51
|
Chen S, Yang C, Zhang W, Mahalingam S, Wang M, Cheng A. Flaviviridae virus nonstructural proteins 5 and 5A mediate viral immune evasion and are promising targets in drug development. Pharmacol Ther 2018; 190:1-14. [PMID: 29742479 DOI: 10.1016/j.pharmthera.2018.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Infections with viruses in the Flaviviridae family have a vast global and economic impact because of the high morbidity and mortality. The pathogenesis of Flaviviridae infections is very complex and not fully understood because these viruses can inhibit multiple immune pathways including the complement system, NK cells, and IFN induction and signalling pathways. The non-structural (NS) 5 and 5A proteins of Flaviviridae viruses are highly conserved and play an important role in resisting host immunity through various evasion mechanisms. This review summarizes the strategies used by the NS5 and 5A proteins of Flaviviridae viruses for evading the innate immune response by inhibiting pattern recognition receptor (PRR) signalling pathways (TLR/MyD88, IRF7), suppressing interferon (IFN) signalling pathways (IFN-γRs, STAT1, STAT2), and impairing the function of IFN-stimulated genes (ISGs) (e.g. protein kinase R [PKR], oligoadenylate synthase [OAS]). All of these immune evasion mechanisms depend on the interaction of NS5 or NS5A with cellular proteins, such as MyD88 and IRF7, IFN-αRs, IFN-γRs, STAT1, STAT2, PKR and OAS. NS5 is the most attractive target for the discovery of broad spectrum compounds against Flaviviridae virus infection. The methyltransferase (MTase) and RNA-dependent RNA polymerase (RdRp) activities of NS5 are the main therapeutic targets for antiviral drugs against Flaviviridae virus infection. Based on our site mapping, the sites involved in immune evasion provide some potential and promising targets for further novel antiviral therapeutics.
Collapse
Affiliation(s)
- Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan 611130, China.
| | - Chao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Wei Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Suresh Mahalingam
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan 611130, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan 611130, China.
| |
Collapse
|
52
|
Mahony R, Broadbent L, Maier-Moore JS, Power UF, Jefferies CA. The RNA binding protein La/SS-B promotes RIG-I-mediated type I and type III IFN responses following Sendai viral infection. Sci Rep 2017; 7:14537. [PMID: 29109527 PMCID: PMC5673980 DOI: 10.1038/s41598-017-15197-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/23/2017] [Indexed: 12/28/2022] Open
Abstract
La/SS-B (or La) is a 48 kDa RNA-binding protein and an autoantigen in autoimmune disorders such as systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS). La involvement in regulating the type I interferon (IFN) response is controversial - acting through both positive and negative regulatory mechanisms; inhibiting the IFN response and enhancing viral growth, or directly inhibiting viral replication. We therefore sought to clarify how La regulates IFN production in response to viral infection. ShRNA knockdown of La in HEK 293 T cells increased Sendai virus infection efficiency, decreased IFN-β, IFN-λ1, and interferon-stimulated chemokine gene expression. In addition, knockdown attenuated CCL-5 and IFN-λ1 secretion. Thus, La has a positive role in enhancing type I and type III IFN production. Mechanistically, we show that La directly binds RIG-I and have mapped this interaction to the CARD domains of RIG-I and the N terminal domain of La. In addition, we showed that this interaction is induced following RIG-I activation and that overexpression of La enhances RIG-I-ligand binding. Together, our results demonstrate a novel role for La in mediating RIG-I-driven responses downstream of viral RNA detection, ultimately leading to enhanced type I and III IFN production and positive regulation of the anti-viral response.
Collapse
Affiliation(s)
- Rebecca Mahony
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Lindsay Broadbent
- Centre for Experimental Medicine, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland
| | - Jacen S Maier-Moore
- The University of Texas at El Paso College of Health Sciences, Clinical Laboratory Sciences Program, 500 W. University Avenue, El Paso, Texas, 79968, USA
| | - Ultan F Power
- Centre for Experimental Medicine, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland
| | - Caroline A Jefferies
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
- Division of Rheumatology, Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Centre, 8700 Beverly Blvd, Los Angeles, California, 90048, USA.
| |
Collapse
|
53
|
Functional validation of ATF4 and GADD34 in Neuro2a cells by CRISPR/Cas9-mediated genome editing. Mol Cell Biochem 2017; 440:65-75. [PMID: 28825160 DOI: 10.1007/s11010-017-3156-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/05/2017] [Indexed: 12/13/2022]
Abstract
Activating transcription factor 4 (ATF4), which is ubiquitously expressed, plays a crucial role in regulating various stress-responsive genes under pathophysiological conditions. Further, growth arrest and DNA damage-inducible gene 34 (GADD34), a downstream target of ATF4, has been reported to negatively regulate ATF4 expression. To understand the relationship between intrinsic ATF4 and GADD34 under resting and ER stress conditions, we used a novel gene editing approach, CRISPR/Cas9, to integrate antibiotic-resistant genes into the target genes, ATF4 and GADD34. First, we manipulated the ATF4 gene in the mouse neuroblastoma cell line, Neuro2a, and compared the ER stress responses between parental and ATF4-edited Neuro2a cells. Next, we established Neuro2a cells with edited GADD34 and ATF4/GADD34 genes and found that ATF4 acts as a proapoptotic factor, but GADD34 depletion did not attenuate the expression of cleaved caspase-3 induced by tunicamycin treatment. These findings provide new insights into the ATF4 signaling cascades. Additionally, the rapid establishment of cells lacking multiple genes using this CRISPR/Cas9 system will be a powerful tool for exploring various cellular issues under pathophysiological conditions.
Collapse
|
54
|
Chang YH, Lau KS, Kuo RL, Horng JT. dsRNA Binding Domain of PKR Is Proteolytically Released by Enterovirus A71 to Facilitate Viral Replication. Front Cell Infect Microbiol 2017; 7:284. [PMID: 28702377 PMCID: PMC5487429 DOI: 10.3389/fcimb.2017.00284] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 06/12/2017] [Indexed: 01/18/2023] Open
Abstract
Enterovirus 71 (EV-A71) causes hand, foot and mouth disease in young children and infants, but can also cause severe neurological complications or even death. The double-stranded RNA (dsRNA)-dependent protein kinase R (PKR), an interferon-induced antiviral protein, phosphorylates the regulatory α-subunit of the eukaryotic translation initiation factor 2 in response to viral infection, thereby blocking the translation of cellular and viral mRNA and promoting apoptosis. The cleavage of PKR after infection with poliovirus, a prototype enterovirus, has been reported by others, but the underlying mechanism of this cleavage and its role in viral replication remain unclear. In the present study, we show that viral 3C protease cleaves PKR at a site, Q188, which differs from the site cleaved during apoptosis, D251. In contrast to the conventional phosphorylation of PKR by dsRNA, EV-A71 3C physically interacts with PKR to mediate the phosphorylation of PKR; this effect is dependent on 3C protease activity. Overexpression of a catalytically inactive PKR mutant (K296H) accelerates viral protein accumulation and increases virus titer, whereas a K64E substitution in the dsRNA binding site abolishes this advantage. We also demonstrate that PKR cleavage mediated by EV-A71 3C protease produces a short N-terminal PKR fragment that can enhance EV-A71 replication, in terms of viral RNA, viral protein, and viral titers. We conclude that PKR is co-opted by EV-A71 via viral protease 3C-mediated proteolytic activation to facilitate viral replication.
Collapse
Affiliation(s)
- Yu-Hsiu Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan.,National Defense Medical Center, Institute of Preventive MedicineTaipei, Taiwan
| | - Kean Seng Lau
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan
| | - Rei-Lin Kuo
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan.,Molecular Infectious Disease Research Center, Chang Gung Memorial HospitalTaoyuan, Taiwan
| | - Jim-Tong Horng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan.,Research Center for Emerging Viral Infections, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan.,Molecular Infectious Disease Research Center, Chang Gung Memorial HospitalTaoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and TechnologyTaoyuan, Taiwan
| |
Collapse
|
55
|
Mahony R, Gargan S, Roberts KL, Bourke N, Keating SE, Bowie AG, O'Farrelly C, Stevenson NJ. A novel anti-viral role for STAT3 in IFN-α signalling responses. Cell Mol Life Sci 2017; 74:1755-1764. [PMID: 27988795 PMCID: PMC11107673 DOI: 10.1007/s00018-016-2435-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 11/16/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
Abstract
The cytokine, Interferon (IFN)-α, induces a wide spectrum of anti-viral mediators, via the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. STAT1 and STAT2 are well characterised to upregulate IFN-stimulated gene (ISG) expression; but even though STAT3 is also activated by IFN-α, its role in anti-viral ISG induction is unclear. Several viruses, including Hepatitis C and Mumps, reduce cellular STAT3 protein levels, via the promotion of ubiquitin-mediated proteasomal degradation. This viral immune evasion mechanism suggests an undiscovered anti-viral role for STAT3 in IFN-α signalling. To investigate STAT3's functional involvement in this Type I IFN pathway, we first analysed its effect upon the replication of two viruses, Influenza and Vaccinia. Viral plaque assays, using Wild Type (WT) and STAT3-/- Murine Embryonic Fibroblasts (MEFs), revealed that STAT3 is required for the inhibition of Influenza and Vaccinia replication. Furthermore, STAT3 shRNA knockdown also enhanced Influenza replication and hindered induction of several, well characterised, anti-viral ISGs: PKR, OAS2, MxB and ISG15; while STAT3 expression had no effect upon induction of a separate ISG group: Viperin, IFI27, CXCL10 and CCL5. These discoveries reveal, for the first time, an anti-viral role for STAT3 in the IFN-α pathway and characterise a requirement for STAT3 in the expression of specific ISGs. These findings also identify STAT3 as a therapeutic target against viral infection and highlight it as an essential pathway component for endogenous and therapeutic IFN-α responsiveness.
Collapse
Affiliation(s)
- Rebecca Mahony
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Siobhán Gargan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Kim L Roberts
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Nollaig Bourke
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sinead E Keating
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
56
|
Inducible Rubicon facilitates viral replication by antagonizing interferon production. Cell Mol Immunol 2017; 14:607-620. [PMID: 28392573 DOI: 10.1038/cmi.2017.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/13/2016] [Accepted: 12/13/2016] [Indexed: 12/16/2022] Open
Abstract
The RUN domain Beclin-1-interacting cysteine-rich-containing (Rubicon) protein is involved in the maturation step of autophagy and the endocytic pathway as a Beclin-1-binding partner, but little is known regarding the role of Rubicon during viral infection. Here, we performed functional studies of the identified target in interferon (IFN) signaling pathways associated with Rubicon to elucidate the mechanisms of viral resistance to IFN. The Rubicon protein levels were elevated in peripheral blood mononuclear cells, sera and liver tissues from patients with hepatitis B virus (HBV) infection relative to those in healthy individuals. Assays of the overexpression and knockdown of Rubicon showed that Rubicon significantly promoted HBV replication. In addition, Rubicon knockdown resulted in the inhibition of enterovirus 71, influenza A virus and vesicular stomatitis virus. The expression o0f Rubicon led to the suppression of virus-induced type-I interferon (IFN-α and IFN-β) and type-III interferon (IFN-λ1). Translocation of activated IRF3 and IRF7 from the cytoplasm to the nucleus was involved in this process, and the NF-κB essential modulator (NEMO), a key factor in the IFN pathway, was the target with which Rubicon interacted. Our results reveal a previously unrecognized function of Rubicon as a virus-induced protein that binds to NEMO, leading to the inhibition of type-I interferon production. Rubicon thus functions as an important negative regulator of the innate immune response, enhances viral replication and may play a role in viral immune evasion.
Collapse
|
57
|
Hu Z, Zhang H, Tang L, Lou M, Geng Y. Silencing nc886, a Non-Coding RNA, Induces Apoptosis of Human Endometrial Cancer Cells-1A In Vitro. Med Sci Monit 2017; 23:1317-1324. [PMID: 28298621 PMCID: PMC5365049 DOI: 10.12659/msm.900320] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The role that nc886, a non-coding microRNA, plays in human endometrial cancer is unknown. The present study aimed to describe the functional role of nc886 in human endometrial cancer-1A (HEC-1A) cell line, which may provide another target for human endometrial cancer treatment. Material/Methods The expression levels of nv886 in normal human endometrial tissue and the early phase and late phase of human endometrial cancer tissues were determined and compared by fluorescence in situ hybridization (FISH). Small interference RNA (siRNA) was used to inhibit nc886, and cell proliferation was evaluated with the MTT test. mRNA levels of PKR, NF-κB, vascular endothelial growth factor (VEGF), and caspase-3 were determined against glyceraldehyde 3-phosphate dehydrogenase (GAPDH between the HEC-1A control group and the silenced group (nc886 silenced with siRNA) by real-time reverse transcription polymerase chain reaction (RT-PCR). The protein levels of PKR (total and phosphorylated form), NF-κB, VEGF, and caspase-3 were determined against GAPDH by Western blotting, and cell apoptosis was determined by flow cytometry. Results Our results indicated that a higher level of nc886 was expressed in the late phase of human endometrial cancer tissue, less than in the early phase but still higher than in normal human endometrial tissue. After nc886 was silenced, protein levels of p-PKR (phosphorylated PKR) and caspase-3 were increased, whereas NF-κB and VEGF were decreased. Conclusions The rate of apoptosis in the silenced group was increased and the rate of cell proliferation was slower in comparison to the control.
Collapse
Affiliation(s)
- Zhuoying Hu
- Department of Obstetrics and Gynecology, The 1st Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Hongyu Zhang
- Department of General Surgery, The 1st Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Liangdan Tang
- Department of Obstetrics and Gynecology, The 1st Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Meng Lou
- Department of Obstetrics and Gynecology, The 1st Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Yanqing Geng
- Department of Obstetrics and Gynecology, The 1st Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
58
|
Xu X, Sun Q, Yu X, Zhao L. Rescue of nonlytic Newcastle Disease Virus (NDV) expressing IL-15 for cancer immunotherapy. Virus Res 2017; 233:35-41. [PMID: 28286036 DOI: 10.1016/j.virusres.2017.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/28/2017] [Accepted: 03/01/2017] [Indexed: 02/05/2023]
Abstract
In order to test and enhance the antitumor activity against mice melanoma by NDV-modified tumor vaccine, a recombinant NDV expressing IL-15 (LX/(IL-15)) was generated by reverse genetics. Then, the expression level and biological activity of IL-15 were examined. Our results showed that mice tumor cell lines infected with LX/(IL-15) expressed IL-15 at a high level, and that expressed IL-15 was biologically active. Expression kinetics demonstrated that the highest expression level of IL-15 was at 48h post infection. The cytotoxicity assay showed that murine melanoma cells modified with LX/(IL-15) could significantly enhance the antitumor immune response in vitro. In vivo study also showed that murine melanoma cells modified with LX/(IL-15) could prevent melanoma growth in mice. Taken together, our data strongly indicated that recombinant LX/(IL-15) is a promising agent for cancer immunotherapy both for human and animal.
Collapse
Affiliation(s)
- Xiaojing Xu
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123 Suzhou, People's Republic of China
| | - Qing Sun
- Laboratory of Animal Infectious Diseases, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, People's Republic of China; Virus Research Unit, Department of Microbiology and Immunology, School of Medicine, University of Otago, New Zealand
| | - Xiao Yu
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123 Suzhou, People's Republic of China
| | - Lixiang Zhao
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123 Suzhou, People's Republic of China.
| |
Collapse
|
59
|
Santhakumar D, Rubbenstroth D, Martinez-Sobrido L, Munir M. Avian Interferons and Their Antiviral Effectors. Front Immunol 2017; 8:49. [PMID: 28197148 PMCID: PMC5281639 DOI: 10.3389/fimmu.2017.00049] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/12/2017] [Indexed: 12/12/2022] Open
Abstract
Interferon (IFN) responses, mediated by a myriad of IFN-stimulated genes (ISGs), are the most profound innate immune responses against viruses. Cumulatively, these IFN effectors establish a multilayered antiviral state to safeguard the host against invading viral pathogens. Considerable genetic and functional characterizations of mammalian IFNs and their effectors have been made, and our understanding on the avian IFNs has started to expand. Similar to mammalian counterparts, three types of IFNs have been genetically characterized in most avian species with available annotated genomes. Intriguingly, chickens are capable of mounting potent innate immune responses upon various stimuli in the absence of essential components of IFN pathways including retinoic acid-inducible gene I, IFN regulatory factor 3 (IRF3), and possibility IRF9. Understanding these unique properties of the chicken IFN system would propose valuable targets for the development of potential therapeutics for a broader range of viruses of both veterinary and zoonotic importance. This review outlines recent developments in the roles of avian IFNs and ISGs against viruses and highlights important areas of research toward our understanding of the antiviral functions of IFN effectors against viral infections in birds.
Collapse
Affiliation(s)
| | - Dennis Rubbenstroth
- Institute for Virology, Faculty of Medicine, University Medical Center, University of Freiburg , Freiburg , Germany
| | - Luis Martinez-Sobrido
- Department of Microbiology and Immunology, University of Rochester Medical Center , Rochester, NY , USA
| | | |
Collapse
|
60
|
Abstract
Organisms throughout biology need to maintain the integrity of their genome. From bacteria to vertebrates, life has established sophisticated mechanisms to detect and eliminate foreign genetic material or to restrict its function and replication. Tremendous progress has been made in the understanding of these mechanisms which keep foreign or unwanted nucleic acids from viruses or phages in check. Mechanisms reach from restriction-modification systems and CRISPR/Cas in bacteria and archaea to RNA interference and immune sensing of nucleic acids, altogether integral parts of a system which is now appreciated as nucleic acid immunity. With inherited receptors and acquired sequence information, nucleic acid immunity comprises innate and adaptive components. Effector functions include diverse nuclease systems, intrinsic activities to directly restrict the function of foreign nucleic acids (e.g., PKR, ADAR1, IFIT1), and extrinsic pathways to alert the immune system and to elicit cytotoxic immune responses. These effects act in concert to restrict viral replication and to eliminate virus-infected cells. The principles of nucleic acid immunity are highly relevant for human disease. Besides its essential contribution to antiviral defense and restriction of endogenous retroelements, dysregulation of nucleic acid immunity can also lead to erroneous detection and response to self nucleic acids then causing sterile inflammation and autoimmunity. Even mechanisms of nucleic acid immunity which are not established in vertebrates are relevant for human disease when they are present in pathogens such as bacteria, parasites, or helminths or in pathogen-transmitting organisms such as insects. This review aims to provide an overview of the diverse mechanisms of nucleic acid immunity which mostly have been looked at separately in the past and to integrate them under the framework nucleic acid immunity as a basic principle of life, the understanding of which has great potential to advance medicine.
Collapse
Affiliation(s)
- G Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, University of Bonn, Bonn, Germany.
| |
Collapse
|
61
|
Pakos-Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A, Gorman AM. The integrated stress response. EMBO Rep 2016; 17:1374-1395. [PMID: 27629041 DOI: 10.15252/embr.201642195] [Citation(s) in RCA: 1538] [Impact Index Per Article: 192.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
In response to diverse stress stimuli, eukaryotic cells activate a common adaptive pathway, termed the integrated stress response (ISR), to restore cellular homeostasis. The core event in this pathway is the phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) by one of four members of the eIF2α kinase family, which leads to a decrease in global protein synthesis and the induction of selected genes, including the transcription factor ATF4, that together promote cellular recovery. The gene expression program activated by the ISR optimizes the cellular response to stress and is dependent on the cellular context, as well as on the nature and intensity of the stress stimuli. Although the ISR is primarily a pro-survival, homeostatic program, exposure to severe stress can drive signaling toward cell death. Here, we review current understanding of the ISR signaling and how it regulates cell fate under diverse types of stress.
Collapse
Affiliation(s)
- Karolina Pakos-Zebrucka
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Izabela Koryga
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Katarzyna Mnich
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Mila Ljujic
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Adrienne M Gorman
- Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
62
|
Abstract
Seasonal and pandemic influenza are the two faces of respiratory infections caused by influenza viruses in humans. As seasonal influenza occurs on an annual basis, the circulating virus strains are closely monitored and a yearly updated vaccination is provided, especially to identified risk populations. Nonetheless, influenza virus infection may result in pneumonia and acute respiratory failure, frequently complicated by bacterial coinfection. Pandemics are, in contrary, unexpected rare events related to the emergence of a reassorted human-pathogenic influenza A virus (IAV) strains that often causes increased morbidity and spreads extremely rapidly in the immunologically naive human population, with huge clinical and economic impact. Accordingly, particular efforts are made to advance our knowledge on the disease biology and pathology and recent studies have brought new insights into IAV adaptation mechanisms to the human host, as well as into the key players in disease pathogenesis on the host side. Current antiviral strategies are only efficient at the early stages of the disease and are challenged by the genomic instability of the virus, highlighting the need for novel antiviral therapies targeting the pulmonary host response to improve viral clearance, reduce the risk of bacterial coinfection, and prevent or attenuate acute lung injury. This review article summarizes our current knowledge on the molecular basis of influenza infection and disease progression, the key players in pathogenesis driving severe disease and progression to lung failure, as well as available and envisioned prevention and treatment strategies against influenza virus infection.
Collapse
Affiliation(s)
- Christin Peteranderl
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Carole Schmoldt
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| |
Collapse
|
63
|
Abd El Halim HM, Alshukri BMH, Ahmad MS, Nakasu EYT, Awwad MH, Salama EM, Gatehouse AMR, Edwards MG. RNAi-mediated knockdown of the voltage gated sodium ion channel TcNav causes mortality in Tribolium castaneum. Sci Rep 2016; 6:29301. [PMID: 27411529 PMCID: PMC4944135 DOI: 10.1038/srep29301] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/24/2016] [Indexed: 01/26/2023] Open
Abstract
The voltage-gated sodium ion channel (VGSC) belongs to the largest superfamily of ion channels. Since VGSCs play key roles in physiological processes they are major targets for effective insecticides. RNA interference (RNAi) is widely used to analyse gene function, but recently, it has shown potential to contribute to novel strategies for selectively controlling agricultural insect pests. The current study evaluates the delivery of dsRNA targeted to the sodium ion channel paralytic A (TcNav) gene in Tribolium castaneum as a viable means of controlling this insect pest. Delivery of TcNav dsRNA caused severe developmental arrest with larval mortalities up to 73% post injection of dsRNA. Injected larvae showed significant (p < 0.05) knockdown in gene expression between 30-60%. Expression was also significantly (p < 0.05) reduced in pupae following injection causing 30% and 42% knockdown for early and late pupal stages, respectively. Oral delivery of dsRNA caused dose-dependant mortalities of between 19 and 51.34%; this was accompanied by significant (p < 0.05) knockdown in gene expression following 3 days of continuous feeding. The majority of larvae injected with, or fed, dsRNA died during the final larval stage prior to pupation. This work provides evidence of a viable RNAi-based strategy for insect control.
Collapse
Affiliation(s)
- Hesham M. Abd El Halim
- Newcastle Institute for Research on Environment and Sustainability, School of Biology, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
- Entomology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Baida M. H. Alshukri
- Newcastle Institute for Research on Environment and Sustainability, School of Biology, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - Munawar S. Ahmad
- Newcastle Institute for Research on Environment and Sustainability, School of Biology, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
- Department of Zoology, University of, Swabi, KPK, Pakistan
| | - Erich Y. T. Nakasu
- Newcastle Institute for Research on Environment and Sustainability, School of Biology, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - Mohammed H. Awwad
- Zoology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Elham M. Salama
- Entomology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Angharad M. R. Gatehouse
- Newcastle Institute for Research on Environment and Sustainability, School of Biology, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - Martin G. Edwards
- Newcastle Institute for Research on Environment and Sustainability, School of Biology, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| |
Collapse
|
64
|
Zhao J, Feng SS. Nanocarriers for delivery of siRNA and co-delivery of siRNA and other therapeutic agents. Nanomedicine (Lond) 2016. [PMID: 26214357 DOI: 10.2217/nnm.15.61] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A major problem in cancer treatment is the multidrug resistance. siRNA inhibitors have great advantages to solve the problem, if the bottleneck of their delivery could be well addressed by the various nanocarriers. Moreover, co-delivery of siRNA together with the various anticancer agents in one nanocarrier may maximize their additive or synergistic effect. This review provides a comprehensive summary on the state-of-the-art of the nanocarriers, which may include prodrugs, micelles, liposomes, dendrimers, nanohydrogels, solid lipid nanoparticles, nanoparticles of biodegradable polymers and nucleic acid nanocarriers for delivery of siRNA and co-delivery of siRNA together with anticancer agents with focus on synthesis of the nanocarrier materials, design and characterization, in vitro and in vivo evaluation, and prospect and challenges of nanocarriers.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Si-Shen Feng
- Department of Chemical & Biomolecular Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
65
|
Dixit U, Pandey AK, Mishra P, Sengupta A, Pandey VN. Staufen1 promotes HCV replication by inhibiting protein kinase R and transporting viral RNA to the site of translation and replication in the cells. Nucleic Acids Res 2016; 44:5271-87. [PMID: 27106056 PMCID: PMC4914112 DOI: 10.1093/nar/gkw312] [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: 03/18/2016] [Accepted: 04/14/2016] [Indexed: 01/23/2023] Open
Abstract
Persistent hepatitis C virus (HCV) infection leads to chronic hepatitis C (CHC), which often progresses to liver cirrhosis (LC) and hepatocellular carcinoma (HCC). The molecular mechanisms that establish CHC and cause its subsequent development into LC and HCC are poorly understood. We have identified a cytoplasmic double-stranded RNA binding protein, Stau1, which is crucial for HCV replication. In this study, Stau1 specifically interacted with the variable-stem-loop region in the 3′ NTR and domain IIId of the HCV-IRES in the 5′ NTR, and promoted HCV replication and translation. Stau1 coimmunoprecipitates HCV NS5B and a cell factor, protein kinase R (PKR), which is critical for interferon-induced cellular antiviral and antiproliferative responses. Like Stau1, PKR displayed binding specificity to domain IIId of HCV-IRES. Stau1 binds to PKR and strongly inhibits PKR-autophosphorylation. We demonstrated that the transport of HCV RNA on the polysomes is Stau1-dependent, being mainly localized in the monosome fractions when Stau1 is downregulated and exclusively localized in the polysomes when Stau1 is overexpressed. Our findings suggest that HCV may appropriate Stau1 to its advantage to prevent PKR-mediated inhibition of eIF2α, which is required for the synthesis of HCV proteins for translocation of viral RNA genome to the polysomes for efficient translation and replication.
Collapse
Affiliation(s)
- Updesh Dixit
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, Rutgers, the State University of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Ashutosh K Pandey
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, Rutgers, the State University of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Priya Mishra
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, Rutgers, the State University of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Amitabha Sengupta
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, Rutgers, the State University of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Virendra N Pandey
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, Rutgers, the State University of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| |
Collapse
|
66
|
Wu C, Hu Y, Fan L, Wang H, Sun Z, Deng S, Liu Y, Hu C. Ctenopharyngodon idella PKZ facilitates cell apoptosis through phosphorylating eIF2α. Mol Immunol 2016; 69:13-23. [DOI: 10.1016/j.molimm.2015.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/08/2015] [Accepted: 11/11/2015] [Indexed: 11/24/2022]
|
67
|
Song Y, Wan X, Gao L, Pan Y, Xie W, Wang H, Guo J. Activated PKR inhibits pancreatic β-cell proliferation through sumoylation-dependent stabilization of P53. Mol Immunol 2015; 68:341-9. [PMID: 26446704 DOI: 10.1016/j.molimm.2015.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/27/2015] [Accepted: 09/02/2015] [Indexed: 12/20/2022]
Abstract
Double-stranded RNA-dependent protein kinase (PKR) is intimately involved in type 2 diabetes due to its role in insulin resistance in peripheral tissues and anti-proliferative effect on pancreatic β-cells. Activated PKR was found to inhibit β-cell proliferation, partially through accumulation of P53. However the molecular mechanisms underlying PKR-dependent upregulation of P53 remain unknown. The results of the present study showed that PKR can be specifically activated in PKR overexpressing β-cells by a low dosage of the previously synthesized compound 1H-benzimidazole1-ethanol,2,3-dihydro-2-imino-a-(phenoxymethyl)-3-(phenylmethyl)-,monohydrochloride (BEPP), and this led to upregulation of P53 through sumoylation-dependent stability. Activated PKR was found to interact with sumo-conjugating enzyme Ubc9, and P53 sumoylation relies on a PKR-Ubc9 protein-protein interaction. Additionally, a ceramide signal was needed for PKR activation to be triggered by glucolipotoxicity and TNFα stimulation, and stabilization of P53 required endogenous ceramide accumulation. Glucolipotoxicity and pro-inflammatory cytokines therefore promote the sumoylation-dependent stability of P53 via the ceramide/PKR/Ubc9 signalling pathway that is involved in pancreatic β-cell proliferation inhibition in the development of type 2 diabetes.
Collapse
Affiliation(s)
- Ying Song
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - XiaoMeng Wan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - LiLi Gao
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - Yi Pan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - WeiPing Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Hong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.
| | - Jun Guo
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| |
Collapse
|
68
|
Bou-Nader C, Pecqueur L, Bregeon D, Kamah A, Guérineau V, Golinelli-Pimpaneau B, Guimarães BG, Fontecave M, Hamdane D. An extended dsRBD is required for post-transcriptional modification in human tRNAs. Nucleic Acids Res 2015; 43:9446-56. [PMID: 26429968 PMCID: PMC4627097 DOI: 10.1093/nar/gkv989] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/19/2015] [Indexed: 12/25/2022] Open
Abstract
In tRNA, dihydrouridine is a conserved modified base generated by the post-transcriptional reduction of uridine. Formation of dihydrouridine 20, located in the D-loop, is catalyzed by dihydrouridine synthase 2 (Dus2). Human Dus2 (HsDus2) expression is upregulated in lung cancers, offering a growth advantage throughout its ability to interact with components of the translation apparatus and inhibit apoptosis. Here, we report the crystal structure of the individual domains of HsDus2 and their functional characterization. HsDus2 is organized into three major modules. The N-terminal catalytic domain contains the flavin cofactor involved in the reduction of uridine. The second module is the conserved α-helical domain known as the tRNA binding domain in HsDus2 homologues. It is connected via a flexible linker to an unusual extended version of a dsRNA binding domain (dsRBD). Enzymatic assays and yeast complementation showed that the catalytic domain binds selectively NADPH but cannot reduce uridine in the absence of the dsRBD. While in Dus enzymes from bacteria, plants and fungi, tRNA binding is essentially achieved by the α-helical domain, we showed that in HsDus2 this function is carried out by the dsRBD. This is the first reported case of a tRNA-modifying enzyme carrying a dsRBD used to bind tRNAs.
Collapse
Affiliation(s)
- Charles Bou-Nader
- Laboratoire de Chimie des Processus Biologiques, CNRS-UMR 8229, Collège De France, France, 11 place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | - Ludovic Pecqueur
- Laboratoire de Chimie des Processus Biologiques, CNRS-UMR 8229, Collège De France, France, 11 place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | - Damien Bregeon
- Sorbonne Universités, UPMC Univ. Paris 06, IBPS, UMR8256, Biology of Aging and Adaptation, F-75005 Paris, France
| | - Amina Kamah
- Université de Lille-Nord de France, CNRS UMR 8576, Institut Fédératif de Recherches 147, Villeneuve d'Ascq, France
| | - Vincent Guérineau
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Béatrice Golinelli-Pimpaneau
- Laboratoire de Chimie des Processus Biologiques, CNRS-UMR 8229, Collège De France, France, 11 place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | - Beatriz G Guimarães
- Synchrotron SOLEIL, L'Orme des Merisiers, Saint-Aubin, 91190 Gif-sur-Yvette, France
| | - Marc Fontecave
- Laboratoire de Chimie des Processus Biologiques, CNRS-UMR 8229, Collège De France, France, 11 place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | - Djemel Hamdane
- Laboratoire de Chimie des Processus Biologiques, CNRS-UMR 8229, Collège De France, France, 11 place Marcelin Berthelot, 75231 Paris Cedex 05, France
| |
Collapse
|
69
|
van ‘t Wout EFA, van Schadewijk A, van Boxtel R, Dalton LE, Clarke HJ, Tommassen J, Marciniak SJ, Hiemstra PS. Virulence Factors of Pseudomonas aeruginosa Induce Both the Unfolded Protein and Integrated Stress Responses in Airway Epithelial Cells. PLoS Pathog 2015; 11:e1004946. [PMID: 26083346 PMCID: PMC4471080 DOI: 10.1371/journal.ppat.1004946] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 05/11/2015] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa infection can be disastrous in chronic lung diseases such as cystic fibrosis and chronic obstructive pulmonary disease. Its toxic effects are largely mediated by secreted virulence factors including pyocyanin, elastase and alkaline protease (AprA). Efficient functioning of the endoplasmic reticulum (ER) is crucial for cell survival and appropriate immune responses, while an excess of unfolded proteins within the ER leads to “ER stress” and activation of the “unfolded protein response” (UPR). Bacterial infection and Toll-like receptor activation trigger the UPR most likely due to the increased demand for protein folding of inflammatory mediators. In this study, we show that cell-free conditioned medium of the PAO1 strain of P. aeruginosa, containing secreted virulence factors, induces ER stress in primary bronchial epithelial cells as evidenced by splicing of XBP1 mRNA and induction of CHOP, GRP78 and GADD34 expression. Most aspects of the ER stress response were dependent on TAK1 and p38 MAPK, except for the induction of GADD34 mRNA. Using various mutant strains and purified virulence factors, we identified pyocyanin and AprA as inducers of ER stress. However, the induction of GADD34 was mediated by an ER stress-independent integrated stress response (ISR) which was at least partly dependent on the iron-sensing eIF2α kinase HRI. Our data strongly suggest that this increased GADD34 expression served to protect against Pseudomonas-induced, iron-sensitive cell cytotoxicity. In summary, virulence factors from P. aeruginosa induce ER stress in airway epithelial cells and also trigger the ISR to improve cell survival of the host. Pseudomonas aeruginosa causes a devastating infection when it affects patients with cystic fibrosis or other chronic lung diseases. It often causes chronic infection due to its resistance to antibiotic treatment and its ability to form biofilms in these patients. The toxic effects of P. aeruginosa are largely mediated by secreted virulence factors. Efficient functioning of the endoplasmic reticulum is crucial for cell survival and appropriate immune responses, while its dysfunction causes stress and activation of the unfolded protein response. In this study, we found that virulence factors secreted by P. aeruginosa trigger the unfolded protein response in human cells by causing endoplasmic reticulum stress. In addition, secreted virulence factors activate the integrated stress response via a parallel independent pathway. Both stress pathways lead to the induction of the protein GADD34, which appears to provide protection against the toxic effects of the secreted virulence factors.
Collapse
Affiliation(s)
- Emily F. A. van ‘t Wout
- Department of Pulmonology, Leiden University Medical Centre, Leiden, the Netherlands
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, United Kingdom
| | | | - Ria van Boxtel
- Department of Molecular Microbiology, Utrecht University, Utrecht, the Netherlands
| | - Lucy E. Dalton
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, United Kingdom
| | - Hanna J. Clarke
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, United Kingdom
| | - Jan Tommassen
- Department of Molecular Microbiology, Utrecht University, Utrecht, the Netherlands
| | - Stefan J. Marciniak
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, United Kingdom
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Centre, Leiden, the Netherlands
- * E-mail:
| |
Collapse
|
70
|
Sheehan KCF, Lazear HM, Diamond MS, Schreiber RD. Selective Blockade of Interferon-α and -β Reveals Their Non-Redundant Functions in a Mouse Model of West Nile Virus Infection. PLoS One 2015; 10:e0128636. [PMID: 26010249 PMCID: PMC4444312 DOI: 10.1371/journal.pone.0128636] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 04/30/2015] [Indexed: 01/12/2023] Open
Abstract
Although type I interferons (IFNs) were first described almost 60 years ago, the ability to monitor and modulate the functional activities of the individual IFN subtypes that comprise this family has been hindered by a lack of reagents. The major type I IFNs, IFN-β and the multiple subtypes of IFN-α, are expressed widely and induce their effects on cells by interacting with a shared heterodimeric receptor (IFNAR). In the mouse, the physiologic actions of IFN-α and IFN-β have been defined using polyclonal anti-type I IFN sera, by targeting IFNAR using monoclonal antibodies or knockout mice, or using Ifnb-/- mice. However, the corresponding analysis of IFN-α has been difficult because of its polygenic nature. Herein, we describe two monoclonal antibodies (mAbs) that differentially neutralize murine IFN-β or multiple subtypes of murine IFN-α. Using these mAbs, we distinguish specific contributions of IFN-β versus IFN-α in restricting viral pathogenesis and identify IFN-α as the key mediator of the antiviral response in mice infected with West Nile virus. This study thus suggests the utility of these new reagents in dissecting the antiviral and immunomodulatory roles of IFN-β versus IFN-α in murine models of infection, immunity, and autoimmunity.
Collapse
Affiliation(s)
- Kathleen C. F. Sheehan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Helen M. Lazear
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
71
|
Xiao CX, Xiao JJ, Xu HZ, Wang HH, Chen X, Liu YS, Li P, Shi Y, Nie YZ, Li S, Wu KC, Liu ZJ, Ren JL, Guleng B. Exome sequencing identifies novel compound heterozygous IFNA4 and IFNA10 mutations as a cause of impaired function in Crohn's disease patients. Sci Rep 2015; 5:10514. [PMID: 26000985 PMCID: PMC4441321 DOI: 10.1038/srep10514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 04/23/2015] [Indexed: 12/15/2022] Open
Abstract
Previous studies have highlighted the role of genetic predispositions in disease, and several genes had been identified as important in Crohn’s disease (CD). However, many of these genes are likely rare and not associated with susceptibility in Chinese CD patients. We found 294 shared identical variants in the CD patients of which 26 were validated by Sanger sequencing. Two heterozygous IFN variants (IFNA10 c.60 T > A; IFNA4 c.60 A > T) were identified as significantly associated with CD susceptibility. The single-nucleotide changes alter a cysteine situated before the signal peptide cleavage site to a stop code (TGA) in IFNA10 result in the serum levels of IFNA10 were significantly decreased in the CD patients compared to the controls. Furthermore, the IFNA10 and IFNA4 mutants resulted in an impairment of the suppression of HCV RNA replication in HuH7 cells, and the administration of the recombinant IFN subtypes restored DSS-induced colonic inflammation through the upregulation of CD4+ Treg cells. We identified heterozygous IFNA10 and IFNA4 variants as a cause of impaired function and CD susceptibility genes in Chinese patients from multiple center based study. These findings might provide clues in the understanding of the genetic heterogeneity of CD and lead to better screening and improved treatment.
Collapse
Affiliation(s)
- Chuan-Xing Xiao
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | | | - Hong-Zhi Xu
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | - Huan-Huan Wang
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | - Xu Chen
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | - Yuan-Sheng Liu
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | - Ping Li
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | - Ying Shi
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | - Yong-Zhan Nie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, West Changle Road 15, Xi'an, 710032, China
| | - Shao Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics, Tsinghua University, Beijing, 100084, China
| | - Kai-Chun Wu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, West Changle Road 15, Xi'an, 710032, China
| | - Zhan-Ju Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Jian-Lin Ren
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China
| | - Bayasi Guleng
- 1] Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, 201 Hubin South Road, Xiamen, Fujian Province, 361004, China [2] Faculty of Clinical Medicine, Medical College of Xiamen University, 168 University Road, Xiamen, Fujian Province, 361005, China [3] State Key Laboratory of Cellular Stress Biology, Xiamen University, 168 University Road, Xiamen, Fujian Province, 361005, China
| |
Collapse
|
72
|
Yong Y, Meng Y, Ding H, Fan Z, Tang Y, Zhou C, Luo J, Ke ZJ. PACT/RAX regulates the migration of cerebellar granule neurons in the developing cerebellum. Sci Rep 2015; 5:7961. [PMID: 25609658 PMCID: PMC4302322 DOI: 10.1038/srep07961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/29/2014] [Indexed: 01/03/2023] Open
Abstract
PACT and its murine ortholog RAX were originally identified as a protein activator for the dsRNA-dependent, interferon-inducible protein kinase PKR. Recent studies indicated that RAX played a role in embryogenesis and neuronal development. In this study, we investigated the expression of RAX during the postnatal development of the mouse cerebellum and its role in the migration of cerebellar granule neurons (CGNs). High expression of RAX was observed in the cerebellum from postnatal day (PD) 4 to PD9, a period when the CGNs migrate from the external granule layer (EGL) to the internal granule layer (IGL). The migration of the EGL progenitor cells in vivo was inhibited by RAX knockdown on PD4. This finding was confirmed by in vitro studies showing that RAX knockdown impaired the migration of CGNs in cerebellar microexplants. PACT/RAX-regulated migration required its third motif and was independent of PKR. PACT/RAX interacted with focal adhesion kinase (FAK) and PACT/RAX knockdown disturbed the FAK phosphorylation in CGNs. These findings demonstrated a novel function of PACT/RAX in the regulation of neuronal migration.
Collapse
Affiliation(s)
- Yue Yong
- 1] Department of Biochemistry, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China [2] Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Ya Meng
- 1] Department of Biochemistry, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China [2] Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Hanqing Ding
- Department of Biochemistry, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Zhiqin Fan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Yifen Tang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenghua Zhou
- 1] Department of Biochemistry, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China [2] Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Jia Luo
- Department of Pharmacology and Nutritonal Sciences, University of Kentucky College of Medicine, Lexington, Kentucky 40536, U.S.A
| | - Zun-Ji Ke
- 1] Department of Biochemistry, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China [2] Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
73
|
Acevedo R, Orench-Rivera N, Quarles KA, Showalter SA. Helical defects in microRNA influence protein binding by TAR RNA binding protein. PLoS One 2015; 10:e0116749. [PMID: 25608000 PMCID: PMC4301919 DOI: 10.1371/journal.pone.0116749] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/12/2014] [Indexed: 01/19/2023] Open
Abstract
Background MicroRNAs (miRNAs) are critical post-transcriptional regulators of gene expression. Their precursors have a globally A-form helical geometry, which prevents most proteins from identifying their nucleotide sequence. This suggests the hypothesis that local structural features (e.g., bulges, internal loops) play a central role in specific double-stranded RNA (dsRNA) selection from cellular RNA pools by dsRNA binding domain (dsRBD) containing proteins. Furthermore, the processing enzymes in the miRNA maturation pathway require tandem-dsRBD cofactor proteins for optimal function, suggesting that dsRBDs play a key role in the molecular mechanism for precise positioning of the RNA within these multi-protein complexes. Here, we focus on the tandem-dsRBDs of TRBP, which have been shown to bind dsRNA tightly. Methodology/Principal Findings We present a combination of dsRNA binding assays demonstrating that TRBP binds dsRNA in an RNA-length dependent manner. Moreover, circular dichroism data shows that the number of dsRBD moieties bound to RNA at saturation is different for a tandem-dsRBD construct than for constructs with only one dsRBD per polypeptide, revealing another reason for the selective pressure to maintain multiple domains within a polypeptide chain. Finally, we show that helical defects in precursor miRNA alter the apparent dsRNA size, demonstrating that imperfections in RNA structure influence the strength of TRBP binding. Conclusion/Significance We conclude that TRBP is responsible for recognizing structural imperfections in miRNA precursors, in the sense that TRBP is unable to bind imperfections efficiently and thus is positioned around them. We propose that once positioned around structural defects, TRBP assists Dicer and the rest of the RNA-induced silencing complex (RISC) in providing efficient and homogenous conversion of substrate precursor miRNA into mature miRNA downstream.
Collapse
Affiliation(s)
- Roderico Acevedo
- Department of Chemistry and Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Nichole Orench-Rivera
- Department of Chemistry and Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Kaycee A. Quarles
- Department of Chemistry and Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Scott A. Showalter
- Department of Chemistry and Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
74
|
Friedmann-Morvinski D, Singer O. Overexpression Models: Lentiviral Modeling of Brain Cancer. ACTA ACUST UNITED AC 2015; 3:121-39. [DOI: 10.1002/9780470942390.mo110271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Oded Singer
- The Salk Institute for Biological Studies; La Jolla California
| |
Collapse
|
75
|
Fung TS, Huang M, Liu DX. Coronavirus-induced ER stress response and its involvement in regulation of coronavirus-host interactions. Virus Res 2014; 194:110-23. [PMID: 25304691 PMCID: PMC7114476 DOI: 10.1016/j.virusres.2014.09.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/25/2014] [Accepted: 09/28/2014] [Indexed: 12/11/2022]
Abstract
Coronavirus replication is structurally and functionally associated with the endoplasmic reticulum (ER), a major site of protein synthesis, folding, modification and sorting in the eukaryotic cells. Disturbance of ER homeostasis may occur under various physiological or pathological conditions. In response to the ER stress, signaling pathways of the unfolded protein response (UPR) are activated. UPR is mediated by three ER transmembrane sensors, namely the PKR-like ER protein kinase (PERK), the inositol-requiring protein 1 (IRE1) and the activating transcriptional factor 6 (ATF6). UPR facilitates adaptation to ER stress by reversible translation attenuation, enhancement of ER protein folding capacity and activation of ER-associated degradation (ERAD). In cells under prolonged and irremediable ER stress, UPR can also trigger apoptotic cell death. Accumulating evidence has shown that coronavirus infection causes ER stress and induces UPR in the infected cells. UPR is closely associated with a number of major signaling pathways, including autophagy, apoptosis, the mitogen-activated protein (MAP) kinase pathways, innate immunity and pro-inflammatory response. Therefore, studies on the UPR are pivotal in elucidating the complicated issue of coronavirus-host interaction. In this paper, we present the up-to-date knowledge on coronavirus-induced UPR and discuss its potential involvement in regulation of innate immunity and apoptosis.
Collapse
Affiliation(s)
- To Sing Fung
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Mei Huang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Ding Xiang Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551.
| |
Collapse
|
76
|
Killiny N, Hajeri S, Tiwari S, Gowda S, Stelinski LL. Double-stranded RNA uptake through topical application, mediates silencing of five CYP4 genes and suppresses insecticide resistance in Diaphorina citri. PLoS One 2014; 9:e110536. [PMID: 25330026 PMCID: PMC4203802 DOI: 10.1371/journal.pone.0110536] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/16/2014] [Indexed: 01/07/2023] Open
Abstract
Silencing of genes through RNA interference (RNAi) in insects has gained momentum during the past few years. RNAi has been used to cause insect mortality, inhibit insect growth, increase insecticide susceptibility, and prevent the development of insecticide resistance. We investigated the efficacy of topically applied dsRNA to induce RNAi for five Cytochrome P450 genes family 4 (CYP4) in Diaphorina citri. We previously reported that these CYP4 genes are associated with the development of insecticide resistance in D. citri. We targeted five CYP4 genes that share a consensus sequence with one dsRNA construct. Quantitative PCR confirmed suppressed expression of the five CYP4 genes as a result of dsRNA topically applied to the thoracic region of D. citri when compared to the expression levels in a control group. Western blot analysis indicated a reduced signal of cytochrome P450 proteins (45 kDa) in adult D. citri treated with the dsRNA. In addition, oxidase activity and insecticide resistance were reduced for D. citri treated with dsRNA that targeted specific CYP4 genes. Mortality was significantly higher in adults treated with dsRNA than in adults treated with water. Our results indicate that topically applied dsRNA can penetrate the cuticle of D. citri and induce RNAi. These results broaden the scope of RNAi as a mechanism to manage pests by targeting a broad range of genes. The results also support the application of RNAi as a viable tool to overcome insecticide resistance development in D. citri populations. However, further research is needed to develop grower-friendly delivery systems for the application of dsRNA under field conditions. Considering the high specificity of dsRNA, this tool can also be used for management of D. citri by targeting physiologically critical genes involved in growth and development.
Collapse
Affiliation(s)
- Nabil Killiny
- Department of Entomology and Nematology, Citrus Research and Education Center, IFAS, University of Florida, Lake Alfred, Florida, United States of America
- * E-mail:
| | - Subhas Hajeri
- Department of Plant Pathology, Citrus Research and Education Center, IFAS, University of Florida, Lake Alfred, Florida, United States of America
| | - Siddharth Tiwari
- Department of Entomology and Nematology, Citrus Research and Education Center, IFAS, University of Florida, Lake Alfred, Florida, United States of America
| | - Siddarame Gowda
- Department of Plant Pathology, Citrus Research and Education Center, IFAS, University of Florida, Lake Alfred, Florida, United States of America
| | - Lukasz L. Stelinski
- Department of Entomology and Nematology, Citrus Research and Education Center, IFAS, University of Florida, Lake Alfred, Florida, United States of America
| |
Collapse
|
77
|
Yamashiro LH, Oliveira SC, Báfica A. Innate immune sensing of nucleic acids from mycobacteria. Microbes Infect 2014; 16:991-7. [PMID: 25284681 DOI: 10.1016/j.micinf.2014.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 01/16/2023]
Abstract
Endosomal and cytosolic receptors engage recognition of mycobacterial-derived nucleic acids (MyNAs). In contrast, virulent mycobacteria may utilize nucleic acid recognition pathways to escape the host immune system. This short review will summarize the mechanisms by which MyNAs are sensed and how they influence host protective responses.
Collapse
Affiliation(s)
- Lívia Harumi Yamashiro
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Brazil; Pharmacology Graduate Program, Federal University of Santa Catarina, Brazil
| | - Sérgio Costa Oliveira
- Laboratory of Immunology and Infectious Diseases, Federal University of Minas Gerais, Brazil
| | - André Báfica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Brazil; Pharmacology Graduate Program, Federal University of Santa Catarina, Brazil.
| |
Collapse
|
78
|
André ND, Silva VAO, Watanabe MAE, De Lucca FL. Intratumoral injection of PKR shRNA expressing plasmid inhibits B16-F10 melanoma growth. Oncol Rep 2014; 32:2267-73. [PMID: 25175769 DOI: 10.3892/or.2014.3410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/04/2014] [Indexed: 11/06/2022] Open
Abstract
The RNA-dependent protein kinase (PKR) is a serine/threonine kinase that is involved in the regulation of important cell processes such as apoptosis, signal transduction, cell proliferation and differentiation. However, the role played by PKR in cancer remains controversial. RNA interference (RNAi) has currently become an important technique in understanding gene function. Previously, we showed that PKR shRNA downregulates PKR expression in B16-F10 melanoma cells and reduces the metastatic potential of these tumor cells. In the present study, we examined the effect of the intratumoral injection of PKR shRNA‑expressing plasmid on the growth of B16-F10 melanoma in mice. The results showed that this treatment significantly reduced tumor growth. Thus, these findings suggested that PKR acts as a tumor suppressor, a finding that is consistent with our previous study on the experimental model of metastasis. Moreover, the results suggested that this effect may be mediated by the transcription factor NF-κB. The present study confirmed the hypothesis that the direct administration of RNAi-based therapeutics in the target tumor is a promising approach for overcoming the obstacles of systemic delivery. The results also suggested that the intratumoral injection of PKR shRNA‑expressing vector is a novel therapeutic approach for human solid tumors such as cutaneous melanoma and breast cancer, since PKR is overexpressed in these tumors.
Collapse
Affiliation(s)
| | - Viviane Aline Oliveira Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Maria Angelica Ehara Watanabe
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, 86057-970 Londrina, PR, Brazil
| | - Fernando Luiz De Lucca
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| |
Collapse
|
79
|
Yong Y, Luo J, Ke ZJ. dsRNA binding protein PACT/RAX in gene silencing, development and diseases. ACTA ACUST UNITED AC 2014; 9:382-388. [PMID: 25554729 DOI: 10.1007/s11515-014-1325-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PACT (Protein kinase, interferon-inducible double stranded RNA dependent activator) and its murine ortholog RAX (PKR-associated protein X) were originally identified as a protein activator for the dsRNA-dependent, interferon-inducible protein kinase (PKR). Endogenous PACT/RAX activates PKR in response to diverse stress signals such as serum starvation, and peroxide or arsenite treatment. PACT/RAX heterodimerized with PKR and activated it with its third motif in the absence of dsRNA. The activation of PKR leads to enhanced eIF2α phosphorylation followed by apoptosis or inhibition of growth. Besides the role of activating PKR, PACT is associated with a ~500 kDa complex that contains Dicer, hAgo2, and TRBP (TAR RNA binding protein) and it associates with Dicer to facilitate the production of small interfering RNA. PACT/RAX plays an important role in diverse physiological and pathological processes. Pact-/- mice exhibit notable developmental abnormalities including microtia, with craniofacial ear, and hearing defects. Pact-/- mice had smaller body sizes and fertility defects, both of which were caused by defective pituitary functions. It was found that dRAX disrupted fly embryos homozygous, displayed highly abnormal commissural axon structure of the central nervous system, and 70% of the flies homozygous for the mutant allele died prior to adulthood. Using high density SNP genotyping arrays, it was found that a mutation in PRKRA (the PACT/RAX gene) is the causative genetic mutation in DYT16, a novel autosomal recessive dystonia-parkinsonism syndrome in Brazilian patients.
Collapse
Affiliation(s)
- Yue Yong
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA
| | - Zun-Ji Ke
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China ; Department of Biochemistry, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| |
Collapse
|
80
|
Antagonizing interferon-mediated immune response by porcine reproductive and respiratory syndrome virus. BIOMED RESEARCH INTERNATIONAL 2014; 2014:315470. [PMID: 25101271 PMCID: PMC4101967 DOI: 10.1155/2014/315470] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/18/2014] [Accepted: 06/18/2014] [Indexed: 11/25/2022]
Abstract
Interferons (IFNs) are important components in innate immunity involved in the first line of defense to protect host against viral infection. Porcine reproductive and respiratory syndrome virus (PRRSV) leads to severe economic losses for swine industry since being first identified in early 1990s. PRRSV interplays with host IFN production and IFN-activated signaling, which may contribute to the delayed onset and low level of neutralizing antibodies, as well as weak cell-mediated immune response in infected pigs. PRRSV encodes several proteins that act as antagonists for the IFN signaling. In this review, we summarized the various strategies used by PRRSV to antagonize IFN production and thwart IFN-activated antiviral signaling, as well as the variable interference with IFN-mediated immune response by different PRRSV strains. Thorough understanding of the interaction between PRRSV and host innate immune response will facilitate elucidation of PRRSV pathogenesis and development of a better strategy to control PRRS.
Collapse
|
81
|
Zhao L, Mei Y, Sun Q, Guo L, Wu Y, Yu X, Hu B, Liu X, Liu H. Autologous tumor vaccine modified with recombinant new castle disease virus expressing IL-7 promotes antitumor immune response. THE JOURNAL OF IMMUNOLOGY 2014; 193:735-45. [PMID: 24943214 DOI: 10.4049/jimmunol.1400004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Autologous tumor vaccine modified with nonlytic Newcastle disease virus (ATV-NDV) is a promising vaccine for cancer immunotherapy. IL-7 plays a critical role in lymphocyte development and homeostasis. To improve the efficacy of ATV-NDV, we inserted the murine IL-7 gene into the genome of nonlytic NDV strain LX using reverse genetic system. The insertion of the IL-7 gene neither affected the main features of NDV replication nor its tumor selectivity. The gene product was biologically active and stable. Then we tested the antitumor effects of the autologous tumor vaccine modified with LX/(IL-7) in the murine tumor models. We showed that tumor cells modified with LX/IL-7 induced a strong antitumor activity both in prophylaxis and therapeutic models. The IFN-γ production and the cytotoxicity of tumor-specific CD8(+) T cells were significantly enhanced after immunization with tumor cells modified with LX/(IL-7) in both models. Although the tumor-infiltrating CD4(+) T cells and CD8(+) T cells were both increased and their IFN-γ productions also were upregulated, the antitumor activity of the tumor vaccine modified with LX/(IL-7) was dependent on CD8(+) T cells. Our results demonstrated that the autologous tumor vaccine modified with NDV strain LX/(IL-7) could promote the antitumor immune responses mediated by CD8(+) T cells and significantly improve the efficacy of the ATV-NDV.
Collapse
Affiliation(s)
- Lixiang Zhao
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yu Mei
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Qing Sun
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; and
| | - Linghua Guo
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yan Wu
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Xiao Yu
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Bo Hu
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; and
| | - Haiyan Liu
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China; Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
82
|
Fung TS, Liu DX. Coronavirus infection, ER stress, apoptosis and innate immunity. Front Microbiol 2014; 5:296. [PMID: 24987391 PMCID: PMC4060729 DOI: 10.3389/fmicb.2014.00296] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/29/2014] [Indexed: 12/27/2022] Open
Abstract
The replication of coronavirus, a family of important animal and human pathogens, is closely associated with the cellular membrane compartments, especially the endoplasmic reticulum (ER). Coronavirus infection of cultured cells was previously shown to cause ER stress and induce the unfolded protein response (UPR), a process that aims to restore the ER homeostasis by global translation shutdown and increasing the ER folding capacity. However, under prolonged ER stress, UPR can also induce apoptotic cell death. Accumulating evidence from recent studies has shown that induction of ER stress and UPR may constitute a major aspect of coronavirus–host interaction. Activation of the three branches of UPR modulates a wide variety of signaling pathways, such as mitogen-activated protein (MAP) kinase activation, autophagy, apoptosis, and innate immune response. ER stress and UPR activation may therefore contribute significantly to the viral replication and pathogenesis during coronavirus infection. In this review, we summarize the current knowledge on coronavirus-induced ER stress and UPR activation, with emphasis on their cross-talking to apoptotic signaling.
Collapse
Affiliation(s)
- To S Fung
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore
| | - Ding X Liu
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore
| |
Collapse
|
83
|
Takahashi T, Zenno S, Ishibashi O, Takizawa T, Saigo K, Ui-Tei K. Interactions between the non-seed region of siRNA and RNA-binding RLC/RISC proteins, Ago and TRBP, in mammalian cells. Nucleic Acids Res 2014; 42:5256-69. [PMID: 24561616 PMCID: PMC4005638 DOI: 10.1093/nar/gku153] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Small interfering RNA (siRNA)-based RNA interference (RNAi) is widely used for target gene silencing in various organisms. We previously showed that 8-nt-long 5′ proximal nucleotides, which include seed sequence (positions 2–8 from the 5′ end of guide strand), and the complementary sequence of the passenger strand are capable of being simultaneously replaced with cognate deoxyribonucleotides without any substantial loss of gene silencing. In the present study, examination was made of RNA requirements in the non-seed region of siRNA. The non-seed region of siRNA was found to be subdivided into four domains, in which two nucleotide pairs (positions 13 and 14) were replaceable with cognate deoxyribonucleotides without reducing RNAi activity. However, RNA sequences at positions 9-12 and 15-18 were essential for effective gene silencing, and these two double-stranded RNA cores are required for binding of the trans-activation response RNA-binding protein (TRBP). The terminal RNA (positions 19–21) provided Argonaute protein binding sites. Argonaute binding was enhanced by the presence of RNAs at positions 15–18. Knockdown experiments showed that, unlike Argonaute and TRBP, Dicer was dispensable for RNAi. Based on these observations, we discuss possible RNA/protein and protein/protein interactions in RNA-induced silencing complex formation.
Collapse
Affiliation(s)
- Tomoko Takahashi
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Department of Biotechnology, Faculty of Engineering, Maebashi Institute of Technology, 460-1 Kamisadori-cho, Maebashi-shi, Gunma 371-0816, Japan and Department of Molecular Medicine and Anatomy, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | | | | | | | | | | |
Collapse
|
84
|
Hughes T, Coffin RS, Lilley CE, Ponce R, Kaufman HL. Critical analysis of an oncolytic herpesvirus encoding granulocyte-macrophage colony stimulating factor for the treatment of malignant melanoma. Oncolytic Virother 2014; 3:11-20. [PMID: 27512660 PMCID: PMC4918360 DOI: 10.2147/ov.s36701] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Oncolytic viruses that selectively lyse tumor cells with minimal damage to normal cells are a new area of therapeutic development in oncology. An attenuated herpesvirus encoding the granulocyte-macrophage colony stimulating factor (GM-CSF), known as talimogene laherparepvec (T-VEC), has been identified as an attractive oncolytic virus for cancer therapy based on preclinical tumor studies and results from early-phase clinical trials and a large randomized Phase III study in melanoma. In this review, we discuss the basic biology of T-VEC, describe the role of GM-CSF as an immune adjuvant, summarize the preclinical data, and report the outcomes of published clinical trials using T-VEC. The emerging data suggest that T-VEC is a safe and potentially effective antitumor therapy in malignant melanoma and represents the first oncolytic virus to demonstrate therapeutic activity against human cancer in a randomized, controlled Phase III study.
Collapse
Affiliation(s)
- Tasha Hughes
- Departments of General Surgery and Immunology and Microbiology, Rush University Medical Center, Chicago IL, USA
| | - Robert S Coffin
- BioVex, Inc, a subsidiary of Amgen, Inc, Sherman Oaks, CA, USA
| | | | - Rafael Ponce
- BioVex, Inc, a subsidiary of Amgen, Inc, Sherman Oaks, CA, USA
| | - Howard L Kaufman
- Departments of General Surgery and Immunology and Microbiology, Rush University Medical Center, Chicago IL, USA
| |
Collapse
|
85
|
Sugimoto K, Kim SR, El-Shamy A, Imoto S, Ando K, Kim KI, Tanaka Y, Yano Y, Kim SK, Hasegawa Y, Fujinami A, Ohta M, Takashi H, Hotta H, Hayashi Y, Kudo M. Factors of response to pegylated interferon/ribavirin combination therapy and mechanism of viral clearance. Dig Dis 2013; 31:421-5. [PMID: 24281015 DOI: 10.1159/000355239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES This study explores viral factors of the interferon (IFN) and ribavirin (RBV) resistance-determining region (IRRDR), the IFN sensitivity-determining region (ISDR) and the core protein, and host factor interleukin 28B associated with response to pegylated IFN (PEG-IFN) and RBV combination therapy, and the correlation of viral and host factors with IFN-λ1. METHODS A total of 58 patients underwent PEG-IFN/RBV combination therapy for 48 weeks. The pretreatment factors associated with rapid virological response (RVR) and sustained virological response (SVR) were analyzed. Pretreatment IFN-λ1 serum levels were compared with the viral and host factors. RESULTS Univariate analysis showed that IRRDR ≥6 and ISDR ≥2 were significant pretreatment predictors of RVR, and multivariate analysis identified IRRDR ≥6 and hemoglobin as significant predictors of SVR. Pretreatment IFN-λ1 was significantly higher in the SVR group than in the non-SVR group and also in the IRRDR ≥6 group than in the IRRDR ≤5 group. CONCLUSIONS IRRDR ≥6 was the only significant predictor of SVR and was correlated with IFN-λ1. High serum levels of IFN-λ1 may be conducive to effective PEG-IFN/RBV combination therapy because of the immunomodulatory system.
Collapse
Affiliation(s)
- Kayo Sugimoto
- Department of Pharmacy, Kobe Asahi Hospital, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
da Silva LF, Jones C. Small non-coding RNAs encoded within the herpes simplex virus type 1 latency associated transcript (LAT) cooperate with the retinoic acid inducible gene I (RIG-I) to induce beta-interferon promoter activity and promote cell survival. Virus Res 2013; 175:101-9. [PMID: 23648811 PMCID: PMC4074922 DOI: 10.1016/j.virusres.2013.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/04/2013] [Accepted: 04/08/2013] [Indexed: 12/23/2022]
Abstract
The herpes simplex virus type 1 (HSV-1) latency-associated transcript (LAT) is abundantly expressed in latently infected trigeminal ganglionic sensory neurons. Expression of the first 1.5 kb of LAT coding sequences restores wild type reactivation to a LAT null HSV-1 mutant. The anti-apoptosis functions of the first 1.5 kb of LAT coding sequences are important for wild type levels of reactivation from latency. Two small non-coding RNAs (sncRNAs) contained within the first 1.5 kb of LAT coding sequences are expressed in trigeminal ganglia of latently infected mice, they cooperate to inhibit apoptosis, and reduce the efficiency of productive infection. In this study, we demonstrated that LAT sncRNA1 cooperates with the RNA sensor, retinoic acid inducible gene I (RIG-I), to stimulate IFN-β promoter activity and NF-κB dependent transcription in human or mouse cells. LAT sncRNA2 stimulated RIG-I induction of NF-κB dependent transcription in mouse neuroblastoma cells (Neuro-2A) but not human 293 cells. Since it is well established that NF-κB interferes with apoptosis, we tested whether the sncRNAs cooperated with RIG-I to inhibit apoptosis. In Neuro-2A cells, both sncRNAs cooperated with RIG-I to inhibit cold-shock induced apoptosis. Double stranded RNA (PolyI:C) stimulates RIG-I dependent signaling; but enhanced cold-shock induced apoptosis. PolyI:C, but not LAT sncRNAs, interfered with protein synthesis when cotransfected with RIG-I, which correlated with increased levels of cold-shock induced apoptosis. LAT sncRNA1 appeared to interact with RIG-I in transiently transfected cells suggesting this interaction stimulates RIG-I.
Collapse
Affiliation(s)
- Leticia Frizzo da Silva
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0900, United States
- Morisson Life Science Center, RM234 Lincoln, NE 68583-0900, United States
| | - Clinton Jones
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0900, United States
- Morisson Life Science Center, RM234 Lincoln, NE 68583-0900, United States
| |
Collapse
|
87
|
Watanabe T, Hiasa Y, Tokumoto Y, Hirooka M, Abe M, Ikeda Y, Matsuura B, Chung RT, Onji M. Protein kinase R modulates c-Fos and c-Jun signaling to promote proliferation of hepatocellular carcinoma with hepatitis C virus infection. PLoS One 2013; 8:e67750. [PMID: 23844083 PMCID: PMC3699507 DOI: 10.1371/journal.pone.0067750] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/22/2013] [Indexed: 12/22/2022] Open
Abstract
Double-stranded RNA-activated protein kinase R (PKR) is known to be upregulated by hepatitis C virus (HCV) and overexpressed in hepatocellular carcinoma (HCC). However, the precise roles of PKR in HCC with HCV infection remain unclear. Two HCV replicating cell lines (JFH-1 and H77s), generated by transfection of Huh7.5.1 cells, were used for experiments reported here. PKR expression was modulated with siRNA and a PKR expression plasmid, and cancer-related genes were assessed by real-time PCR and Western blotting; cell lines were further analyzed using a proliferation assay. Modulation of genes by PKR was also assessed in 34 human HCC specimens. Parallel changes in c-Fos and c-Jun gene expression with PKR were observed. Levels of phosphorylated c-Fos and c-Jun were upregulated by an increase of PKR, and were related to levels of phosphorylated JNK1 and Erk1/2. DNA binding activities of c-Fos and c-Jun also correlated with PKR expression, and cell proliferation was dependent on PKR-modulated c-Fos and c-Jun expression. Coordinate expression of c-Jun and PKR was confirmed in human HCC specimens with HCV infection. PKR upregulated c-Fos and c-Jun activities through activation of Erk1/2 and JNK1, respectively. These modulations resulted in HCC cell proliferation with HCV infection. These findings suggest that PKR-related proliferation pathways could be an attractive therapeutic target.
Collapse
Affiliation(s)
- Takao Watanabe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
- * E-mail:
| | - Yoshio Tokumoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masanori Abe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yoshio Ikeda
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Bunzo Matsuura
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Raymond T. Chung
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Morikazu Onji
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
88
|
Fournier P, Schirrmacher V. Oncolytic Newcastle Disease Virus as Cutting Edge between Tumor and Host. BIOLOGY 2013; 2:936-75. [PMID: 24833054 PMCID: PMC3960873 DOI: 10.3390/biology2030936] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/11/2013] [Accepted: 06/18/2013] [Indexed: 12/17/2022]
Abstract
Oncolytic viruses (OVs) replicate selectively in tumor cells and exert anti-tumor cytotoxic activity. Among them, Newcastle Disease Virus (NDV), a bird RNA virus of the paramyxovirus family, appears outstanding. Its anti-tumor effect is based on: (i) oncolytic activity and (ii) immunostimulation. Together these activities facilitate the induction of post-oncolytic adaptive immunity. We will present milestones during the last 60 years of clinical evaluation of this virus. Two main strategies of clinical application were followed using the virus (i) as a virotherapeutic agent, which is applied systemically or (ii) as an immunostimulatory agent combined with tumor cells for vaccination of cancer patients. More recently, a third strategy evolved. It combines the strategies (i) and (ii) and includes also dendritic cells (DCs). The first step involves systemic application of NDV to condition the patient. The second step involves intradermal application of a special DC vaccine pulsed with viral oncolysate. This strategy, called NDV/DC, combines anti-cancer activity (oncolytic virotherapy) and immune-stimulatory properties (oncolytic immunotherapy) with the high potential of DCs (DC therapy) to prime naive T cells. The aim of such treatment is to first prepare the cancer-bearing host for immunocompetence and then to instruct the patient's immune system with information about tumor-associated antigens (TAAs) of its own tumor together with danger signals derived from virus infection. This multimodal concept should optimize the generation of strong polyclonal T cell reactivity targeted against the patient's TAAs and lead to the establishment of a long-lasting memory T cell repertoire.
Collapse
Affiliation(s)
- Philippe Fournier
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Volker Schirrmacher
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
89
|
Zhou P, Cowled C, Wang LF, Baker ML. Bat Mx1 and Oas1, but not Pkr are highly induced by bat interferon and viral infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 40:240-247. [PMID: 23541614 DOI: 10.1016/j.dci.2013.03.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/10/2013] [Accepted: 03/12/2013] [Indexed: 06/02/2023]
Abstract
Bats harbour many emerging and re-emerging viruses, several of which are highly pathogenic in other mammals but cause no diseases in bats. As the interferon (IFN) response represents a first line of defence against viral infection, the ability of bats to control viral replication may be linked to the activation of the IFN system. The three most studied antiviral IFN-stimulated genes (ISGs) in other mammals; Pkr, Mx1 and Oas1 were examined in our model bat species, Pteropus alecto. Our results demonstrate that the three ISGs from P. alecto are highly conserved in their functional domains and promoter elements compared to corresponding genes from other mammals. However, P. alecto Oas1 contains two IFN-stimulated response elements (ISRE) in its promoter region compared with the single ISRE present in human OAS1 which may lead to higher IFN inducibility of the bat gene. Both Oas1 and Mx1 were induced in a highly IFN-dependent manner following stimulation with IFN or synthetic double-strand RNA (dsRNA) whereas Pkr showed evidence of being induced in an IFN-independent manner. Furthermore, bat Oas1 appeared to be the most inducible of the three ISGs following either IFN stimulation or viral infection, providing evidence that Oas1 may play a more important role in antiviral activity in bats compared with Mx1 or Pkr. Our results have important implications for the different roles of ISGs in bats and provide the first step in understanding the role of these molecules in the ability of bats to coexist with viruses.
Collapse
Affiliation(s)
- Peng Zhou
- CSIRO Australian Animal Health Laboratory, Geelong, Victoria 3220, Australia
| | | | | | | |
Collapse
|
90
|
Liu ZY, Jia KT, Li C, Weng SP, Guo CJ, He JG. A truncated Danio rerio PKZ isoform functionally interacts with eIF2α and inhibits protein synthesis. Gene 2013; 527:292-300. [PMID: 23742890 DOI: 10.1016/j.gene.2013.05.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 04/15/2013] [Accepted: 05/06/2013] [Indexed: 10/26/2022]
Abstract
A protein kinase containing Z-DNA binding domains (PKZ), which resembles protein kinase R (PKR) in domain organization, was recently discovered to be a member of the eIF2α kinase family in fish. PKR has roles in antiviral immunity through inhibiting protein synthesis and activating NF-κB; therefore, it is thought that PKZ may have a similar role in fish antiviral immunity. In the present study, the roles of two Danio rerio PKZ isoforms (DrPKZ-A and DrPKZ-B) in eIF2α phosphorylation and protein synthesis regulation were explored. DrPKZ-A and DrPKZ-B possess N-terminal Z-DNA binding domains and a conserved eIF2α kinase domain; however, they have domains of differing lengths inserted between kinase subdomains IV and V. DrPKZ-A has an insert domain of 73 amino acids (aa), whereas DrPKZ-B has an insert sequence of only 10 aa, suggesting that DrPKZ-B could be a dysfunctional isoform or could interact with different substrates. Our results show that both DrPKZ-A and DrPKZ-B functionally interact with eIF2α and inhibit protein synthesis, although DrPKZ-B possesses attenuated kinase activity. Our results also show that deletion of the insert in either isoform results in the complete abrogation of kinase activity, suggesting that the insert is critical for PKZ kinase activity. Kinase activity appears to be independent of insert length but may depend on the presence of specific amino acids within the insert domain. Furthermore, the effects of the N-terminal regulatory domain on kinase activity were analyzed. Deletion of the N-terminus results in reduced kinase activity of these isoforms relative to the wild-type forms, indicating that the isolated kinase domain is sufficient for eIF2α phosphorylation and that DrPKZ-A and DrPKZ-B may be regulated in a similar manner. Overall, our results show that DrPKZ-B is a functional kinase in zebrafish and contribute to our understanding of the function of PKZ in fish.
Collapse
Affiliation(s)
- Zhao-Yu Liu
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, PR China
| | | | | | | | | | | |
Collapse
|
91
|
Duwi Fanata WI, Lee SY, Lee KO. The unfolded protein response in plants: a fundamental adaptive cellular response to internal and external stresses. J Proteomics 2013; 93:356-68. [PMID: 23624343 DOI: 10.1016/j.jprot.2013.04.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/07/2013] [Accepted: 04/10/2013] [Indexed: 10/26/2022]
Abstract
In eukaryotic cells, proteins that enter the secretory pathway are translated on membrane-bound ribosomes and translocated into the endoplasmic reticulum (ER), where they are subjected to chaperone-assisted folding, post-translational modification and assembly. During the evolution of the eukaryotic cell, a homeostatic mechanism was developed to maintain the functions of the ER in the face of various internal and external stresses. The most severe stresses imposed on eukaryotic cells can induce ER stress that can overwhelm the processing capacity of the ER, leading to the accumulation of unfolded proteins in the ER lumen. To cope with this accumulation of unfolded proteins, the unfolded protein response (UPR) is activated to alter transcriptional programs through inositol-requiring enzyme 1 (IRE1) and bZIP17/28 in plants. In addition to transcriptional induction of UPR genes, quality control (QC), translational attenuation, ER-associated degradation (ERAD) and ER stress-induced apoptosis are also conserved as fundamental adaptive cellular responses to ER stress in plants. This article is part of a Special Issue entitled: Translational Plant Proteomics.
Collapse
Affiliation(s)
- Wahyu Indra Duwi Fanata
- Division of Applied Life Science (BK21 Program) and PMBBRC, Gyeongsang National University, 501 Jinju-daero, Jinju, 660-701, Republic of Korea
| | | | | |
Collapse
|
92
|
Transcriptional and post-translational regulation of mouse cation transport regulator homolog 1. Mol Cell Biochem 2013; 380:97-106. [DOI: 10.1007/s11010-013-1663-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/17/2013] [Indexed: 01/12/2023]
|
93
|
Handy I, Patel RC. STAT1 requirement for PKR-induced cell cycle arrest in vascular smooth muscle cells in response to heparin. Gene 2013; 524:15-21. [PMID: 23597922 DOI: 10.1016/j.gene.2013.03.124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 03/18/2013] [Accepted: 03/27/2013] [Indexed: 11/27/2022]
Abstract
Interferons (IFNs) are a family of cytokines that exhibit antiviral, antiproliferative, and immunomodulatory properties. PKR (protein kinase, RNA activated) is of central importance in mediating the antiproliferative actions of IFNs. Our research has established that PKR inhibits vascular smooth muscle cell (VSMC) proliferation by regulating G1 to S transition. Many cardiovascular diseases result from complications of atherosclerosis, a chronic and progressive inflammatory condition often characterized by excessive proliferation of VSMC. Thus, an effective method for inhibiting VSMC proliferation is likely to arrest atherosclerosis and restenosis at early stages. Our research establishes that PKR activation in VSMC leads to a G1 arrest brought about by an inhibition of cyclin-dependent kinase 2 (Cdk2) activity by p27(kip1). In quiescent VSMC, p27(kip1) levels are high and when stimulated by serum/growth factors, p27(kip1) levels drop by destabilization of the protein. Under conditions that lead to activation of PKR, there is a marked inhibition of p27(kip1) down-regulation due to increased stability of p27(kip1) protein. In order to understand the mechanism of heparin-induced stabilization of p27(kip1) in VSMC, we examined the involvement of the Signal Transducer and Activator of Transcription-1 (STAT1), which is an important player in mediating antiproliferative effects of IFNs. Our results demonstrate that PKR overexpression in VSMC leads to an increase in p27(kip1) protein levels and this increase requires the catalytic activity of PKR. PKR activation induced by antiproliferative agent heparin leads to phosphorylation of STAT1 on serine 727, which is essential for the cell cycle block. STAT1 null VSMCs are largely defective in heparin-induced cell cycle arrest and in PKR null cells the STAT1 phosphorylation in response to heparin was absent. These results establish that heparin causes STAT1 phosphorylation on serine 727 via activation of PKR and that this event is required for the G1 arrest in VSMC.
Collapse
Affiliation(s)
- Indhira Handy
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208,USA
| | | |
Collapse
|
94
|
Disease-causing allele-specific silencing by RNA interference. Pharmaceuticals (Basel) 2013; 6:522-35. [PMID: 24276122 PMCID: PMC3816697 DOI: 10.3390/ph6040522] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/28/2013] [Accepted: 04/02/2013] [Indexed: 12/19/2022] Open
Abstract
Small double-stranded RNAs (dsRNAs) of approximately 21-nucleotides in size, referred to as small interfering RNA (siRNA) duplexes, can induce sequence-specific posttranscriptional gene silencing, or RNA interference (RNAi). Since chemically synthesized siRNA duplexes were found to induce RNAi in mammalian cells, RNAi has become a powerful reverse genetic tool for suppressing the expression of a gene of interest in mammals, including human, and its application has been expanding to various fields. Recent studies further suggest that synthetic siRNA duplexes have the potential for specifically inhibiting the expression of an allele of interest without suppressing the expression of other alleles, i.e., siRNA duplexes likely confer allele-specific silencing. Such gene silencing by RNAi is an advanced technique with very promising applications. In this review, I would like to discuss the potential utility of allele-specific silencing by RNAi as a therapeutic method for dominantly inherited diseases, and describe possible improvements in siRNA duplexes for enhancing their efficacy.
Collapse
|
95
|
Gibbert K, Schlaak JF, Yang D, Dittmer U. IFN-α subtypes: distinct biological activities in anti-viral therapy. Br J Pharmacol 2013; 168:1048-58. [PMID: 23072338 PMCID: PMC3594665 DOI: 10.1111/bph.12010] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 08/15/2012] [Accepted: 09/07/2012] [Indexed: 12/12/2022] Open
Abstract
During most viral infections, the immediate host response is characterized by an induction of type I IFN. These cytokines have various biological activities, including anti-viral, anti-proliferative and immunomodulatory effects. After induction, they bind to their IFN-α/β receptor, which leads to downstream signalling resulting in the expression of numerous different IFN-stimulated genes. These genes encode anti-viral proteins that directly inhibit viral replication as well as modulate immune function. Thus, the induction of type I IFN is a very powerful tool for the host to fight virus infections. Many viruses evade this response by various strategies like the direct suppression of IFN induction or inhibition of the IFN signalling pathway. Therefore, the therapeutic application of exogenous type I IFN or molecules that induce strong IFN responses should be of great potential for future immunotherapies against viral infections. Type I IFN is currently used as a treatment in chronic hepatitis B and C virus infection, but as yet is not widely utilized for other viral infections. One reason for this restricted clinical use is that type I IFN belongs to a multigene family that includes 13 different IFN-α subtypes and IFN-β, whose individual anti-viral and immunomodulatory properties have so far not been investigated in detail to improve IFN therapy against viral infections in humans. In this review, we summarize the recent achievements in defining the distinct biological functions of type I IFN subtypes in cell culture and in animal models of viral infection as well as their clinical usage in chronic hepatitis virus infections.
Collapse
Affiliation(s)
- K Gibbert
- Department of Virology, University Hospital Essen, Essen, Germany.
| | | | | | | |
Collapse
|
96
|
Brenner C, Simmonds RE, Wood S, Rose V, Feldmann M, Turner J. TLR signalling and adapter utilization in primary human in vitro differentiated adipocytes. Scand J Immunol 2012; 76:359-70. [PMID: 22690903 DOI: 10.1111/j.1365-3083.2012.02744.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Toll-like receptors (TLRs) are central to innate immunity and yet their expression is widespread and not restricted to professional inflammatory cells. TLRs have been reported on adipocytes and have been implicated in obesity-associated pathologies such as diabetes. Why TLRs are found on adipocytes is not clear although one hypothesis is that they may coordinate energy utilization for the energy intensive process of an immune response. We have explored TLR signalling in primary human in vitro differentiated adipocytes and investigated the specific adapter molecules that are involved. Only lipopolysaccharide (LPS), poly(I:C), Pam3CSK4 and MALP-2 could induce the production of IL-6, IL-8 and MCP-1 by adipocytes. Poly(I:C) alone caused a strong induction of type I interferons, as assessed by IP-10 production. Using siRNA, it was confirmed that LPS-dependent signalling in adipocytes occurs via TLR4 utilizing the adapter molecules MyD88, Mal and TRIF and caused rapid degradation of IκBα. Pam3CSK4 signalling utilized TLR2, MyD88 and Mal (but not TRIF). However, the response to poly(I:C) observed in these cells appeared not to require TRIF, but MyD88 was required for induction of NFκB-dependent cytokines by Poly(I:C). Despite this, IκBα degradation could not be detected in poly(I:C) stimulated adipocytes at any time-point up to 4 h. Indeed, IL-6 transcription was not induced until 8-16 h after exposure. These data suggest that Pam3CSK4 and LPS signal via the expected routes in human adipocytes, whereas poly(I:C)/TLR3 signalling may act via a TRIF-independent, MyD88-dependent route.
Collapse
Affiliation(s)
- C Brenner
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, London, UK
| | | | | | | | | | | |
Collapse
|
97
|
Gentz SHL, Bertollo CM, Souza-Fagundes EM, da Silva AM. Implication of eIF2α kinase GCN2 in induction of apoptosis and endoplasmic reticulum stress-responsive genes by sodium salicylate. ACTA ACUST UNITED AC 2012; 65:430-40. [PMID: 23356852 DOI: 10.1111/jphp.12002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 10/11/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Sodium salicylate (NaSal) can disturb cell viability by affecting the activity of multiple cellular molecules. In this work, we investigated the involvement of stress-responsive kinase GCN2 in regulating cell death and expression of stress genes in mouse embryonic fibroblasts (MEFs) upon exposure to NaSal. METHODS Cell viability was assayed using the 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide (MTT) method, and apoptosis was evaluated by annexin V and propidium iodide staining. A polymerase chain reaction (PCR) array approach was used to analyse differential expression of a panel of 84 endoplasmic reticulum (ER) stress-associated genes. Gene reporter assays were carried out to determine activity of ER stress element (ERSE), and the protein levels of activating transcription factor 6 (ATF6) and C/EBP homologous protein (CHOP) were determined by western blot. KEY FINDINGS NaSal treatment resulted in reduction of cellular viability and induction of apoptosis in wild-type but not Gcn2(-/-) cells. Many genes with important functions in protein synthesis/degradation, transcriptional regulation and apoptosis were induced by NaSal and most of these were dependent on GCN2. The activation of ERSE within Ddit3 and the production of CHOP and ATF6 induced by NaSal required GCN2. CONCLUSIONS Our data provide evidence for the involvement of GCN2 in apoptosis and gene expression triggered by NaSal, and contributes to the understanding of molecular events occurring in NaSal-treated cells.
Collapse
Affiliation(s)
- Solange H L Gentz
- Laboratory of Inflammatory Genes, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | | | | |
Collapse
|
98
|
Gullerova M, Proudfoot NJ. Convergent transcription induces transcriptional gene silencing in fission yeast and mammalian cells. Nat Struct Mol Biol 2012; 19:1193-201. [PMID: 23022730 PMCID: PMC3504457 DOI: 10.1038/nsmb.2392] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 08/27/2012] [Indexed: 01/03/2023]
Abstract
We show that convergent transcription induces transcriptional gene silencing (TGS) in trans for both fission yeast and mammalian cells. This method has advantages over existing strategies to induce gene silencing. Previous studies in fission yeast have characterized TGS as a cis-specific process involving RNA interference that maintains heterochromatic regions such as centromeres. In contrast, in mammalian cells, gene silencing is known to occur through a post-transcriptional mechanism that uses exogenous short interfering RNAs or endogenous microRNAs to inactivate mRNA. We now show that the introduction of convergent transcription plasmids into either Schizosaccharomyces pombe or mammalian cells allows the production of double-stranded RNA from inserted gene fragments, resulting in TGS of endogenous genes. We predict that using convergent transcription to induce gene silencing will be a generally useful strategy and allow for a fuller molecular understanding of the biology of TGS.
Collapse
Affiliation(s)
- Monika Gullerova
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | | |
Collapse
|
99
|
Cell-specific regulation of nucleic acid sensor cascades: a controlling interest in the antiviral response. J Virol 2012; 86:13303-12. [PMID: 23015711 DOI: 10.1128/jvi.02296-12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In this study, we examined the capacities of non-antigen-presenting cell types to propagate antiviral signals following infection with recombinant adenovirus or by direct nucleic acid transfection. Three murine cell lines (RAW264.7 macrophages as a positive control, FL83B hepatocytes, and MS1 endothelial cells) were assessed following exposure to adenovirus, DNA, or RNA ligands. Based on primary (interferon response factor 3 [IRF3] phosphorylation) and secondary (STAT1/2 phosphorylation) response markers, we found each cell line presented a unique response profile: RAW cells were highly responsive, MS1 cells were modified in their response, and FL83B cells were essentially nonresponsive. Comparative reverse transcription-quantitative PCR (RT-qPCR) of nucleic acid sensing components revealed major differences between the three cell types. A prominent difference was at the level of adaptor molecules; TRIF, MyD88, MAVS, and STING. TRIF was absent in MS1 and FL83B cells, whereas MyD88 levels were diminished in FL83B hepatocytes. These differences resulted in compromised TLR-mediated activation. While the cytosolic adaptor MAVS was well represented in all cell lines, the DNA adaptor STING was deficient in FL83B hepatocytes (down by nearly 3 log units). The absence of STING provides an explanation for the lack of DNA responsiveness in these cells. This hypothesis was confirmed by acquisition of IRF3 activation in Flag-STING FL83B cells following DNA transfection. To consolidate the central role of adaptors in MS1 endothelial cells, short hairpin RNA (shRNA) knockdown of STING and MAVS resulted in a ligand-specific loss of IRF3 responsiveness. In contrast to the requirement for specific adaptor proteins, a requirement for a specific DNA sensor (AIM2, DDx41, or p204) in the IRF3 activation response was not detected by shRNA knockdown in MS1 cells. The data reveal that cell-specific regulation of nucleic acid sensing cascade components influences antiviral recognition responses, that controlling levels of adaptor molecules is a recurring strategy in regulating antiviral recognition response functions, and that comparative RT-qPCR has predictive value for antiviral/innate response functions in these cells.
Collapse
|
100
|
Shi B, Ren G, Hu Y, Wang S, Zhang Z, Yuan Z. HBsAg inhibits IFN-α production in plasmacytoid dendritic cells through TNF-α and IL-10 induction in monocytes. PLoS One 2012; 7:e44900. [PMID: 23024774 PMCID: PMC3443230 DOI: 10.1371/journal.pone.0044900] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 08/14/2012] [Indexed: 01/04/2023] Open
Abstract
Type I Interferon (IFN) is one of the first lines of defense against viral infection. Plasmacytoid dendritic cells (pDCs) are professional IFN-α-producing cells that play an important role in the antiviral immune response. Previous studies have reported that IFN-α production is impaired in chronic hepatitis B (CHB) patients. However, the mechanisms underlying the impairment in IFN-α production are not fully understood. Here, we report that plasma-derived hepatitis B surface antigen (HBsAg) and HBsAg expressed in CHO cells can significantly inhibit toll like receptor (TLR) 9-mediated Interferon-α (IFN-α) production in peripheral blood mononuclear cells (PBMCs) from healthy donors. Further analysis indicated that monocytes participate in the inhibitory effect of HBsAg on pDCs through the secretion of TNF-α and IL-10. Furthermore, TLR9 expression on pDCs was down-regulated by TNF-α, IL-10 and HBsAg treatment. This down-regulation may partially explain the inhibition of IFN-α production in pDCs. In conclusion, we determined that HBsAg inhibited the production of IFN-α by pDCs through the induction of monocytes that secreted TNF-α and IL-10 and through the down-regulation of TLR9 expression on pDCs. These data may aid in the development of effective antiviral treatments and lead to the immune control of the viral infections.
Collapse
Affiliation(s)
- Bisheng Shi
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Guangxu Ren
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
- Laboratory of Molecular Virology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Yunwen Hu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Sen Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Zhanqing Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Zhenghong Yuan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
- Laboratory of Molecular Virology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- * E-mail:
| |
Collapse
|