, Hakhverdyan M, Baule C. Equine arteritis virus induced cell death is associated with activation of the intrinsic apoptotic signalling pathway. Virus Res 2012; 171:222-6. [PMID: 23079113 DOI: 10.1016/j.virusres.2012.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/04/2012] [Accepted: 10/05/2012] [Indexed: 01/09/2023]
Abstract
Equine arteritis virus (EAV) causes a respiratory and reproductive disease in horses, equine viral arteritis. Though cell death in infection with EAV is considered to occur by apoptosis, the underlying molecular mechanism has not been extensively elucidated. We investigated the expression of mRNA of pro-apoptotic and caspase genes during EAV infection in BHK21 cells, a well-established cell type for EAV replication. Using a SYBR Green real-time PCR, mRNA of p53, Bax, caspase 3 and caspase 9 were found up-regulated in a time dependent manner in EAV infected cells. Western blot analysis for caspase 3 and caspase 9 showed expression of cleaved forms of these proteins during EAV infection. In addition, a luminescence-based cell assay for caspase 3/7 activation as a hallmark in apoptosis confirmed apoptotic cell death. The findings demonstrate that cell death in EAV infected BHK21 cells results from apoptosis mediated through the intrinsic signalling pathway.
Collapse
Affiliation(s)
- Harindranath Cholleti
- R&D Unit for Virology, Department of Virology, Immunobiology and Parasitology of the National Veterinary Institute (SVA), Ulls väg 2B, SE-751 89 Uppsala, Sweden
| | | | | | | | | |
Collapse
|
26
|
Hou J, Wang L, Quan R, Fu Y, Zhang H, Feng WH. Induction of interleukin-10 is dependent on p38 mitogen-activated protein kinase pathway in macrophages infected with porcine reproductive and respiratory syndrome virus. Virol J 2012; 9:165. [PMID: 22909062 PMCID: PMC3441385 DOI: 10.1186/1743-422x-9-165] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 08/07/2012] [Indexed: 01/12/2023] Open
Abstract
Background Porcine reproductive and respiratory syndrome virus (PRRSV) causes reproductive failure and respiratory illness in pigs and usually establishes a persistent infection. Previous studies suggested that interleukin-10 (IL-10) could play a critical role in PRRSV-induced immunosuppression. However, the ability of PRRSV to induce IL-10 in infected cells is controversial. In this study, we further investigated this issue using PRRSV strain CH-1a, which is the first North American genotype strain isolated in China. Results PRRSV strain CH-1a could significantly up-regulate IL-10 production both at mRNA and protein levels in porcine alveolar macrophages (PAMs), bone marrow-derived macrophages (BMDMs), and monocyte-derived macrophages (MDMs). However, up-regulation of IL-10 by PRRSV was retarded by specific inhibitors of p38 mitogen-activated protein kinase (MAPK) (SB203580) and NF-κB (BAY11-7082). Additionally, p38 MAPK and NF-κB pathways but not ERK1/2 MAPK were actually activated in PRRSV-infected BMDMs as demonstrated by western blot analysis, suggesting that p38 MAPK and NF-κB pathways are involved in the induction of IL-10 by PRRSV infection. Transfection of PAMs and PAM cell line 3D4/21 (CRL-2843) with viral structural genes showed that glycoprotein5 (GP5) could significantly up-regulate IL-10 production, which was dependent on p38 MAPK and signal transducer and activator of transcription-3 (STAT3) activation. We also demonstrated that a full-length glycoprotein was essential for GP5 to induce IL-10 production. Conclusions PRRSV strain CH-1a could significantly up-regulate IL-10 production through p38 MAPK activation.
Collapse
Affiliation(s)
- Jun Hou
- State Key Laboratories of Agrobiotechnology, Department of Microbiology and Immunology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | | | | | | | | | | |
Collapse
|
27
|
Post-transcriptional control of type I interferon induction by porcine reproductive and respiratory syndrome virus in its natural host cells. Viruses 2012; 4:725-33. [PMID: 22754646 PMCID: PMC3386621 DOI: 10.3390/v4050725] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 04/18/2012] [Accepted: 04/24/2012] [Indexed: 12/28/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is not only a poor inducer of type I interferon but also inhibits the efficient induction of type I interferon by porcine transmissible gastroenteritis virus (TGEV) and synthetic dsRNA molecules, Poly I:C. However, the mechanistic basis by which PRRSV interferes with the induction of type I interferon in its natural host cells remains less well defined. The purposes of this review are to summarize the key findings in supporting the post-transcriptional control of type I interferon in its natural host cells and to propose the possible role of translational control in the regulation of type I interferon induction by PRRSV.
Collapse
|