1
|
Li S, Xie Y, Yu C, Zheng C, Xu Z. The battle between host antiviral innate immunity and immune evasion by cytomegalovirus. Cell Mol Life Sci 2024; 81:341. [PMID: 39120730 PMCID: PMC11335264 DOI: 10.1007/s00018-024-05369-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Cytomegalovirus (CMV) has successfully established a long-lasting latent infection in humans due to its ability to counteract the host antiviral innate immune response. During coevolution with the host, the virus has evolved various evasion techniques to evade the host's innate immune surveillance. At present, there is still no vaccine available for the prevention and treatment of CMV infection, and the interaction between CMV infection and host antiviral innate immunity is still not well understood. However, ongoing studies will offer new insights into how to treat and prevent CMV infection and its related diseases. Here, we update recent studies on how CMV evades antiviral innate immunity, with a focus on how CMV proteins target and disrupt critical adaptors of antiviral innate immune signaling pathways. This review also discusses some classic intrinsic cellular defences that are crucial to the fight against viral invasion. A comprehensive review of the evasion mechanisms of antiviral innate immunity by CMV will help investigators identify new therapeutic targets and develop vaccines against CMV infection.
Collapse
Affiliation(s)
- Shuang Li
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuanyang Xie
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
Effects of Bacillus subtilis Natto Strains on Antiviral Responses in Resiquimod-Stimulated Human M1-Phenotype Macrophages. Foods 2023; 12:foods12020313. [PMID: 36673407 PMCID: PMC9858497 DOI: 10.3390/foods12020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Bacillus subtilis natto is used in the production of natto, a traditional fermented soy food, and has beneficial immunomodulatory effects in humans. Single-stranded RNA (ssRNA) viruses, including influenza and coronavirus, often cause global pandemics. We proposed a human cell culture model mimicking ssRNA viral infection and investigated the ability of B. subtilis natto to induce antiviral effects in the model. The gene expressions were analyzed using quantitative real-time reverse transcription PCR. M1-phenotype macrophages derived from THP-1 cells strongly express the Toll-like receptor 8 (76.2-hold), CD80 (64.2-hold), and CCR7 (45.7-hold) mRNA compared to M0 macrophages. One µg/mL of resiquimod (RSQ)-stimulation induced the expression of IRF3 (1.9-hold), CXCL10 (14.5-hold), IFNβ1 (3.5-hold), ISG20 (4.4-hold), and MxA (1.7-hold) mRNA in the M1-phenotype macrophages. Based on these results, the RSQ-stimulated M1-phenotype macrophages were used as a cell culture model mimicking ssRNA viral infection. Moreover, the B. subtilis natto XF36 strain induced the expression of genes associated with antiviral activities (IFNβ1, IFNλ1, ISG20, and RNase L) and anti-inflammatory activities (IL-10) in the cell culture model. Thus, it is suggested that the XF36 suppresses viral infections and excessive inflammation by inducing the expression of genes involved in antiviral and anti-inflammatory activities.
Collapse
|
3
|
Lee MF, Voon GZ, Lim HX, Chua ML, Poh CL. Innate and adaptive immune evasion by dengue virus. Front Cell Infect Microbiol 2022; 12:1004608. [PMID: 36189361 PMCID: PMC9523788 DOI: 10.3389/fcimb.2022.1004608] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/26/2022] [Indexed: 11/26/2022] Open
Abstract
Dengue is a mosquito-borne disease which causes significant public health concerns in tropical and subtropical countries. Dengue virus (DENV) has evolved various strategies to manipulate the innate immune responses of the host such as ‘hiding’ in the ultrastructure of the host, interfering with the signaling pathway through RNA modifications, inhibiting type 1 IFN production, as well as inhibiting STAT1 phosphorylation. DENV is also able to evade the adaptive immune responses of the host through antigenic variation, antigen-dependent enhancement (ADE), partial maturation of prM proteins, and inhibition of antigen presentation. miRNAs are important regulators of both innate and adaptive immunity and they have been shown to play important roles in DENV replication and pathogenesis. This makes them suitable candidates for the development of anti-dengue therapeutics. This review discusses the various strategies employed by DENV to evade innate and adaptive immunity. The role of miRNAs and DENV non-structural proteins (NS) are promising targets for the development of anti-dengue therapeutics.
Collapse
|
4
|
Sui N, Zhang R, Jiang Y, Yu H, Xu G, Wang J, Zhu Y, Xie Z, Hu J, Jiang S. Long Noncoding RNA Expression Rofiles Elucidate the Potential Roles of lncRNA- XR_003496198 in Duck Hepatitis A Virus Type 1 Infection. Front Cell Infect Microbiol 2022; 12:858537. [PMID: 35531338 PMCID: PMC9074814 DOI: 10.3389/fcimb.2022.858537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/24/2022] [Indexed: 12/11/2022] Open
Abstract
Duck hepatitis A virus type 1 (DHAV-1) is a highly lethal virus that severely affects the duck industry worldwide. Long noncoding RNAs (lncRNAs) exert crucial roles in pathogen attacks. Here, we conducted deep transcriptome analysis to investigate the dynamic changes of host lncRNAs profiles in DHAV-1-infected duck embryo fibroblasts. We identified 16,589 lncRNAs in total and characterized their genomic features. Moreover, 772 and 616 differentially expressed lncRNAs (DELs) were screened at 12 and 24 h post-infection. Additionally, we predicted the DELs' cis- and trans-target genes and constructed lncRNA-target genes regulatory networks. Functional annotation analyses indicated that the putative target genes of DELs participated in diverse vital biological processed, including immune responses, cellular metabolism, and autophagy. For example, we confirmed the dysregulation of pattern recognition receptors (TLR3, RIG-I, MDA5, LGP2, cGAS), signal transducers (STAT1), transcription factors (IRF7), immune response mediators (IL6, IL10, TRIM25, TRIM35, TRIM60, IFITM1, IFITM3, IFITM5), and autophagy-related genes (ULK1, ULK2, EIF4EBP2) using RT-qPCR. Finally, we confirmed that one DHAV-1 induced lncRNA-XR_003496198 is likely to inhibit DHAV-1 replication in DEFs. Our study comprehensively analyzed the lncRNA profiles upon DHAV-1 infection and screened the target genes involved in the innate immune response and autophagy signaling pathway, thereby revealing the essential roles of duck lncRNAs and broadening our understanding of host-virus interactions.
Collapse
Affiliation(s)
- Nana Sui
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Ruihua Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Yue Jiang
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Honglei Yu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Guige Xu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Jingyu Wang
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Yanli Zhu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Zhijing Xie
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| | - Jiaqing Hu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
- Shandong GreenBlue Biotechnology Co. Ltd., Taian, China
| | - Shijin Jiang
- College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, China
| |
Collapse
|
5
|
Valenzuela B, Benavides A, Gutierrez D, Blamey L, Monsalves MT, Modak B, Blamey JM. Violacein from an Antarctic Iodobacter sp. 7MAnt and its function as immunomodulator of the defence mechanism of innate immunity in fish cells. JOURNAL OF FISH DISEASES 2022; 45:485-489. [PMID: 34850980 DOI: 10.1111/jfd.13559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 06/13/2023]
Affiliation(s)
- Beatriz Valenzuela
- Laboratory of Natural Products Chemistry, Centre of Aquatic Biotechnology, Department of Environmental Sciences, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago, Chile
| | - Almendra Benavides
- Laboratory of Natural Products Chemistry, Centre of Aquatic Biotechnology, Department of Environmental Sciences, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago, Chile
| | - Daniela Gutierrez
- Laboratory of Natural Products Chemistry, Centre of Aquatic Biotechnology, Department of Environmental Sciences, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago, Chile
| | | | | | - Brenda Modak
- Laboratory of Natural Products Chemistry, Centre of Aquatic Biotechnology, Department of Environmental Sciences, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago, Chile
| | - Jenny M Blamey
- Fundación Biociencia, Santiago, Chile
- Biology Department, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago, Chile
| |
Collapse
|
6
|
Sánchez-González MT, Cienfuegos-Jiménez O, Álvarez-Cuevas S, Pérez-Maya AA, Borrego-Soto G, Marino-Martínez IA. Prevalence of the SNP rs10774671 of the OAS1 gene in Mexico as a possible predisposing factor for RNA virus disease. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2021; 12:52-60. [PMID: 34336138 PMCID: PMC8310884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/31/2021] [Indexed: 06/13/2023]
Abstract
UNLABELLED The COVID-19 pandemic has revealed the susceptibility of certain populations to RNA virus infection. This variety of agents is currently the cause of severe respiratory diseases (SARS-CoV2 and Influenza), Hepatitis C, measles and of high prevalence tropical diseases that are detected throughout the year (Dengue and Zika). The rs10774671 polymorphism is a base change from G to A in the last nucleotide of intron-5 of the OAS1 gene. This change modifies a splicing site and generates isoforms of the OAS1 protein with a higher molecular weight and a demonstrated lower enzymatic activity. The low activity of these OAS1 isoforms makes the innate immune response against RNA virus infections less efficient, representing a previously unattended risk factor for certain populations. OBJECTIVE Determine the distribution of rs10774671 in the open population of Mexico. METHODS In 98 healthy volunteers, allelic and genotypic frequencies were determined by qPCR using allele specific labeled probes, and the Hardy-Weinberg equilibrium was determined. RESULTS The A-allele turned out to be the most prevalent in the analyzed population. CONCLUSIONS Our population is genetically susceptible to RNA virus disease due to the predominant presence of the A allele of rs10774671 in the OAS1 gene.
Collapse
Affiliation(s)
- María Teresa Sánchez-González
- Universidad Autónoma de Nuevo León, Centro de Investigación y Desarrollo en Ciencias de la SaludMonterrey, Nuevo León, México
| | - Oscar Cienfuegos-Jiménez
- Universidad Autónoma de Nuevo León, Centro de Investigación y Desarrollo en Ciencias de la SaludMonterrey, Nuevo León, México
| | - Salomón Álvarez-Cuevas
- Universidad Autónoma de Nuevo León, Departamento de Patología, Facultad de MedicinaMonterrey, Nuevo León, México
| | - Antonio Ali Pérez-Maya
- Universidad Autónoma de Nuevo León, Departamento de Bioquímica y Medicina Molecular, Facultad de MedicinaMonterrey, Nuevo León, México
| | - Gissela Borrego-Soto
- Department of Molecular Biosciences, University of Texas at AustinAustin, Texas, United States of America
| | - Iván Alberto Marino-Martínez
- Universidad Autónoma de Nuevo León, Centro de Investigación y Desarrollo en Ciencias de la SaludMonterrey, Nuevo León, México
- Universidad Autónoma de Nuevo León, Departamento de Patología, Facultad de MedicinaMonterrey, Nuevo León, México
| |
Collapse
|
7
|
Strumillo ST, Kartavykh D, de Carvalho FF, Cruz NC, de Souza Teodoro AC, Sobhie Diaz R, Curcio MF. Host-virus interaction and viral evasion. Cell Biol Int 2021; 45:1124-1147. [PMID: 33533523 PMCID: PMC8014853 DOI: 10.1002/cbin.11565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/24/2021] [Indexed: 12/12/2022]
Abstract
With each infectious pandemic or outbreak, the medical community feels the need to revisit basic concepts of immunology to understand and overcome the difficult times brought about by these infections. Regarding viruses, they have historically been responsible for many deaths, and such a peculiarity occurs because they are known to be obligate intracellular parasites that depend upon the host's cell machinery for their replication. Successful infection with the production of essential viral components requires constant viral evolution as a strategy to manipulate the cellular environment, including host internal factors, the host's nonspecific and adaptive immune responses to viruses, the metabolic and energetic state of the infected cell, and changes in the intracellular redox environment during the viral infection cycle. Based on this knowledge, it is fundamental to develop new therapeutic strategies for controlling viral dissemination, by means of antiviral therapies, vaccines, or antioxidants, or by targeting the inhibition or activation of cell signaling pathways or metabolic pathways that are altered during infection. The rapid recovery of altered cellular homeostasis during viral infection is still a major challenge. Here, we review the strategies by which viruses evade the host's immune response and potential tools used to develop more specific antiviral therapies to cure, control, or prevent viral diseases.
Collapse
Affiliation(s)
- Scheilla T Strumillo
- Department of Biochemistry, Laboratory of Cell Signaling, Federal University of São Paulo, São Paulo, Brazil
| | - Denis Kartavykh
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| | - Fábio F de Carvalho
- Departament of Educational Development, Getulio Vargas Foundation, São Paulo, Brazil
| | - Nicolly C Cruz
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| | - Ana C de Souza Teodoro
- Department of Biochemistry, Laboratory of Cell Signaling, Federal University of São Paulo, São Paulo, Brazil
| | - Ricardo Sobhie Diaz
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| | - Marli F Curcio
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Sohn KM, Lee SG, Kim HJ, Cheon S, Jeong H, Lee J, Kim IS, Silwal P, Kim YJ, Paik S, Chung C, Park C, Kim YS, Jo EK. COVID-19 Patients Upregulate Toll-like Receptor 4-mediated Inflammatory Signaling That Mimics Bacterial Sepsis. J Korean Med Sci 2020; 35:e343. [PMID: 32989935 PMCID: PMC7521960 DOI: 10.3346/jkms.2020.35.e343] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Observational studies of the ongoing coronavirus disease 2019 (COVID-19) outbreak suggest that a 'cytokine storm' is involved in the pathogenesis of severe illness. However, the molecular mechanisms underlying the altered pathological inflammation in COVID-19 are largely unknown. We report here that toll-like receptor (TLR) 4-mediated inflammatory signaling molecules are upregulated in peripheral blood mononuclear cells (PBMCs) from COVID-19 patients, compared with healthy controls (HC). METHODS A total of 48 subjects including 28 COVID-19 patients (8 severe/critical vs. 20 mild/moderate cases) admitted to Chungnam National University Hospital, and age/sex-matched 20 HC were enrolled in this study. PBMCs from the subjects were processed for nCounter Human Immunology gene expression assay to analyze the immune related transcriptome profiles. Recombinant proteins of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) were used to stimulate the PBMCs and monocyte-derived macrophages, and real-time polymerase chain reaction was performed to quantify the mRNA expressions of the pro-inflammatory cytokines/chemokines. RESULTS Among the most highly increased inflammatory mediators in severe/critically ill patients, S100A9, an alarmin and TLR4 ligand, was found as a noteworthy biomarker, because it inversely correlated with the serum albumin levels. We also observed that recombinant S2 and nucleocapsid proteins of SARS-CoV-2 significantly increased pro-inflammatory cytokines/chemokines and S100A9 in human primary PBMCs. CONCLUSION These data support a link between TLR4 signaling and pathological inflammation during COVID-19 and contribute to develop therapeutic approaches through targeting TLR4-mediated inflammation.
Collapse
Affiliation(s)
- Kyung Mok Sohn
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sung Gwon Lee
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Hyeon Ji Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Shinhyea Cheon
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyeongseok Jeong
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jooyeon Lee
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - In Soo Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Chaeuk Chung
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea.
| | - Yeon Sook Kim
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea.
| | - Eun Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea.
| |
Collapse
|
9
|
Ovejero T, Sadones O, Sánchez-Fito T, Almenar-Pérez E, Espejo JA, Martín-Martínez E, Nathanson L, Oltra E. Activation of Transposable Elements in Immune Cells of Fibromyalgia Patients. Int J Mol Sci 2020; 21:E1366. [PMID: 32085571 PMCID: PMC7072917 DOI: 10.3390/ijms21041366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/06/2020] [Accepted: 02/14/2020] [Indexed: 02/07/2023] Open
Abstract
Advancements in nucleic acid sequencing technology combined with an unprecedented availability of metadata have revealed that 45% of the human genome constituted by transposable elements (TEs) is not only transcriptionally active but also physiologically necessary. Dysregulation of TEs, including human retroviral endogenous sequences (HERVs) has been shown to associate with several neurologic and autoimmune diseases, including Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). However, no study has yet addressed whether abnormal expression of these sequences correlates with fibromyalgia (FM), a disease frequently comorbid with ME/CFS. The work presented here shows, for the first time, that, in fact, HERVs of the H, K and W types are overexpressed in immune cells of FM patients with or without comorbid ME/CFS. Patients with increased HERV expression (N = 14) presented increased levels of interferon (INF-β and INF-γ) but unchanged levels of TNF-α. The findings reported in this study could explain the flu-like symptoms FM patients present with in clinical practice, in the absence of concomitant infections. Future work aimed at identifying specific genomic loci differentially affected in FM and/or ME/CFS is warranted.
Collapse
Affiliation(s)
- Tamara Ovejero
- School of Medicine, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | | | - Teresa Sánchez-Fito
- Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, 46008 Valencia, Spain; (T.S.-F.); (E.A.-P.)
| | - Eloy Almenar-Pérez
- Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, 46008 Valencia, Spain; (T.S.-F.); (E.A.-P.)
| | - José Andrés Espejo
- School of Biotechnology, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | | | - Lubov Nathanson
- Institute for Neuro Immune Medicine, Nova Southeastern University, Ft Lauderdale, FL 33314, USA;
| | - Elisa Oltra
- School of Medicine, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
- Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain
| |
Collapse
|
10
|
Tang Y, Zhu X, Han R, Zhou Q, Cheng H. Expressionof langerhans cell and plasmacytoid dendritic cell markers, and toll-like receptor 7/9 signaling pathway proteins in verruca vulgaris lesions. Medicine (Baltimore) 2020; 99:e19214. [PMID: 32080113 PMCID: PMC7034720 DOI: 10.1097/md.0000000000019214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Langerhans cells (LCs) and plasmacytoid dendritic cells (pDCs) play an important role in the cutaneous immune response to viral infection. Verruca vulgaris (VV) is a chronic benign disease caused by human papillomavirus (HPV) infection.To investigate the possible roles of LCs, pDCs and toll-like receptor (TLR)7/9 signaling pathways in the pathogenesis of VV, we detected the expression of CD1a, CD2AP, CD123, TLR7/9, IFN regulatory factor 7 (IRF7), and interleukin-1 receptor-associated kinase 1 (IRAK1) in VV lesions.The expression of CD1a, CD2AP, CD123, TLR7/9, IRF7, and IRAK1 in 20 VV lesions was tested by immunohistochemistry. The density and number of stained cells were compared between VV lesions and the perilesional normal skin.The density and number of CD1a-, CD2AP-, CD123-, TLR9-, and IRAK1-positive cells in the papillary layer of VV lesions were significantly higher than those in the perilesional normal skin (P < .05). There were no significant differences in the density and positive rate of CD1a+ cells in the epidermis and of TLR7 and IRF7 cells in the dermis between VV lesions and the perilesional normal skin at the edge (P > .05).In VV, the number of LCs increases only in the dermis, indicating that LC's antigen-presenting function might not be inhibited. The increased number of pDCs in VV lesions suggests that HPV infection may recruit the pDCs to the virus-infected epithelium. We speculate that the TLR7/9 downstream signaling pathway is not fully activated in VV, leading to difficulty of HPV removal and the relapse of HPV-infected lesions.
Collapse
Affiliation(s)
- Yi Tang
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou
| | - Xiaoxia Zhu
- Department of Dermatology, Ningbo First Hospital, Ningbo, Zhejiang
| | - Rui Han
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiang Zhou
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hao Cheng
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Interferon-Stimulated Genes-Mediators of the Innate Immune Response during Canine Distemper Virus Infection. Int J Mol Sci 2019; 20:ijms20071620. [PMID: 30939763 PMCID: PMC6480560 DOI: 10.3390/ijms20071620] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022] Open
Abstract
The demyelinating canine distemper virus (CDV)-leukoencephalitis represents a translational animal model for multiple sclerosis. The present study investigated the expression of type I interferon (IFN-I) pathway members in CDV-induced cerebellar lesions to gain an insight into their role in lesion development. Gene expression of 110 manually selected genes in acute, subacute and chronic lesions was analyzed using pre-existing microarray data. Interferon regulatory factor (IRF) 3, IRF7, signal transducer and activator of transcription (STAT) 1, STAT2, MX protein, protein kinase R (PKR), 2'-5'-oligoadenylate synthetase (OAS) 1 and interferon-stimulated gene (ISG) 15 expression were also evaluated using immunohistochemistry. Cellular origin of STAT1, STAT2, MX and PKR were determined using immunofluorescence. CDV infection caused an increased expression of the antiviral effector proteins MX, PKR, OAS1 and ISG15, which probably contributed to a restricted viral replication, particularly in neurons and oligodendrocytes. This increase might be partly mediated by IRF-dependent pathways due to the lack of changes in IFN-I levels and absence of STAT2 in astrocytes. Nevertheless, activated microglia/macrophages showed a strong expression of STAT1, STAT2 and MX proteins in later stages of the disease, indicating a strong activation of the IFN-I signaling cascade, which might be involved in the aggravation of bystander demyelination.
Collapse
|
12
|
Jung YS, Chae D, Park K. Population PK-PD Model of Pegylated Interferon Alfa-2a in Healthy Korean Men. J Pharm Sci 2018; 107:3171-3178. [PMID: 30179597 DOI: 10.1016/j.xphs.2018.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 08/09/2018] [Accepted: 08/14/2018] [Indexed: 10/28/2022]
Abstract
Pegylated interferon alfa-2a (PEG-IFN alfa-2a), which was developed to overcome the disadvantages of conventional formulations, is widely prescribed for hepatitis B or C virus infection. It is characterized by pharmacokinetic (PK) and pharmacodynamic (PD) properties much different from those of conventional forms. The present study developed a population PK-PD model of subcutaneous PEG-IFN alfa-2a in a Korean population. For PK, IFN alfa-2a concentrations were described by a 1-compartment model with first-order absorption, preceded by skin-to-depot first-order input. For PD, serum 2'-5' oligoadenylsynthetase activity was described by an effect compartment model incorporating a tolerance compartment. The baseline serum 2'-5' oligoadenylsynthetase level was found to have an inverse relationship with sensitivity to tolerance, leading to high tolerance at low baseline. The model revealed that subjects with low baselines experienced time delay, while those with high baselines showed tolerance in their concentration-effect relationships. The developed models matched well with data and simulation results, and the model showed that the optimal dose decreases with the baseline, with no dose effective for a baseline >250 pmol/dL. Our results can serve as a basis for improving dosing regimens of PEG-IFN alfa-2a in adult patients with chronic hepatitis B or C infection.
Collapse
Affiliation(s)
- Yun Seob Jung
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Korea
| | - Dongwoo Chae
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Korea
| | - Kyungsoo Park
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
13
|
Vahedi F, Lee AJ, Collins SE, Chew MV, Lusty E, Chen B, Dubey A, Richards CD, Feld JJ, Russell RS, Mossman KL, Ashkar AA. IL-15 and IFN-γ signal through the ERK pathway to inhibit HCV replication, independent of type I IFN signaling. Cytokine 2018; 124:154439. [PMID: 29908921 DOI: 10.1016/j.cyto.2018.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 05/17/2018] [Accepted: 06/06/2018] [Indexed: 12/18/2022]
Abstract
Despite effective new treatments for Hepatitis C virus (HCV) infection, development of drug resistance, safety concerns and cost are remaining challenges. More importantly, there is no vaccine available against hepatitis C infection. Recent data suggest that there is a strong correlation between spontaneous HCV clearance and human NK cell function, particularly IFN-γ production. Further, IL-15 has innate antiviral activity and is also one of the main factors that activates NK cells to produce IFN-γ. To examine whether IL-15 and IFN-γ have direct antiviral activity against HCV, Huh7.5 cells were treated with either IFN-γ or IL-15 prior to HCV infection. Our data demonstrate that IFN-γ and IL-15 block HCV replication in vitro. Additionally, we show that IL-15 and IFN-γ do not induce anti-HCV effects through the type I interferon signaling pathway or nitric oxide (NO) production. Instead, IL-15 and IFN-γ provide protection against HCV via the ERK pathway. Treatment of Huh7.5 cells with a MEK/ERK inhibitor abrogated the anti-HCV effects of IL-15 and IFN-γ and overexpression of ERK1 prevented HCV replication compared to control transfection. Our in vitro data support the hypothesis that early production of IL-15 and activation of NK cells in the liver lead to control of HCV replication.
Collapse
Affiliation(s)
- Fatemeh Vahedi
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Amanda J Lee
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Susan E Collins
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Marianne V Chew
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Evan Lusty
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Branson Chen
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Anisha Dubey
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Carl D Richards
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Jordan J Feld
- Toronto Centre for Liver Disease, University Health Network, University of Toronto, ON, Canada
| | - Rodney S Russell
- Immunology and Infectious Diseases, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Karen L Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada
| | - Ali A Ashkar
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada; MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
14
|
Yu L, Zhang X, Wu T, Su J, Wang Y, Wang Y, Ruan B, Niu X, Wu Y. Avian infectious bronchitis virus disrupts the melanoma differentiation associated gene 5 (MDA5) signaling pathway by cleavage of the adaptor protein MAVS. BMC Vet Res 2017; 13:332. [PMID: 29132350 PMCID: PMC5683607 DOI: 10.1186/s12917-017-1253-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/31/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Melanoma differentiation associated gene 5 (MDA5) and retinoic acid-inducible gene-I (RIG-I) selectively sense cytoplasmic viral RNA to induce an antiviral immune response. Infectious bronchitis virus (IBV) is one of the most important infectious agents in chickens, and in chicken cells, it can be recognized by MDA5 to activate interferon production. RIG-I is considered to be absent in chickens. However, the absence of RIG-I in chickens raises the question of whether this protein influences the antiviral immune response against IBV infection. RESULTS Here, we showed that chicken cells transfected with domestic goose RIG-I (dgRIG-I) exhibited increased IFN-β activity after IBV infection. We also found that IBV can cleave MAVS, an adaptor protein downstream of RIG-I and MDA5 that acts as a platform for antiviral innate immunity at an early stage of infection. CONCLUSIONS Although chicken MDA5 (chMDA5) is functionally active during IBV infection, the absence of RIG-I may increase the susceptibility of chickens to IBV infection, and IBV may disrupt the activation of the host antiviral response through the cleavage of MAVS.
Collapse
Affiliation(s)
- Liping Yu
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Xiaorong Zhang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Tianqi Wu
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Jin Su
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Yuyang Wang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Yuexin Wang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Baoyang Ruan
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Xiaosai Niu
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Yantao Wu
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
15
|
Canine dorsal root ganglia satellite glial cells represent an exceptional cell population with astrocytic and oligodendrocytic properties. Sci Rep 2017; 7:13915. [PMID: 29066783 PMCID: PMC5654978 DOI: 10.1038/s41598-017-14246-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022] Open
Abstract
Dogs can be used as a translational animal model to close the gap between basic discoveries in rodents and clinical trials in humans. The present study compared the species-specific properties of satellite glial cells (SGCs) of canine and murine dorsal root ganglia (DRG) in situ and in vitro using light microscopy, electron microscopy, and immunostainings. The in situ expression of CNPase, GFAP, and glutamine synthetase (GS) has also been investigated in simian SGCs. In situ, most canine SGCs (>80%) expressed the neural progenitor cell markers nestin and Sox2. CNPase and GFAP were found in most canine and simian but not murine SGCs. GS was detected in 94% of simian and 71% of murine SGCs, whereas only 44% of canine SGCs expressed GS. In vitro, most canine (>84%) and murine (>96%) SGCs expressed CNPase, whereas GFAP expression was differentially affected by culture conditions and varied between 10% and 40%. However, GFAP expression was induced by bone morphogenetic protein 4 in SGCs of both species. Interestingly, canine SGCs also stimulated neurite formation of DRG neurons. These findings indicate that SGCs represent an exceptional, intermediate glial cell population with phenotypical characteristics of oligodendrocytes and astrocytes and might possess intrinsic regenerative capabilities in vivo.
Collapse
|
16
|
Maoto, a Traditional Japanese Herbal Medicine, Inhibits Uncoating of Influenza Virus. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:1062065. [PMID: 28904550 PMCID: PMC5585631 DOI: 10.1155/2017/1062065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/20/2017] [Indexed: 11/17/2022]
Abstract
We previously reported in randomized controlled trials that maoto, a traditional herbal medicine, showed clinical and virological efficacy for seasonal influenza. In this study, a culturing system for influenza was used to test the effect of maoto. A549 cells in the culture were infected with influenza virus A (PR8) and followed after treatment with maoto; the virus titers in the culture supernatant, intracellular viral proteins, and viral RNA were determined. When infected cells were cultured with maoto for 24 hr, the virus titer and protein were significantly reduced compared with medium only. Other subtypes, A/H3N2, H1N1pdm, and B, were also inhibited by maoto. Proliferation of viral RNA in a 6 hr culture was inhibited by maoto in the early phase, especially in the first 30 min. Focusing on the entry step of the influenza virus, we found that endosomal pH, regulated by vacuolar-type H+ ATPase (V-ATPase) located in the membrane, was increased when treated with maoto. We also found that uncoating of influenza viruses was also inhibited by maoto, resulting in the increase of the number of virus particles in endosomes. These results strongly suggest that the inhibition of endosomal acidification by maoto results in blocking influenza virus entry to cytoplasm, probably through the inhibition of V-ATPase. The present study provides evidence that supports the clinical use of maoto for the treatment of influenza.
Collapse
|
17
|
Erwin-Cohen RA, Porter AI, Pittman PR, Rossi CA, DaSilva L. Human transcriptome response to immunization with live-attenuated Venezuelan equine encephalitis virus vaccine (TC-83): Analysis of whole blood. Hum Vaccin Immunother 2016; 13:169-179. [PMID: 27870591 PMCID: PMC5287313 DOI: 10.1080/21645515.2016.1227900] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is an important human and animal alphavirus pathogen transmitted by mosquitoes. The virus is endemic in Central and South America, but has also caused equine outbreaks in southwestern areas of the United States. In an effort to better understand the molecular mechanisms of the development of immunity to this important pathogen, we performed transcriptional analysis from whole, unfractionated human blood of patients who had been immunized with the live-attenuated vaccine strain of VEEV, TC-83. We compared changes in the transcriptome between naïve individuals who were mock vaccinated with saline to responses of individuals who received TC-83. Significant transcriptional changes were noted at days 2, 7, and 14 following vaccination. The top canonical pathways revealed at early and intermediate time points (days 2 and 7) included the involvement of the classic interferon response, interferon-response factors, activation of pattern recognition receptors, and engagement of the inflammasome. By day 14, the top canonical pathways included oxidative phosphorylation, the protein ubiquitination pathway, natural killer cell signaling, and B-cell development. Biomarkers were identified that differentiate between vaccinees and control subjects, at early, intermediate, and late stages of the development of immunity as well as markers which were common to all 3 stages following vaccination but distinct from the sham-vaccinated control subjects. The study represents a novel examination of molecular processes that lead to the development of immunity against VEEV in humans and which may be of value as diagnostic targets, to enhance modern vaccine design, or molecular correlates of protection.
Collapse
Affiliation(s)
- Rebecca A Erwin-Cohen
- a Virology Division, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Aimee I Porter
- a Virology Division, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Phillip R Pittman
- b Division of Medicine, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Cynthia A Rossi
- c Diagnostics Systems Division, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Luis DaSilva
- d Center for Aerobiological Sciences, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| |
Collapse
|
18
|
Valadão ALC, Aguiar RS, de Arruda LB. Interplay between Inflammation and Cellular Stress Triggered by Flaviviridae Viruses. Front Microbiol 2016; 7:1233. [PMID: 27610098 PMCID: PMC4996823 DOI: 10.3389/fmicb.2016.01233] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae family comprises several human pathogens, including Dengue, Zika, Yellow Fever, West Nile, Japanese Encephalitis viruses, and Hepatitis C Virus. Those are enveloped, single-stranded positive sense RNA viruses, which replicate mostly in intracellular compartments associated to endoplasmic reticulum (ER) and Golgi complex. Virus replication results in abundant viral RNAs and proteins, which are recognized by cellular mechanisms evolved to prevent virus infection, resulting in inflammation and stress responses. Virus RNA molecules are sensed by Toll-like receptors (TLRs), RIG-I-like receptors (RIG-I and MDA5) and RNA-dependent protein kinases (PKR), inducing the production of inflammatory mediators and interferons. Simultaneously, the synthesis of virus RNA and proteins are distinguished in different compartments such as mitochondria, ER and cytoplasmic granules, triggering intracellular stress pathways, including oxidative stress, unfolded protein response pathway, and stress granules assembly. Here, we review the new findings that connect the inflammatory pathways to cellular stress sensors and the strategies of Flaviviridae members to counteract these cellular mechanisms and escape immune response.
Collapse
Affiliation(s)
- Ana L C Valadão
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Renato S Aguiar
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Luciana B de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Farias MVN, Lendez PA, Marin M, Quintana S, Martínez-Cuesta L, Ceriani MC, Dolcini GL. Toll-like receptors, IFN-γ and IL-12 expression in bovine leukemia virus-infected animals with low or high proviral load. Res Vet Sci 2016; 107:190-195. [DOI: 10.1016/j.rvsc.2016.06.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/09/2016] [Accepted: 06/18/2016] [Indexed: 10/21/2022]
|
20
|
Local Innate Responses to TLR Ligands in the Chicken Trachea. Viruses 2016; 8:v8070207. [PMID: 27455308 PMCID: PMC4974541 DOI: 10.3390/v8070207] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/11/2022] Open
Abstract
The chicken upper respiratory tract is the portal of entry for respiratory pathogens, such as avian influenza virus (AIV). The presence of microorganisms is sensed by pathogen recognition receptors (such as Toll-like receptors (TLRs)) of the innate immune defenses. Innate responses are essential for subsequent induction of potent adaptive immune responses, but little information is available about innate antiviral responses of the chicken trachea. We hypothesized that TLR ligands induce innate antiviral responses in the chicken trachea. Tracheal organ cultures (TOC) were used to investigate localized innate responses to TLR ligands. Expression of candidate genes, which play a role in antiviral responses, was quantified. To confirm the antiviral responses of stimulated TOC, chicken macrophages were treated with supernatants from stimulated TOC, prior to infection with AIV. The results demonstrated that TLR ligands induced the expression of pro-inflammatory cytokines, type I interferons and interferon stimulated genes in the chicken trachea. In conclusion, TLR ligands induce functional antiviral responses in the chicken trachea, which may act against some pathogens, such as AIV.
Collapse
|
21
|
Zeng M, Chen S, Wang M, Jia R, Zhu D, Liu M, Sun K, Yang Q, Wu Y, Chen X, Cheng A. Molecular identification and comparative transcriptional analysis of myxovirus resistance GTPase (Mx) gene in goose (Anser cygnoide) after H9N2 AIV infection. Comp Immunol Microbiol Infect Dis 2016; 47:32-40. [PMID: 27477505 DOI: 10.1016/j.cimid.2016.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/20/2016] [Accepted: 05/23/2016] [Indexed: 12/18/2022]
Abstract
Interferon (IFN)-induced myxovirus resistance (Mx) GTPases belong to the family of dynamin-like GTPases and control a diverse range of viruses. In this study, the identified goose Mx (goMx) mRNA is 2009bp long, shares partially conserved exons with other homologues, and shares highly conserved domains in its primary structure. The amino acid position 629 (629aa) of the goMx protein was identified as serine (Ser), in contrast to the Ser located at 631aa in chicken Mx, which is considered to be responsible for the lack of chicken Mx antiviral activity. In addition, the goMx 142aa residue in the dynamin family signature differs from that of other functional Mx proteins. Transcriptional analysis revealed that goMx was mainly expressed in the digestive, respiratory and immune systems in an age-specific manner. GoMx transcript levels in goose peripheral blood mononuclear cells (PBMCs) were found to be significantly up-regulated by various agonists and avian viruses. Furthermore, a time course study of the effects of H9N2 avian influenza virus (AIV) on goMx expression in infected goslings suggested that H9N2 AIV affected goMx expression. However, significant changes in goMx expression were observed in the trachea, lung and small intestine of infected birds. Altogether, these results indicate that goMx protein may have acquired its broad antiviral activity by changing only a few amino acids at select sites, even as it shares a conserved architectures with species.
Collapse
Affiliation(s)
- Miao Zeng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Dekang Zhu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Kunfeng Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiaoyue Chen
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| |
Collapse
|
22
|
Yu K, Deng S, Wang H, Zhang Y, Chen X, Wang K, Hu R, Lian Z, Li N. Small interfering RNA expression inhibits avian infectious bronchitis virus replication and inflammatory response. Antivir Ther 2016; 21:469-479. [PMID: 26835751 DOI: 10.3851/imp3027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2016] [Indexed: 12/09/2022]
Abstract
BACKGROUND Avian infectious bronchitis virus (IBV) is a major cause of poor weight gain and mortality among chicks. METHODS A lentivirus vector was used to generate transgenic chickens expressing small interfering RNA (siRNA) targeting the M protein of IBV. Offspring of generation 0 (G0) were screened to identify G1 transgenic chickens (Tg). Monocytes from G1 Tg were stimulated with IBV in vitro. RESULTS Monocytes producing siRNA efficiently inhibit IBV replication. Expression of inflammatory cytokines, Mx protein and nitric oxide levels were lower in early IBV infection in Tg. In vivo experiments show that siRNA expression inhibits IBV replication, significantly decreases mortality and increases weight gain. Inflammatory responses and oxidative damage were significantly decreased, yielding minimal tissue injury. The inflammatory responses indicate that the cellular immune response is most effective during the initial stage, while the humoral immune response is more significant in later stages of infection. CONCLUSIONS Small interfering RNA expression inhibits avian IBV replication and inflammatory response.
Collapse
Affiliation(s)
- Kun Yu
- National Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China.,Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shoulong Deng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hai Wang
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yi Zhang
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xuehui Chen
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Kejun Wang
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rui Hu
- Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhengxing Lian
- National Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China.,Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Li
- National Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
23
|
Li L, Ulrich R, Baumgärtner W, Gerhauser I. Interferon-stimulated genes-essential antiviral effectors implicated in resistance to Theiler's virus-induced demyelinating disease. J Neuroinflammation 2015; 12:242. [PMID: 26703877 PMCID: PMC4690264 DOI: 10.1186/s12974-015-0462-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/17/2015] [Indexed: 01/08/2023] Open
Abstract
Background Experimental infection of mice with Theiler’s murine encephalomyelitis virus (TMEV) is used as an animal model of human multiple sclerosis. TMEV persists in susceptible mouse strains and causes a biphasic disease consisting of acute polioencephalomyelitis and chronic demyelinating leukomyelitis. In contrast, resistant mice eliminate the virus within 2 to 4 weeks, which seems to be based on a strong antiviral innate immune response including the activation of the type I interferon (IFN) pathway. Several interferon-stimulated genes (ISGs) such as IFN-stimulated protein of 15 kDa (ISG15), protein kinase R (PKR), and 2′5′-oligoadenylate synthetase (OAS) function as antiviral effectors and might contribute to virus elimination. Nevertheless, detailed investigations of the type I IFN pathway during TMEV-induced demyelinating disease (TMEV-IDD) are lacking. Methods The present study evaluated microarray data of the spinal cord obtained from susceptible SJL/J mice after TMEV infection focusing on IFN-related genes. Moreover, ISG gene and protein expression was determined in mock- and TMEV-infected SJL/J mice and compared to its expression in resistant C57BL/6 mice using real- time PCR, immunohistochemistry, and immunofluorescence. Results Interestingly, despite of increased ISG gene expression during TMEV-IDD, ISG protein expression was impaired in SJL/J mice and mainly restricted to demyelinated lesions. In contrast, high ISG protein levels were found in spinal cord gray and white matter of C57BL/6 compared to SJL/J mice in the acute and chronic phase of TMEV-IDD. In both mouse strains, ISG15 was mainly found in astrocytes and endothelial cells, whereas PKR was predominantly expressed by microglia/macrophages, oligodendrocytes, and neurons. Only few cells were immunopositive for OAS proteins. Conclusions High levels of antiviral ISG15 and PKR proteins in the spinal cord of C57BL/6 mice might block virus replication and play an important role in the resistance to TMEV-IDD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0462-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lin Li
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany. .,Center of Systems Neuroscience Hannover, Hannover, Germany.
| | - Reiner Ulrich
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany. .,Center of Systems Neuroscience Hannover, Hannover, Germany.
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany. .,Center of Systems Neuroscience Hannover, Hannover, Germany.
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
| |
Collapse
|
24
|
Reduction of avian influenza virus shedding by administration of Toll-like receptor ligands to chickens. Vaccine 2015; 33:4843-9. [PMID: 26238721 DOI: 10.1016/j.vaccine.2015.07.070] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 07/20/2015] [Accepted: 07/22/2015] [Indexed: 11/23/2022]
Abstract
Avian influenza viruses (AIV) are of concern to the poultry industry. Outbreaks of AIV highlight the urgent need for effective control measures. Prophylactic strategies should be explored that rapidly elicit immunity against the virus. Toll-like receptors (TLRs) are innate immune molecules that can induce anti-viral responses, therefore the application of TLR ligands as prophylactic agents in chickens is gaining more attention. We hypothesized that treatment of chickens with TLR ligands reduces the shedding of AIV from infected birds. In addition, the effects of TLR ligand dose and route of administration on the efficiency of TLR ligands to reduce AIV shedding were examined. Chickens were treated with TLR2, 4, 7 and 21 ligands using different doses and routes of administration, 18h before AIV infection. Moreover, the expression of several candidate genes, such as type I interferons, PKR, OAS, viperin and IFITM3 was quantified at 3, 8 and 18h post-treatment with TLR ligands. The results revealed that route of administration and dosage affect the efficacy of TLR ligands to reduce virus shedding. Furthermore, varying effects were observed when different ligands were applied. Our results demonstrated that all TLR ligand treatments reduced AIV shedding, with the CpG-ODN 1826 being the most efficacious to reduce oral virus shedding, whereas LPS from Escherichia coli 026:B6 resulted in the largest reduction in cloacal virus shedding. Moreover, TLR ligands induced the expression of genes involved in antiviral responses such as type I interferons and interferon-stimulated genes in chicken trachea and cecal tonsils. These results raise the possibility of treatment of chickens with TLR ligands as anti-viral agents.
Collapse
|
25
|
Hafidh RR, Abdulamir AS, Abu Bakar F, Sekawi Z, Jahansheri F, Jalilian FA. Novel antiviral activity of mung bean sprouts against respiratory syncytial virus and herpes simplex virus -1: an in vitro study on virally infected Vero and MRC-5 cell lines. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:179. [PMID: 26062546 PMCID: PMC4461985 DOI: 10.1186/s12906-015-0688-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 05/21/2015] [Indexed: 01/26/2023]
Abstract
BACKGROUND New sources for discovering novel antiviral agents are desperately needed. The current antiviral products are both expensive and not very effective. METHODS The antiviral activity of methanol extract of mung bean sprouts (MBS), compared to Ribavarin and Acyclovir, on respiratory syncytial virus (RSV) and Herpes Simplex virus -1 (HSV-1) was investigated using cytotoxicity, virus yield reduction, virucidal activity, and prophylactic activity assays on Vero and MRC-5 cell lines. Moreover, the level of antiviral cytokines, IFNβ, TNFα, IL-1, and IL-6 was assessed in MBS-treated, virally infected, virally infected MBS-treated, and control groups of MRC-5 cells using ELISA. RESULTS MBS extract showed reduction factors (RF) 2.2 × 10 and 0.5 × 10(2) for RSV and HSV-1, respectively. The 2 h incubation virucidal and prophylactic selectivity indices (SI) of MBS on RSV were 14.18 and 12.82 versus Ribavarin SI of 23.39 and 21.95, respectively, and on HSV-1, SI were 18.23 and 10.9 versus Acyclovir, 22.56 and 15.04, respectively. All SI values were >10 indicating that MBS has a good direct antiviral and prophylactic activities on both RSV and HSV-1. Moreover, interestingly, MBS extract induced vigorously IFNβ, TNFα, IL-1, and IL-6 cytokines in MRC-5 infected-treated group far more than other groups (P < 0.05) and induced TNFα and IL-6 in treated group more than infected group (P < 0.05). CONCLUSIONS MBS extract has potent antiviral and to a lesser extent, prophylactic activities against both RSV and HSV-1, and in case of HSV-1, these activities were comparable to Acyclovir. Part of the underlying mechanism(s) of these activities is attributed to MBS potential to remarkably induce antiviral cytokines in human cells. Hence, we infer that MBS methanol extract could be used as such or as purified active component in protecting and treating RSV and HSV-1 infections. More studies are needed to pinpoint the exact active components responsible for the MBS antiviral activities.
Collapse
|
26
|
Castillo Ramirez JA, Urcuqui-Inchima S. Dengue Virus Control of Type I IFN Responses: A History of Manipulation and Control. J Interferon Cytokine Res 2015; 35:421-30. [PMID: 25629430 DOI: 10.1089/jir.2014.0129] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The arthropod-borne diseases caused by dengue virus (DENV) are a major and emerging problem of public health worldwide. Infection with DENV causes a series of clinical manifestations ranging from mild flu syndrome to severe diseases that include hemorrhage and shock. It has been demonstrated that the innate immune response plays a key role in DENV pathogenesis. However, in recent years, it was shown that DENV evades the innate immune response by blocking type I interferon (IFN-I). It has been demonstrated that DENV can inhibit both the production and the signaling of IFN-I. The viral proteins, NS2A and NS3, inhibit IFN-I production by degrading cellular signaling molecules. In addition, the viral proteins, NS2A, NS4A, NS4B, and NS5, can inhibit IFN-I signaling by blocking the phosphorylation of the STAT1 and STAT2 molecules. Finally, NS5 mediates the degradation of STAT2 using the proteasome machinery. In this study, we briefly review the most recent insights regarding the IFN-I response to DENV infection and its implication for pathogenesis.
Collapse
Affiliation(s)
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA , Medellín, Colombia
| |
Collapse
|
27
|
Lai KY, Ng WYG, Cheng FF. Human Ebola virus infection in West Africa: a review of available therapeutic agents that target different steps of the life cycle of Ebola virus. Infect Dis Poverty 2014; 3:43. [PMID: 25699183 PMCID: PMC4334593 DOI: 10.1186/2049-9957-3-43] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022] Open
Abstract
The recent outbreak of the human Zaire ebolavirus (EBOV) epidemic is spiraling out of control in West Africa. Human EBOV hemorrhagic fever has a case fatality rate of up to 90%. The EBOV is classified as a biosafety level 4 pathogen and is considered a category A agent of bioterrorism by Centers for Disease Control and Prevention, with no approved therapies and vaccines available for its treatment apart from supportive care. Although several promising therapeutic agents and vaccines against EBOV are undergoing the Phase I human trial, the current epidemic might be outpacing the speed at which drugs and vaccines can be produced. Like all viruses, the EBOV largely relies on host cell factors and physiological processes for its entry, replication, and egress. We have reviewed currently available therapeutic agents that have been shown to be effective in suppressing the proliferation of the EBOV in cell cultures or animal studies. Most of the therapeutic agents in this review are directed against non-mutable targets of the host, which is independent of viral mutation. These medications are approved by the Food and Drug Administration (FDA) for the treatment of other diseases. They are available and stockpileable for immediate use. They may also have a complementary role to those therapeutic agents under development that are directed against the mutable targets of the EBOV.
Collapse
Affiliation(s)
- Kang Yiu Lai
- />Department of Intensive Care, Queen Elizabeth Hospital, HKSAR, B6, 30 Gascoigne Rd, Kowloon, Hong Kong SAR China
| | - Wing Yiu George Ng
- />Department of Intensive Care, Queen Elizabeth Hospital, HKSAR, B6, 30 Gascoigne Rd, Kowloon, Hong Kong SAR China
| | - Fan Fanny Cheng
- />Department of Medicine, Queen Elizabeth Hospital, HKSAR, Kowloon, Hong Kong SARChina
| |
Collapse
|
28
|
Cervantes JL, Hawley KL, Benjamin SJ, Weinerman B, Luu SM, Salazar JC. Phagosomal TLR signaling upon Borrelia burgdorferi infection. Front Cell Infect Microbiol 2014; 4:55. [PMID: 24904837 PMCID: PMC4033037 DOI: 10.3389/fcimb.2014.00055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/09/2014] [Indexed: 12/31/2022] Open
Abstract
Internalization and degradation of live Bb within phagosomal compartments of monocytes, macrophages and dendritic cells (DCs), allows for the release of lipoproteins, nucleic acids and other microbial products, triggering a broad and robust inflammatory response. Toll-like receptors (TLRs) are key players in the recognition of spirochetal ligands from whole viable organisms (i.e., vita-PAMPs). Herein we will review the role of endosomal TLRs in the response to the Lyme disease spirochete.
Collapse
Affiliation(s)
- Jorge L Cervantes
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA
| | - Kelly L Hawley
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA
| | - Sarah J Benjamin
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA
| | - Bennett Weinerman
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA
| | - Stephanie M Luu
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center Farmington, CT, USA
| | - Juan C Salazar
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA ; Department of Immunology, University of Connecticut Health Center Farmington, CT, USA
| |
Collapse
|
29
|
Toll-like receptor activation and expression in bovine alpha-herpesvirus infections. Res Vet Sci 2014; 96:196-203. [DOI: 10.1016/j.rvsc.2013.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 10/07/2013] [Accepted: 11/30/2013] [Indexed: 01/19/2023]
|
30
|
Lee CC, Wu CC, Lin TL. Chicken melanoma differentiation-associated gene 5 (MDA5) recognizes infectious bursal disease virus infection and triggers MDA5-related innate immunity. Arch Virol 2014; 159:1671-86. [PMID: 24452668 PMCID: PMC7086882 DOI: 10.1007/s00705-014-1983-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 11/09/2013] [Indexed: 12/24/2022]
Abstract
The objective of the present study was to determine if chicken melanoma differentiation-associated gene 5 (MDA5) senses infectious bursal disease virus (IBDV) infection to initiate and amplify an innate immune response in the chicken MDA5 (chMDA5) signaling pathway. Chicken embryo fibroblast DF-1 cells were infected with IBDV LP1 at a multiplicity of infection (MOI) of 0.5 or 10. In addition, knockdown and overexpression of chMDA5 were performed by transfecting DF-1 cells with chMDA5-targeting small interfering RNA (siRNA) or chMDA5-expressing DNA. The transfected cells were infected with IBDV LP1 at an MOI of 10. Cell culture supernatants and lysates were collected at 2, 8, 16 and 24 hours postinfection (hpi) for IBDV titer determination and RNA extraction, respectively. IBDV RNA loads and mRNA expression levels of chicken MDA5, interferon-β (IFN-β) promoter stimulator 1 (IPS-1), interferon regulatory factor-3 (IRF-3), IFN-β, double-stranded RNA-dependent protein kinase (PKR), 2′,5′-oligoadenylate synthetase (OAS), myxovirus resistance gene (Mx), and major histocompatibility complex class I (MHC class I) were determined by real-time RT-PCR. The IBDV titer increased up to 1.4 × 107 plaque-forming units (PFU)/mL at 24 hpi, and the IBDV RNA load reached 464 ng/μL at 24 hpi. The mRNA expression levels of chicken MDA5, IRF-3, IFN-β, PKR, OAS, Mx and MHC class I in IBDV-infected DF-1 cells exhibited significant (p < 0.05) upregulation up to 906-, 199-, 26,310-, 12-, 66,144-, 64,039- and 33-fold, respectively. Expressed chMDA5 from transfection and double-stranded RNA from IBDV infection were localized or colocalized in the cytoplasm of DF-1 cells at 16 hpi. When chMDA5 was knocked down in DF-1 cells, IBDV titers and RNA loads were significantly higher (p < 0.05) than those in DF-1 cells without chMDA5 knockdown at 24 hpi. The expression levels of chicken MDA5, IRF-3, IFN-β and MHC class I in chMDA5-knockdown DF-1 cells were significantly lower (p < 0.05) at 16 and 24 hpi. DF-1 cells overexpressing chMDA5 by transfection with chMDA5 expressing DNA had significantly lower (p < 0.05) IBDV titers and RNA loads at 16 and 24 hpi and showed significantly higher (p < 0.05) expression of chicken MDA5, IRF-3, IFN-β, PKR, OAS, Mx and MHC class I at 2 hpi. The results indicated that chicken MDA5 recognized IBDV infection and that this interaction resulted in the activation of chMDA5-related innate immune genes and upregulation of chicken MHC class I.
Collapse
Affiliation(s)
- Chih-Chun Lee
- Department of Comparative Pathobiology, Purdue University, 406, S. University St, West Lafayette, IN, 47907, USA
| | | | | |
Collapse
|
31
|
Muralidharan S, Mandrekar P. Cellular stress response and innate immune signaling: integrating pathways in host defense and inflammation. J Leukoc Biol 2013; 94:1167-84. [PMID: 23990626 DOI: 10.1189/jlb.0313153] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Extensive research in the past decade has identified innate immune recognition receptors and intracellular signaling pathways that culminate in inflammatory responses. Besides its role in cytoprotection, the importance of cell stress in inflammation and host defense against pathogens is emerging. Recent studies have shown that proteins in cellular stress responses, including the heat shock response, ER stress response, and DNA damage response, interact with and regulate signaling intermediates involved in the activation of innate and adaptive immune responses. The effect of such regulation by cell stress proteins may dictate the inflammatory profile of the immune response during infection and disease. In this review, we describe the regulation of innate immune cell activation by cell stress pathways, present detailed descriptions of the types of stress response proteins and their crosstalk with immune signaling intermediates that are essential in host defense, and illustrate the relevance of these interactions in diseases characteristic of aberrant immune responses, such as chronic inflammatory diseases, autoimmune disorders, and cancer. Understanding the crosstalk between cellular stress proteins and immune signaling may have translational implications for designing more effective regimens to treat immune disorders.
Collapse
Affiliation(s)
- Sujatha Muralidharan
- 1.LRB 221, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605.
| | | |
Collapse
|
32
|
Hayashi T, Watanabe C, Suzuki Y, Tanikawa T, Uchida Y, Saito T. Chicken MDA5 senses short double-stranded RNA with implications for antiviral response against avian influenza viruses in chicken. J Innate Immun 2013; 6:58-71. [PMID: 23860388 DOI: 10.1159/000351583] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/19/2013] [Indexed: 12/24/2022] Open
Abstract
Mammalian melanoma differentiation-associated gene-5 (MDA5) and retinoic acid-inducible gene-I (RIG-I) selectively sense double-stranded RNA (dsRNA) according to length, as well as various RNA viruses to induce an antiviral response. RIG-I, which plays a predominant role in the induction of antiviral responses against influenza virus infection, has been considered to be lacking in chicken, putting the function of chicken MDA5 (chMDA5) under the spotlight. Here, we show that chMDA5, unlike mammalian MDA5, preferentially senses shorter dsRNA synthetic analogues, poly(I:C), in chicken DF-1 fibroblasts. A requirement for caspase activation and recruitment domains for chMDA5-mediated chicken interferon beta (chIFNβ) induction and its interaction with mitochondrial antiviral signaling proteins were demonstrated. We also found that chMDA5 is involved in chIFNβ induction against avian influenza virus infection. Our findings imply that chMDA5 compensates in part the function of RIG-I in chicken, and highlights the importance of chMDA5 in the innate immune response in chicken.
Collapse
Affiliation(s)
- Tsuyoshi Hayashi
- Viral diseases and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Tsukuba, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Mattoscio D, Segré CV, Chiocca S. Viral manipulation of cellular protein conjugation pathways: The SUMO lesson. World J Virol 2013; 2:79-90. [PMID: 24175232 PMCID: PMC3785051 DOI: 10.5501/wjv.v2.i2.79] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/23/2013] [Accepted: 02/06/2013] [Indexed: 02/05/2023] Open
Abstract
Small ubiquitin-like modifier (SUMO)ylation is a key post-translational modification mechanism that controls the function of a plethora of proteins and biological processes. Given its central regulatory role, it is not surprising that it is widely exploited by viruses. A number of viral proteins are known to modify and/or be modified by the SUMOylation system to exert their function, to create a cellular environment more favorable for virus survival and propagation, and to prevent host antiviral responses. Since the SUMO pathway is a multi-step cascade, viral proteins engage with it at many levels, to advance and favor each stage of a typical infection cycle: replication, viral assembly and immune evasion. Here we review the current knowledge on the interplay between the host SUMO system and viral lifecycle.
Collapse
|
34
|
Abstract
In vitro transcription of DNA with phage RNA polymerases is currently the most efficient method to produce long sequence-specific RNA. While the reaction can yield large quantities of RNA, it contains impurities due to various unwanted activities of the polymerases. Here, we described an easily performed HPLC purification that removes multiple contaminants from in vitro transcribed RNA and is scalable. The purified RNA is translated at much greater levels, especially in primary cells and in vivo. HPLC purification of RNA containing modified nucleosides that suppress RNA-mediated activation of innate immune sensors leads to a non-immunogenic RNA with superior translational capacity.
Collapse
|
35
|
Cervantes JL, Weinerman B, Basole C, Salazar JC. TLR8: the forgotten relative revindicated. Cell Mol Immunol 2012; 9:434-8. [PMID: 23085951 PMCID: PMC3498840 DOI: 10.1038/cmi.2012.38] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The endosomal Toll-like receptors (TLRs) TLR3, TLR7, TLR8 and TLR9 are important in sensing foreign nucleic acids encountered by phagocytes. Because TLR8 was initially thought to be non-functional in mice, less is known about TLR8 than the genetically and functionally related TLR7. Originally associated with the recognition of single-stranded RNA of viral origin, there is now evidence that human TLR8 is also able to sense bacterial RNA released within phagosomal vacuoles, inducing the production of both nuclear factor (NF)-κB-dependent cytokines and type I interferons (IFNs), such as IFN-β. The functions of TLR8 extend beyond the recognition of foreign pathogens and include cross-talk with other endosomal TLRs, a process that may also have a role in the generation of autoimmunity.
Collapse
Affiliation(s)
- Jorge L Cervantes
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | |
Collapse
|
36
|
Zhao X, Ai M, Guo Y, Zhou X, Wang L, Li X, Yao C. Poly I:C-induced tumor cell apoptosis mediated by pattern-recognition receptors. Cancer Biother Radiopharm 2012; 27:530-4. [PMID: 23062195 DOI: 10.1089/cbr.2012.1226] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Poly I:C is a synthetic dsRNA that can imitate a viral infection and elicit host immune responses by triggering specific pattern-recognition receptors (PRRs) such as toll-like receptor 3 and retinoic acid inducible gene I(RIG-I)-like receptors, including RIG-I and melanoma differentiation-associated gene 5. Activation of these PRRs by poly I:C triggers a signal transduction cascade that results in the activation of NF-κB and production of type I interferon. Poly I:C has been used as a vaccine adjuvant for cancer immunotherapy for several decades. Evidence from recent studies indicates that poly I:C can directly induce apoptosis in several types of tumor cells, thus providing a new therapeutic approach for cancer treatment. However, the molecular mechanism underlying the induction of apoptosis by poly I:C is still unclear. In this review, we summarize the current knowledge of poly I:C-induced tumor cell apoptosis, focusing on the key molecules and pathways involved in this process.
Collapse
Affiliation(s)
- Xiangzhong Zhao
- Key Laboratory for Rare Diseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Science, Jinan, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Sugamata R, Dobashi H, Nagao T, Yamamoto KI, Nakajima N, Sato Y, Aratani Y, Oshima M, Sata T, Kobayashi K, Kawachi S, Nakayama T, Suzuki K. Contribution of neutrophil-derived myeloperoxidase in the early phase of fulminant acute respiratory distress syndrome induced by influenza virus infection. Microbiol Immunol 2012; 56:171-82. [PMID: 22211924 DOI: 10.1111/j.1348-0421.2011.00424.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Because the pathogenesis of acute respiratory distress syndrome (ARDS) induced by influenza virus infection remains unknown, we can only improve on existing therapeutic interventions. To approach the subject, we investigated immunological etiology focused on cytokines and an acute lung damage factor in influenza-induced ARDS by using a PR-8 (A/H1N1)-infected mouse model. The infected mouse showed fulminant severe pneumonia with leukocyte infiltration, claudin alteration on tight junctions, and formation of hyaline membranes. In addition to interferon (IFN)-α, plenty of keratinocyte-derived chemokines (KC), macrophage inflammatory protein 2 (MIP-2), regulated on activation normal T-cell expressed and secreted (RANTES), and monocyte chemotactic protein 1 (MCP-1) were significantly released into bronchoalveolar lavage fluid (BALF) of the model. We focused on neutrophil myeloperoxidase (MPO) as a potent tissue damage factor and examined its contribution in influenza pneumonia by using mice genetically lacking in MPO. The absence of MPO reduced inflammatory damage with suppression of leakage of total BALF proteins associated with alteration of claudins in the lung. MPO(-/-) mice also suppressed viral load in the lung. The present study suggests that MPO-mediated OCl(-) generation affects claudin molecules and leads to protein leakage and viral spread as a damage factor in influenza-induced ARDS.
Collapse
Affiliation(s)
- Ryuichi Sugamata
- Inflammation Program, Graduate School of Medicine, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba-city, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hundeshagen A, Hecker M, Paap BK, Angerstein C, Kandulski O, Fatum C, Hartmann C, Koczan D, Thiesen HJ, Zettl UK. Elevated type I interferon-like activity in a subset of multiple sclerosis patients: molecular basis and clinical relevance. J Neuroinflammation 2012; 9:140. [PMID: 22727118 PMCID: PMC3464734 DOI: 10.1186/1742-2094-9-140] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 06/22/2012] [Indexed: 12/26/2022] Open
Abstract
Background A subset of patients with multiple sclerosis (MS) shows an increased endogenous IFN-like activity before initiation of IFN-beta treatment. The molecular basis of this phenomenon and its relevance to predict individual therapy outcomes are not yet fully understood. We studied the expression patterns of these patients, the prognostic value of an elevated IFN-like activity, and the gene regulatory effects of exogenously administered IFN-beta. Methods Microarray gene expression profiling was performed for 61 MS patients using peripheral blood mononuclear cells obtained before and after 1 month of IFN-beta therapy. Expression levels of genes involved in pathways either inducing or being activated by IFN-beta were compared between patients with high (MX1high cohort) and low (MX1low cohort) endogenous IFN-like activity. Patients were followed for 5 years and relapses as well as progression on the expanded disability status scale (EDSS) were documented. Results Before the start of therapy, 11 patients presented elevated mRNA levels of IFN-stimulated genes indicative of a relatively high endogenous IFN-like activity (MX1high). In these patients, pathogen receptors (for example, TLR7, RIG-I and IFIH1) and transcription factors were also expressed more strongly, which could be attributed to an overactivity of IFN-stimulated gene factor 3 (ISGF3, a complex formed by STAT1, STAT2 and IFN regulatory factor 9). After 1 month of IFN-beta therapy, the expression of many pathway genes was significantly induced in MX1low patients, but remained unaltered in MX1high patients. During follow-up, relapse rate and changes in EDSS were comparable between both patient groups, with differences seen between different types of IFN-beta drug application. Conclusions Therapeutic IFN-beta induces the transcription of several genes involved in IFN-related pathways. In a subgroup of MS patients, the expression of these genes is already increased before therapy initiation, possibly driven by an overexpression of ISGF3. Patients with high and low endogenous IFN-like activity showed similar clinical long-term courses of disease. Different results were obtained for different IFN-beta drug preparations, and this merits further investigation.
Collapse
Affiliation(s)
- Alexander Hundeshagen
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Lester K, Hall M, Urquhart K, Gahlawat S, Collet B. Development of an in vitro system to measure the sensitivity to the antiviral Mx protein of fish viruses. J Virol Methods 2012; 182:1-8. [DOI: 10.1016/j.jviromet.2012.01.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 01/12/2012] [Accepted: 01/16/2012] [Indexed: 11/27/2022]
|
40
|
Wilson VG. Sumoylation at the host-pathogen interface. Biomolecules 2012; 2:203-27. [PMID: 23795346 PMCID: PMC3685863 DOI: 10.3390/biom2020203] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 03/21/2012] [Accepted: 03/27/2012] [Indexed: 12/11/2022] Open
Abstract
Many viral proteins have been shown to be sumoylated with corresponding regulatory effects on their protein function, indicating that this host cell modification process is widely exploited by viral pathogens to control viral activity. In addition to using sumoylation to regulate their own proteins, several viral pathogens have been shown to modulate overall host sumoylation levels. Given the large number of cellular targets for SUMO addition and the breadth of critical cellular processes that are regulated via sumoylation, viral modulation of overall sumoylation presumably alters the cellular environment to ensure that it is favorable for viral reproduction and/or persistence. Like some viruses, certain bacterial plant pathogens also target the sumoylation system, usually decreasing sumoylation to disrupt host anti-pathogen responses. The recent demonstration that Listeria monocytogenes also disrupts host sumoylation, and that this is required for efficient infection, extends the plant pathogen observations to a human pathogen and suggests that pathogen modulation of host sumoylation may be more widespread than previously appreciated. This review will focus on recent aspects of how pathogens modulate the host sumoylation system and how this benefits the pathogen.
Collapse
Affiliation(s)
- Van G Wilson
- Department of Microbial & Molecular Pathogenesis, College of Medicine, Texas A&M Health Science Center, 8447 HWY 47, Bryan, TX 77807-1359
| |
Collapse
|
41
|
Brown MG, McAlpine SM, Huang YY, Haidl ID, Al-Afif A, Marshall JS, Anderson R. RNA sensors enable human mast cell anti-viral chemokine production and IFN-mediated protection in response to antibody-enhanced dengue virus infection. PLoS One 2012; 7:e34055. [PMID: 22479521 PMCID: PMC3316603 DOI: 10.1371/journal.pone.0034055] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 02/24/2012] [Indexed: 12/13/2022] Open
Abstract
Dengue hemorrhagic fever and/or dengue shock syndrome represent the most serious pathophysiological manifestations of human dengue virus infection. Despite intensive research, the mechanisms and important cellular players that contribute to dengue disease are unclear. Mast cells are tissue-resident innate immune cells that play a sentinel cell role in host protection against infectious agents via pathogen-recognition receptors by producing potent mediators that modulate inflammation, cell recruitment and normal vascular homeostasis. Most importantly, mast cells are susceptible to antibody-enhanced dengue virus infection and respond with selective cytokine and chemokine responses. In order to obtain a global view of dengue virus-induced gene regulation in mast cells, primary human cord blood-derived mast cells (CBMCs) and the KU812 and HMC-1 mast cell lines were infected with dengue virus in the presence of dengue-immune sera and their responses were evaluated at the mRNA and protein levels. Mast cells responded to antibody-enhanced dengue virus infection or polyinosiniċpolycytidylic acid treatment with the production of type I interferons and the rapid and potent production of chemokines including CCL4, CCL5 and CXCL10. Multiple interferon-stimulated genes were also upregulated as well as mRNA and protein for the RNA sensors PKR, RIG-I and MDA5. Dengue virus-induced chemokine production by KU812 cells was significantly modulated by siRNA knockdown of RIG-I and PKR, in a negative and positive manner, respectively. Pretreatment of fresh KU812 cells with supernatants from dengue virus-infected mast cells provided protection from subsequent infection with dengue virus in a type I interferon-dependent manner. These findings support a role for tissue-resident mast cells in the early detection of antibody-enhanced dengue virus infection via RNA sensors, the protection of neighbouring cells through interferon production and the potential recruitment of leukocytes via chemokine production.
Collapse
Affiliation(s)
- Michael G. Brown
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Sarah M. McAlpine
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Yan Y. Huang
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Ian D. Haidl
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ayham Al-Afif
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Jean S. Marshall
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Robert Anderson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
- Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
- * E-mail:
| |
Collapse
|
42
|
Lee CC, Wu CC, Lin TL. Characterization of chicken melanoma differentiation-associated gene 5 (MDA5) from alternative translation initiation. Comp Immunol Microbiol Infect Dis 2012; 35:335-43. [PMID: 22459580 DOI: 10.1016/j.cimid.2012.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 01/05/2012] [Accepted: 02/21/2012] [Indexed: 12/24/2022]
Abstract
The present study was undertaken to identify and characterize chicken melanoma differentiation-associated gene 5 (MDA5) from alternative translation initiation. The alternatively translated chicken MDA5 had an open reading frame of 3309 base pairs, shorter than the predicted chicken MDA5 sequence by 549 base pairs and longer than the recently published chicken MDA5 (GU570144) by 303 base pairs. The domain architecture was conserved among MDA5 molecules from various vertebrates. The alternatively translated chicken MDA5 shared high genetic identity to zebrafinch MDA5, but not to fish or mammalian MDA5. Various concentrations of chicken MDA5 transcripts were detected in different chicken tissues, with the highest concentration found in the intestine (1.4×10(10)copy/mg tissue). Chicken interferon (IFN-β) expression was dependent on the dose of pCDNA3.1-MDA5-long transfection in DF1 cells without double-stranded RNA stimulation. DF1 cells transfected with polyinosinic-polycytidylic acid (poly(I:C)) had significantly (p<0.05) increased fold changes in chicken MDA5 and IFN-β mRNA expression as compared to those in the controls. DF1 cells incubated with poly(I:C) did not have significant (p>0.05) changes in mRNA expression of chicken MDA5 and IFN-β. Chicken MDA5 mRNA level did not change significantly (p>0.05) in DF1 cells treated with imiquimod or bacterial DNA. The results indicate that although chicken MDA5 is phylogenetically different from mammalian MDA5, the domain architecture remains conserved. Chicken MDA5 can be detected in various tissues and functions to signal the activation of chicken IFN-β when overexpressing or being stimulated by poly(I:C).
Collapse
Affiliation(s)
- Chih-Chun Lee
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
43
|
Nabeshima S, Kashiwagi K, Ajisaka K, Masui S, Takeoka H, Ikematsu H, Kashiwagi S. A randomized, controlled trial comparing traditional herbal medicine and neuraminidase inhibitors in the treatment of seasonal influenza. J Infect Chemother 2012; 18:534-43. [PMID: 22350323 DOI: 10.1007/s10156-012-0378-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 01/23/2012] [Indexed: 11/26/2022]
Abstract
The herbal medicine, maoto, has been traditionally prescribed to patients with influenza in Japan. To better understand the efficacy of maoto for the treatment of influenza, a randomized trial was conducted for comparison with oseltamivir or zanamivir. Adult patients with influenza symptoms, including fever, positive for quick diagnostic kit for influenza within 48 h of fever onset were assessed for enrollment. The data of 28 patients randomly assigned to maoto (n = 10), oseltamivir (n = 8), or zanamivir (n = 10) were analyzed for the duration of fever (>37.5°C) and total symptom score from symptom cards recorded by the patient. Viral isolation and serum cytokine measurements were also done on days 1, 3, and 5. Maoto granules, a commercial medical dosage form, are made from four plants: Ephedra Herb, Apricot Kernel, Cinnamon Bark, and Glycyrrhiza Root. Median durations of fever of patients assigned maoto, oseltamivir, or zanamivir were 29, 46, or 27 h, respectively, significantly different for maoto and oseltamivir. No significant between-group differences were found in total symptom score among three groups. Viral persistent rates and serum cytokine levels (IFN-α, IL-6, IL-8, IL-10, and TNF-α) during the study period showed no differences among three groups. The administration of oral maoto granules to healthy adults with seasonal influenza was well tolerated and associated with equivalent clinical and virological efficacy to neuraminidase inhibitors.
Collapse
Affiliation(s)
- Shigeki Nabeshima
- General Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| | | | | | | | | | | | | |
Collapse
|
44
|
Hösel M, Broxtermann M, Janicki H, Esser K, Arzberger S, Hartmann P, Gillen S, Kleeff J, Stabenow D, Odenthal M, Knolle P, Hallek M, Protzer U, Büning H. Toll-like receptor 2-mediated innate immune response in human nonparenchymal liver cells toward adeno-associated viral vectors. Hepatology 2012; 55:287-97. [PMID: 21898480 DOI: 10.1002/hep.24625] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Adeno-associated viral vectors (rAAV) are frequently used in gene therapy trials. Although rAAV vectors are of low immunogenicity, humoral as well as T cell responses may be induced. While the former limits vector reapplication, the expansion of cytotoxic T cells correlates with liver inflammation and loss of transduced hepatocytes. Because adaptive immune responses are a consequence of recognition by the innate immune system, we aimed to characterize cell autonomous immune responses elicited by rAAV in primary human hepatocytes and nonparenchymal liver cells. Surprisingly, Kupffer cells, but also liver sinusoidal endothelial cells, mounted responses to rAAV, whereas neither rAAV2 nor rAAV8 were recognized by hepatocytes. Viral capsids were sensed at the cell surface as pathogen-associated molecular patterns by Toll-like receptor 2. In contrast to the Toll-like receptor 9-mediated recognition observed in plasmacytoid dendritic cells, immune recognition of rAAV in primary human liver cells did not induce a type I interferon response, but up-regulated inflammatory cytokines through activation of nuclear factor κB. CONCLUSION Using primary human liver cells, we identified a novel mechanism of rAAV recognition in the liver, demonstrating that alternative means of sensing rAAV particles have evolved. Minimizing this recognition will be key to improving rAAV-mediated gene transfer and reducing side effects in clinical trials due to immune responses against rAAV.
Collapse
Affiliation(s)
- Marianna Hösel
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Raus S, Coin S, Monsurrò V. Adenovirus as a new agent for multiple myeloma therapies: Opportunities and restrictions. THE KOREAN JOURNAL OF HEMATOLOGY 2011; 46:229-38. [PMID: 22259628 PMCID: PMC3259514 DOI: 10.5045/kjh.2011.46.4.229] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 12/19/2011] [Indexed: 01/01/2023]
Abstract
Multiple myeloma is a malignancy of B-cells that is characterized by the clonal expansion and accumulation of malignant plasma cells in the bone marrow. This disease remains incurable, and a median survival of 3-5 years has been reported with the use of current treatments. Viral-based therapies offer promising alternatives or possible integration with current therapeutic regimens. Among several gene therapy vectors and oncolytic agents, adenovirus has emerged as a promising agent, and it is already being used for the treatment of solid tumors in humans. The main concern with the clinical use of this vector has been its high immunogenicity; adenovirus is often able to induce a strong immune response in the host. Furthermore, new limitations in the efficacy of this therapy, intrinsic to the nature of tumor cells, have been recently observed. For example, our group showed a strong antiviral phenotype in vitro and in vivo in a subset of tumors, shedding new insights that may explain the partial failure of clinical trials based on this promising new therapy. In this review, we describe novel therapeutic approaches that implement viral-based treatments in hematological malignancies and address the novelty as well as the possible limitations of these new therapies, especially in the context of the use of adenoviral vectors for treating multiple myeloma.
Collapse
Affiliation(s)
- Svjetlana Raus
- Department of Pathology and Diagnostics, University of Verona Medical School, Verona, Italy
| | | | | |
Collapse
|
46
|
Bovine herpesvirus 1 protein bICP0 represses the transcription of bISG15 in fetal bovine lung cells. Virol Sin 2011; 26:403-8. [PMID: 22160940 DOI: 10.1007/s12250-011-3213-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 10/27/2011] [Indexed: 11/27/2022] Open
Abstract
The ubiquitin-like modifier bISG15 is an antiviral protein found in fetal bovine lung (FBL) cells. Bovine Herpesvirus 1(BHV-1), which is a viral pathogen of cattle, can infect FBL cells and induce cytopathic effects. Real-time PCR assays showed that BHV-1's infection could repress the basal or inducible transcription of bISG15 in FBL cells. It demonstrates that this repression effect depends on BHV-1 viral infection and new protein synthesis. Our previous work showed that bIRF-3 was the key factor in the stimulation of bISG15 in FBL cells, so the effect of BHV-1 viral protein on bIRF-3 activating the promoter of bISG15 was confirmed. The luciferase assay showed the BHV-1 viral protein bICP0 inhibited the activation of bISG15 promoter stimulated by bIRF-3. Taken together, our work suggested that BHV-1 had some molecular mechanism to resist the cellular bISG15's antiviral functions.
Collapse
|
47
|
A nonreplicating subunit vaccine protects mice against lethal Ebola virus challenge. Proc Natl Acad Sci U S A 2011; 108:20695-700. [PMID: 22143779 DOI: 10.1073/pnas.1117715108] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Ebola hemorrhagic fever is an acute and often deadly disease caused by Ebola virus (EBOV). The possible intentional use of this virus against human populations has led to design of vaccines that could be incorporated into a national stockpile for biological threat reduction. We have evaluated the immunogenicity and efficacy of an EBOV vaccine candidate in which the viral surface glycoprotein is biomanufactured as a fusion to a monoclonal antibody that recognizes an epitope in glycoprotein, resulting in the production of Ebola immune complexes (EICs). Although antigen-antibody immune complexes are known to be efficiently processed and presented to immune effector cells, we found that codelivery of the EIC with Toll-like receptor agonists elicited a more robust antibody response in mice than did EIC alone. Among the compounds tested, polyinosinic:polycytidylic acid (PIC, a Toll-like receptor 3 agonist) was highly effective as an adjuvant agent. After vaccinating mice with EIC plus PIC, 80% of the animals were protected against a lethal challenge with live EBOV (30,000 LD(50) of mouse adapted virus). Surviving animals showed a mixed Th1/Th2 response to the antigen, suggesting this may be important for protection. Survival after vaccination with EIC plus PIC was statistically equivalent to that achieved with an alternative viral vector vaccine candidate reported in the literature. Because nonreplicating subunit vaccines offer the possibility of formulation for cost-effective, long-term storage in biothreat reduction repositories, EIC is an attractive option for public health defense measures.
Collapse
|
48
|
The cellular TAR RNA binding protein, TRBP, promotes HIV-1 replication primarily by inhibiting the activation of double-stranded RNA-dependent kinase PKR. J Virol 2011; 85:12614-21. [PMID: 21937648 DOI: 10.1128/jvi.05240-11] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The TAR RNA binding protein, TRBP, is a cellular double-stranded RNA (dsRNA) binding protein that can promote the replication of HIV-1 through interactions with the viral TAR element as well as with cellular proteins that affect the efficiency of translation of viral transcripts. The structured TAR element, present on all viral transcripts, can impede efficient translation either by sterically blocking access of translation initiation factors to the 5'-cap or by activating the dsRNA-dependent kinase, PKR. Several mechanisms by which TRBP can facilitate translation of viral transcripts have been proposed, including the binding and unwinding of TAR and the suppression of PKR activation. Further, TRBP has been identified as a cofactor of Dicer in the processing of microRNAs (miRNAs), and sequestration of TRBP by TAR in infected cells has been proposed as a viral countermeasure to potential host cell RNA interference-based antiviral activities. Here, we have addressed the relative importance of these various roles for TRBP in HIV-1 replication. Using Jurkat T cells, primary human CD4(+) T cells, and additional cultured cell lines, we show that depletion of TRBP has no effect on viral replication when PKR activation is otherwise blocked. Moreover, the presence of TAR-containing mRNAs does not affect the efficacy of cellular miRNA silencing pathways. These results establish that TRBP, when expressed at physiological levels, promotes HIV-1 replication mainly by suppressing the PKR-mediated antiviral response, while its contribution to HIV-1 replication through PKR-independent pathways is minimal.
Collapse
|
49
|
Rácz Z, Godó M, Révész C, Hamar P. Immune activation and target organ damage are consequences of hydrodynamic treatment but not delivery of naked siRNAs in mice. Nucleic Acid Ther 2011; 21:215-24. [PMID: 21749298 PMCID: PMC3198622 DOI: 10.1089/nat.2010.0248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 04/12/2011] [Indexed: 12/22/2022] Open
Abstract
Short-interfering RNAs (siRNAs), key mediators of RNA interference comprise a promising therapeutic tool, although side effects such as interferon (IFN) response are still not perfectly understood. Further, delivery to target organs is a major challenge, possibly associated with side effects including immune activation or organ damage. We investigated whether immune activation as a consequence of double-stranded RNA induced IFN response (Jak/STAT pathway activation or cytokine production) or target organ damage is induced by in vivo low-volume (LV) or high-volume (HV) hydrodynamic delivery or treatment with naked siRNA. NMRI mice were injected with naked siRNAs or saline by hydrodynamic injection (HDI) and positive control mice received polyinosinic-polycytidilic acid (poly I:C). LV (1 mL/mouse) and HV (10% of body weight) HDI were compared. After LV HDI, STAT1 and OAS1 gene expression inflammatory cytokine plasma levels and target organ injury were assessed. LV HDI induced slight alanine aminotransferase elevation and mild hepatocyte injury, whereas HV HDI resulted in high ALAT level and extensive hepatocyte necrosis. STAT1 or OAS1 was not induced by LV siRNA; however, HV saline led to a time-dependent slight increase in gene expression. Inflammatory cytokine plasma level and organ histology and functional parameters demonstrated no damage following LV HDI with or without siRNA. Our data demonstrate that naked siRNAs may be harnessed, without the induction of IFN response or immune activation, and that LV HDI is preferable, because HV HDI may cause organ damage.
Collapse
Affiliation(s)
- Zsuzsanna Rácz
- Faculty of Medicine, Institute of Pathophysiology, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
| | | | | | | |
Collapse
|
50
|
Levitzki A, Shir A. Targeting the immune system to cancer. POLYM ADVAN TECHNOL 2011. [DOI: 10.1002/pat.1857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|