Norovirus Replication in Human Intestinal Epithelial Cells Is Restricted by The

Total Page:16

File Type:pdf, Size:1020Kb

Norovirus Replication in Human Intestinal Epithelial Cells Is Restricted by The bioRxiv preprint doi: https://doi.org/10.1101/731802; this version posted August 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 Norovirus replication in human intestinal epithelial cells is restricted by the 2 interferon-induced JAK/STAT signalling pathway and RNA Polymerase II 3 mediated transcriptional responses. 4 5 Myra Hosmilloa#, Yasmin Chaudhrya, Komal Nayakb , Frederic Sorgeloosa, Bon- 6 Kyoung Kooc,d, Alessandra Merendac*, Reidun Lillestole, Lydia Drumrighte, 7 Matthias Zilbauerb and Ian Goodfellowa# 8 9 a Division of Virology, Department of Pathology, University of Cambridge, 10 Cambridge, UK 11 b Department of Paediatrics, University of Cambridge, Cambridge, UK 12 c Wellcome Trust-Medical Research Council Stem Cell Institute, University of 13 Cambridge, Cambridge, UK 14 d Institute of Molecular Biotechnology of the Austrian Academy of Sciences 15 (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria. 16 e Department of Medicine, Addenbrooke’s Hospital, University of Cambridge, 17 Cambridge, UK 18 19 Running title: Restriction of HuNoV infection in intestinal mucosa 20 21 #Address correspondence to Ian Goodfellow, [email protected] and Myra 22 Hosmillo, [email protected] 23 *Present address: bioRxiv preprint doi: https://doi.org/10.1101/731802; this version posted August 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 24 Alessandra Merenda, Oncode Institute and Department of Cell Biology, Centre 25 for Molecular Medicine, University Medical Centre Utrecht, 3584 CX Utrecht, the 26 Netherlands 27 28 Abstract: 217 words 29 Text: 7,105 words 30 bioRxiv preprint doi: https://doi.org/10.1101/731802; this version posted August 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 31 Abstract 32 Human noroviruses (HuNoV) are a leading cause of viral gastroenteritis 33 worldwide and a significant cause of morbidity and mortality in all age groups. 34 The recent finding that HuNoV can be propagated in B cells and mucosa derived 35 intestinal epithelial organoids (IEOs), has transformed our capability to dissect 36 the life cycle of noroviruses. Using RNA-Seq of HuNoV infected intestinal 37 epithelial cells (IECs), we have found that replication of HuNoV in IECs results in 38 interferon-induced transcriptional responses and that HuNoV replication in IECs 39 is sensitive to IFN. This contrasts with previous studies that suggest that the 40 innate immune response may play no role in the restriction of HuNoV replication 41 in immortalised cells. We demonstrate that the inhibition of JAK1/JAK2 42 enhances HuNoV replication in IECs. Surprisingly, targeted inhibition of cellular 43 RNA polymerase II-mediated transcription was not detrimental to HuNoV 44 replication, but enhanced replication to a greater degree compared to blocking 45 of JAK signalling directly. Furthermore, we demonstrate for the first time that 46 IECs generated from genetically modified intestinal organoids, engineered to 47 be deficient in the interferon response, are more permissive to HuNoV 48 infection. Together our work identifies the IFN-induced transcriptional 49 responses restrict HuNoV replication in IECs and demonstrates that the 50 inhibition of these responses by modifications to the culture conditions can 51 greatly enhance the robustness of the norovirus culture system. 52 53 bioRxiv preprint doi: https://doi.org/10.1101/731802; this version posted August 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 54 Importance 55 Noroviruses are a major cause of gastroenteritis worldwide yet the challenges 56 associated with their growth culture has greatly hampered the development of 57 therapeutic approaches and has limited our understanding of cellular pathways 58 that control infection. Here we show that human intestinal epithelial cells, the 59 first point of entry of human noroviruses into the host, limit virus replication by 60 the induction of the innate responses. Furthermore we show that modulating 61 the ability of intestinal epithelial cells to induce transcriptional responses to 62 HuNoV infection can significantly enhance human norovirus replication in 63 culture. Collectively our findings provide new insights into the biological 64 pathways that control norovirus infection but also identify mechanisms to 65 enhance the robustness of norovirus culture. 66 67 bioRxiv preprint doi: https://doi.org/10.1101/731802; this version posted August 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 68 Introduction 69 The induction of the host innate response plays an essential role in the 70 suppression of pathogen infection. The synthesis of interferons (IFN) and the 71 subsequent signalling cascades that leads to the induction of IFN-stimulated 72 genes (ISGs), determine the outcome of viral infection (1, 2). An understanding of 73 the mechanisms underlying the interplay between pathogens and innate immune 74 responses is vital to understanding viral pathogenesis and can greatly aid the 75 identification of potential therapeutic and/or preventive strategies. 76 77 Human noroviruses (HuNoV) are widely recognised as the leading cause of viral 78 gastroenteritis worldwide (3). Noroviruses are classified into at least seven 79 genogroups based on the sequence of the major capsid protein VP1 and regions 80 within ORF1 (3–5). HuNoVs belong to one of three norovirus genogroups (GI, 81 GII, or GIV), which are further divided into >25 genetic clusters or genotypes (6– 82 8). Epidemiological studies reveal that over 75% of confirmed human norovirus 83 infections are associated with HuNoV GII (9, 10). Whilst norovirus gastroenteritis 84 typically results in an acute and self-limiting disease, the socioeconomic impact in 85 both developed and developing countries is estimated to be more than $60.3 86 billion per annum (11). HuNoV infection is particularly severe and prolonged in 87 immunocompromised patients, including young children, elderly, or patients 88 receiving treatment for cancer. In these cases infections can last from months to 89 years (12, 13). 90 bioRxiv preprint doi: https://doi.org/10.1101/731802; this version posted August 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 91 Our understanding of the molecular mechanisms that control HuNoV infection 92 has been limited by the lack of robust culture systems that facilitate the detailed 93 analysis of the viral life cycle. As a result, murine norovirus (MNV) and other 94 members of the Caliciviridae family of positive sense RNA viruses, such as feline 95 calicivirus (FCV) and porcine sapovirus (PSaV), are often used as surrogate 96 models (14–17). MNV, FCV and PSaV can all be efficiently cultured in 97 immortalised cells and are amenable to reverse genetics (16–20). These model 98 systems have been critical to understanding many aspects of the life cycle of 99 members of the Caliciviridae (15). 100 101 Recent efforts have led to the establishment of two HuNoV culture systems 102 based on immortalised B cells (21, 22) and intestinal epithelial cells (IECs) 103 generated from biopsy-derived human intestinal epithelial organoids (IEOs) (23). 104 Whilst authentic replication of HuNoV can be observed in both the B-cell and 105 IEC-based culture systems, repeated long-term passage of HuNoV and the 106 generation of high titre viral stocks is not possible, suggesting that replication is 107 restricted in some manner. In the current study we sought to better understand 108 the cellular response to HuNoV infection and to identify pathways that restrict 109 HuNoV replication in organoid-derived IECs. Using RNA-Seq we observed that 110 HuNoV infection of IECs results in an interferon-mediated antiviral transcriptional 111 response. We show for the first time that HuNoV replication in IECs is sensitive to 112 both Type I and III interferon and that HuNoV replication is restricted by virus- 113 induced innate response. Pharmacological inhibition of the interferon response 114 or genetic modification of organoids to prevent the activation of the interferon bioRxiv preprint doi: https://doi.org/10.1101/731802; this version posted August 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 115 response significantly improved HuNoV replication in IECs. Furthermore, we 116 show that ongoing HuNoV replication is enhanced by the inhibition of RNA Pol II 117 mediated transcription. Overall this work provides new insights into the cellular 118 responses to HuNoV infection of the gut epithelium and identifies modifications to 119 the HuNoV culture system that significantly enhances its utility. 120 121 122 Materials and Methods. 123 Stool samples. Stool specimens were anonymized with written consent from 124 patients at Addenbrooke’s Hospital, Cambridge, who tested positive of HuNoV 125 infection.
Recommended publications
  • The Role of Type I Interferon in the Immunobiology of Chikungunya Virus
    The role of type I interferon in the immunobiology of chikungunya virus Jane Amelia Clare Wilson B. App. Sc. (Human Biology), B. App. Sc. (Hons) A thesis submitted for the degree of Doctor of Philosophy at The University of Queensland in 2015 School of Medicine & QIMR Berghofer Medical Research Institute I Abstract Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that can cause explosive outbreaks of a febrile, arthritic/arthralgic disease usually lasting weeks to months, and in rare cases, more than a year. In 2004, the largest ever CHIKV outbreak began in Kenya, spreading to islands of the Indian Ocean, India, South East Asia and major outbreaks have recently occurred in the South Pacific Islands and the Caribbean. The host type I interferon (IFN) response is crucial for effective control of CHIKV infection. Herein, the dynamics, source and responses generated by the type I IFNs following CHIKV infection were investigated. Interferon regulatory factors 3 (IRF3) and IRF7 are key transcription factors for the type I IFN response. While CHIKV infection of wild-type mice is non-lethal, infection of mice deficient in both IRF3 and IRF7 (IRF3/7-/-) resulted in mortality, illustrating that these factors are essential for protection. Using knockout mice for the adaptor molecules upstream of IRF3 and 7, IPS1 was found to be the most important for type I IFN production, with TRIF and MyD88 also contributing to the response. Mortality in IRF3/7-/- mice was also associated with type I IFN suppression of pathological levels of IFNγ and haemorrhagic shock. Heterozygous reporter mice, in which eGFP was expressed under the control of either the IFNβ or the IFNα6 promoter on one chromosome, were employed to try and identify the cellular source of type I IFN production following CHIVK infection.
    [Show full text]
  • Stimulation of Innate and Adaptive Immunity by Using Filamentous Bacteriophage Fd Targeted to DEC-205
    Hindawi Publishing Corporation Journal of Immunology Research Volume 2015, Article ID 585078, 11 pages http://dx.doi.org/10.1155/2015/585078 Research Article Stimulation of Innate and Adaptive Immunity by Using Filamentous Bacteriophage fd Targeted to DEC-205 Luciana D’Apice,1 Valerio Costa,2 Rossella Sartorius,1 Maria Trovato,1 Marianna Aprile,2 and Piergiuseppe De Berardinis1 1 Institute of Protein Biochemistry (IBP), National Council of Research, 80131 Naples, Italy 2Institute of Genetics and Biophysics “A. Buzzati-Traverso” (IGB), National Council of Research, 80131 Naples, Italy Correspondence should be addressed to Luciana D’Apice; [email protected] Received 26 March 2015; Accepted 27 May 2015 Academic Editor: L. Leite Copyright © 2015 Luciana D’Apice et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The filamentous bacteriophage fd, codisplaying antigenic determinants and a single chain antibody fragment directed against the dendritic cell receptor DEC-205, is a promising vaccine candidate for its safety and its ability to elicit innate and adaptive immune response in absence of adjuvants. By using a system vaccinology approach based on RNA-Sequencing (RNA-Seq) analysis, we describe a relevant gene modulation in dendritic cells pulsed with anti-DEC-205 bacteriophages fd. RNA-Seq data analysis indicates that the bacteriophage fd virions are sensed as a pathogen by dendritic cells; they activate the danger receptors that trigger an innate immune response and thus confer a strong adjuvanticity that is needed to obtain a long-lasting adaptive immune response.
    [Show full text]
  • Type I Interferon Remodels Lysosome Function and Modifies Intestinal Epithelial Defense
    Type I interferon remodels lysosome function and modifies intestinal epithelial defense Hailong Zhanga,b,c, Abdelrahim Zoueda,b,c, Xu Liua,b,c, Brandon Sitb,c, and Matthew K. Waldora,b,c,1 aHoward Hughes Medical Insitute, Boston, MA 02115; bDivision of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA 02115; and cDepartment of Microbiology, Harvard Medical School, Boston, MA 02115 Edited by Jorge E. Galán, Yale University, New Haven, CT, and approved October 14, 2020 (received for review May 29, 2020) Organelle remodeling is critical for cellular homeostasis, but host the full spectrum of IFN-I-mediated changes in cellular function factors that control organelle function during microbial infection is incomplete. Although IFN-Is are known to play critical roles in remain largely uncharacterized. Here, a genome-scale CRISPR/Cas9 antiviral responses, their functions in bacterial infection are less screen in intestinal epithelial cells with the prototypical intracellu- clear, and IFN-I signaling has been reported to be either pro- lar bacterial pathogen Salmonella led us to discover that type I IFN tective or detrimental to the host depending on the specific (IFN-I) remodels lysosomes. Even in the absence of infection, IFN-I bacterial pathogen (19). signaling modified the localization, acidification, protease activity, Here, we carried out a genome-scale CRISPR/Cas9 screen to and proteomic profile of lysosomes. Proteomic and genetic analyses identify the host factors that contribute to Stm’s cytotoxicity to revealed that multiple IFN-I–stimulated genes including IFITM3, SLC15A3, IECs. This screen revealed IFN-I signaling as a key susceptibility and CNP contribute to lysosome acidification.
    [Show full text]
  • Microglial Responses to Peripheral Type 1 Interferon Ernest Aw1,2, Yingying Zhang1 and Michael Carroll1*
    Aw et al. Journal of Neuroinflammation (2020) 17:340 https://doi.org/10.1186/s12974-020-02003-z RESEARCH Open Access Microglial responses to peripheral type 1 interferon Ernest Aw1,2, Yingying Zhang1 and Michael Carroll1* Abstract Background: Interferon α (IFNα) is a cytokine whose production is increased endogenously in response to viral infection and in autoimmune diseases such as systemic lupus erythematosus (SLE). An elevated IFNα signature has been associated with clinically observed neuro-behavioural deficits such as mild cognitive impairment, fatigue, depression and psychosis in these diseases. However, the mechanisms underlying these neuropsychiatric symptoms remain largely unknown, and it is as yet unclear how IFNα signalling might influence central nervous system (CNS) function. Aberrant microglia-mediated synaptic pruning and function has recently been implicated in several neurodegenerative and neuropsychiatric diseases, but whether and how IFNα modulates these functions are not well defined. Methods: Using a model of peripheral IFNα administration, we investigated gene expression changes due to IFNAR signalling in microglia. Bulk RNA sequencing on sorted microglia from wild type and microglia-specific Ifnar1 conditional knockout mice was performed to evaluate IFNα and IFNAR signalling-dependent changes in gene expression. Furthermore, the effects of IFNα on microglia morphology and synapse engulfment were assessed, via immunohistochemistry and flow cytometry. Results: We found that IFNα exposure through the periphery induces a unique gene signature in microglia that includes the expected upregulation of multiple interferon-stimulated genes (ISGs), as well as the complement component C4b. We additionally characterized several IFNα-dependent changes in microglial phenotype, including expression of CD45 and CD68, cellular morphology and presynaptic engulfment, that reveal subtle brain region- specific differences.
    [Show full text]
  • Combined Genetic and Transcriptome Analysis of Patients With
    Systemic lupus erythematosus Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214379 on 5 June 2019. Downloaded from TRANSLATIONAL SCIENCE Combined genetic and transcriptome analysis of patients with SLE: distinct, targetable signatures for susceptibility and severity Nikolaos I Panousis, 1,2,3 George K Bertsias, 4,5 Halit Ongen,1,2,3 Irini Gergianaki,4,5 Maria G Tektonidou, 6,7 Maria Trachana,8 Luciana Romano-Palumbo,1 Deborah Bielser,1 Cedric Howald,1,2,3 Cristina Pamfil,9 Antonis Fanouriakis, 10 Despoina Kosmara,4,5 Argyro Repa,4 Prodromos Sidiropoulos,4,5 Emmanouil T Dermitzakis,1,2,3,11 Dimitrios T Boumpas5,7,10,11,12 Handling editor Josef S ABSTRact Key messages Smolen Objectives Systemic lupus erythematosus (SLE) diagnosis and treatment remain empirical and the ► Additional material is What is already known about this subject? published online only. To view molecular basis for its heterogeneity elusive. We explored ► Previous DNA microarray gene expression please visit the journal online the genomic basis for disease susceptibility and severity. studies have identified gene signatures (http:// dx. doi. org/ 10. 1136/ Methods mRNA sequencing and genotyping in blood involved in systemic lupus erythematosus (SLE) annrheumdis- 2018- 214379). from 142 patients with SLE and 58 healthy volunteers. such as those linked to granulocytes, pattern Abundances of cell types were assessed by CIBERSORT For numbered affiliations see recognition receptors, type I interferon and and cell-specific effects by interaction terms in linear end of article. plasmablasts. models. Differentially expressed genes (DEGs) were used Correspondence to to train classifiers (linear discriminant analysis) of SLE What does this study add? Professor Emmanouil T versus healthy individuals in 80% of the dataset and A more comprehensive profiling of the ‘genomic Dermitzakis, Department of were validated in the remaining 20% running 1000 ► Genetic Medicine and architecture’ of SLE by combining genetic and iterations.
    [Show full text]
  • An Inflammatory Gene Signature Distinguishes Neurofibroma
    www.nature.com/scientificreports OPEN An inflammatory gene signature distinguishes neurofibroma Schwann cells and macrophages Received: 26 May 2016 Accepted: 25 January 2017 from cells in the normal peripheral Published: 03 March 2017 nervous system Kwangmin Choi1, Kakajan Komurov1, Jonathan S. Fletcher1, Edwin Jousma1, Jose A. Cancelas1,2, Jianqiang Wu1 & Nancy Ratner1 Neurofibromas are benign peripheral nerve tumors driven byNF1 loss in Schwann cells (SCs). Macrophages are abundant in neurofibromas, and macrophage targeted interventions may have therapeutic potential in these tumors. We generated gene expression data from fluorescence- activated cell sorted (FACS) SCs and macrophages from wild-type and mutant nerve and neurofibroma to identify candidate pathways involved in SC-macrophage cross-talk. While in 1-month-old Nf1 mutant nerve neither SCs nor macrophages significantly differed from their normal counterparts, both macrophages and SCs showed significantly altered cytokine gene expression in neurofibromas. Computationally reconstructed SC-macrophage molecular networks were enriched for inflammation- associated pathways. We verified that neurofibroma SC conditioned medium contains macrophage chemo-attractants including colony stimulation factor 1 (CSF1). Network analysis confirmed previously implicated pathways and predict novel paracrine and autocrine loops involving cytokines, chemokines, and growth factors. Network analysis also predicted a central role for decreased type-I interferon signaling. We validated type-I interferon expression in neurofibroma by protein profiling, and show that treatment of neurofibroma-bearing mice with polyethylene glycolyated (PEGylated) type-I interferon- α2b reduces the expression of many cytokines overexpressed in neurofibroma. These studies reveal numerous potential targetable interactions between Nf1 mutant SCs and macrophages for further analyses. Neurofibromatosis type 1 (NF1) is one of the most common human monogenic disorders, affecting about 0.3% of the human population.
    [Show full text]
  • (Hies) Infected with Enteric RNA Viruses Identifies Universal and Virus-Specific Epithelial Responses
    viruses Article Comparative Analysis of Public RNA-Sequencing Data from Human Intestinal Enteroid (HIEs) Infected with Enteric RNA Viruses Identifies Universal and Virus-Specific Epithelial Responses Roberto J. Cieza 1,2 , Jonathan L. Golob 2, Justin A. Colacino 3,4 and Christiane E. Wobus 1,* 1 Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA; [email protected] 2 Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA; [email protected] 3 Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA; [email protected] 4 Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA * Correspondence: [email protected] Abstract: Acute gastroenteritis (AGE) has a significant disease burden on society. Noroviruses, rotaviruses, and astroviruses are important viral causes of AGE but are relatively understudied Citation: Cieza, R.J.; Golob, J.L.; enteric pathogens. Recent developments in novel biomimetic human models of enteric disease are Colacino, J.A.; Wobus, C.E. opening new possibilities for studying human-specific host–microbe interactions. Human intestinal Comparative Analysis of Public enteroids (HIE), which are epithelium-only intestinal organoids derived from stem cells isolated from RNA-Sequencing Data from Human human intestinal biopsy tissues, have been successfully used to culture representative norovirus, Intestinal Enteroid (HIEs) Infected rotavirus, and astrovirus strains. Previous studies investigated host–virus interactions at the intestinal with Enteric RNA Viruses Identifies epithelial interface by individually profiling the epithelial transcriptional response to a member of Universal and Virus-Specific each virus family by RNA sequencing (RNA-seq).
    [Show full text]
  • SNP) at Codon 72 of P53 in the Innate Immune Response Against Virus Infection
    International Journal of Molecular Sciences Review SARS-CoV-2: Understanding the Transcriptional Regulation of ACE2 and TMPRSS2 and the Role of Single Nucleotide Polymorphism (SNP) at Codon 72 of p53 in the Innate Immune Response against Virus Infection Niraj Lodhi 1,* , Rubi Singh 2, Satya Prakash Rajput 3 and Quaiser Saquib 4 1 Clinical Research (Research and Development Division) miRNA Analytics LLC, Harlem Bio-Space, New York, NY 10027, USA 2 Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA; [email protected] 3 School of Life Sciences, Warwick University, Coventry CV47AL, UK; [email protected] 4 Department of Zoology, College of Sciences, King Saud University, Riyadh 12372, Saudi Arabia; [email protected] * Correspondence: [email protected] Abstract: Human ACE2 and the serine protease TMPRSS2 of novel SARS-CoV-2 are primary entry receptors in host cells. Expression of these genes at the transcriptional level has not been much discussed in detail. The ISRE elements of the ACE2 promoter are a binding site for the ISGF3 complex of the JAK/STAT signaling pathway. TMPRSS2, including IFNβ, STAT1, and STAT2, has the PARP1 Citation: Lodhi, N.; Singh, R.; Rajput, binding site near to TSS either up or downstream promoter region. It is well documented that PARP1 S.P.; Saquib, Q. SARS-CoV-2: regulates gene expression at the transcription level. Therefore, to curb virus infection, both promoting Understanding the Transcriptional type I IFN signaling to boost innate immunity and prevention of virus entry by inhibiting PARP1, Regulation of ACE2 and TMPRSS2 ACE2 or TMPRSS2 are safe options.
    [Show full text]
  • Influenza Infection Triggers Disease in a Genetic Model of Experimental
    Influenza infection triggers disease in a genetic model PNAS PLUS of experimental autoimmune encephalomyelitis Stephen Blackmorea, Jessica Hernandeza, Michal Judaa, Emily Ryderb, Gregory G. Freunda,c, Rodney W. Johnsona,b,d, and Andrew J. Steelmana,b,d,1 aDepartment of Animal Sciences, University of Illinois Urbana–Champaign, Urbana, IL 61801; bNeuroscience Program, University of Illinois Urbana–Champaign, Urbana, IL 61801; cDepartment of Pathology, University of Illinois Urbana–Champaign, Urbana, IL 61801; and dDivision of Nutritional Sciences, University of Illinois Urbana–Champaign, Urbana, IL 61801 Edited by Lawrence Steinman, Stanford University School of Medicine, Stanford, CA, and approved June 13, 2017 (received for review December 13, 2016) Multiple sclerosis (MS) is an autoimmune disease of the central affect the progression of many neurological diseases including nervous system. Most MS patients experience periods of symptom Alzheimer’s disease (10, 11), Parkinson’s disease (12), and exacerbation (relapses) followed by periods of partial recovery multiple sclerosis (13–17). (remission). Interestingly, upper-respiratory viral infections increase We are interested in determining how upper-respiratory in- the risk for relapse. Here, we used an autoimmune-prone T-cell fection contributes to the progression of neurological dis- receptor transgenic mouse (2D2) and a mouse-adapted human eases including multiple sclerosis (MS), the most prominent influenza virus to test the hypothesis that upper-respiratory viral autoimmune-mediated demyelinating and neurodegenerating infection can cause glial activation, promote immune cell trafficking disease of the CNS. The majority of MS patients exhibit an os- to the CNS, and trigger disease. Specifically, we inoculated 2D2 mice cillating disease course that is characterized by relatively short with influenza A virus (Puerto Rico/8/34; PR8) and then monitored periods of neurological dysfunction followed by periods of re- them for symptoms of inflammatory demyelination.
    [Show full text]
  • IFN-Stimulated Genes in Zebrafish and Humans Define an Ancient Arsenal of Antiviral Immunity
    IFN-Stimulated Genes in Zebrafish and Humans Define an Ancient Arsenal of Antiviral Immunity This information is current as Jean-Pierre Levraud, Luc Jouneau, Valérie Briolat, Valerio of September 23, 2021. Laghi and Pierre Boudinot J Immunol published online 15 November 2019 http://www.jimmunol.org/content/early/2019/11/14/jimmun ol.1900804 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2019/11/15/jimmunol.190080 Material 4.DCSupplemental http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on September 23, 2021 *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2019 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published November 15, 2019, doi:10.4049/jimmunol.1900804 The Journal of Immunology IFN-Stimulated Genes in Zebrafish and Humans Define an Ancient Arsenal of Antiviral Immunity Jean-Pierre Levraud,* Luc Jouneau,† Vale´rie Briolat,* Valerio Laghi,* and Pierre Boudinot† The evolution of the IFN system, the major innate antiviral mechanism of vertebrates, remains poorly understood.
    [Show full text]
  • Transcriptional Dysregulation of Interferome in Experimental and Human Multiple Sclerosis
    www.nature.com/scientificreports Correction: Author Correction OPEN Transcriptional dysregulation of Interferome in experimental and human Multiple Sclerosis Received: 23 May 2017 Sundararajan Srinivasan1,2, Martina Severa3, Fabiana Rizzo3, Ramesh Menon1, Elena Brini1, Accepted: 18 July 2017 Rosella Mechelli4, Vittorio Martinelli1, Paul Hertzog5, Marco Salvetti4,6, Roberto Furlan 1, Published: xx xx xxxx Gianvito Martino 1,2, Giancarlo Comi1,2, Eliana M. Coccia3 & Cinthia Farina1 Recent evidence indicates that single multiple sclerosis (MS) susceptibility genes involved in interferon (IFN) signaling display altered transcript levels in peripheral blood of untreated MS subjects, suggesting that responsiveness to endogenous IFN is dysregulated during neuroinfammation. To prove this hypothesis we exploited the systematic collection of IFN regulated genes (IRG) provided by the Interferome database and mapped Interferome changes in experimental and human MS. Indeed, central nervous system tissue and encephalitogenic CD4 T cells during experimental autoimmune encephalomyelitis were characterized by massive changes in Interferome transcription. Further, the analysis of almost 500 human blood transcriptomes showed that (i) several IRG changed expression at distinct MS stages with a core of 21 transcripts concordantly dysregulated in all MS forms compared with healthy subjects; (ii) 100 diferentially expressed IRG were validated in independent case-control cohorts; and (iii) 53 out of 100 dysregulated IRG were targeted by IFN-beta treatment in vivo. Finally, ex vivo and in vitro experiments established that IFN-beta administration modulated expression of two IRG, ARRB1 and CHP1, in immune cells. Our study confrms the impairment of Interferome in experimental and human MS, and describes IRG signatures at distinct disease stages which can represent novel therapeutic targets in MS.
    [Show full text]
  • Chicken Interferome: Avian Interferon-Stimulated Genes Identified by Microarray and RNA-Seq of Primary Chick Embryo Fibroblasts
    Giotis et al. Vet Res (2016) 47:75 DOI 10.1186/s13567-016-0363-8 RESEARCH ARTICLE Open Access Chicken interferome: avian interferon‑stimulated genes identified by microarray and RNA‑seq of primary chick embryo fibroblasts treated with a chicken type I interferon (IFN‑α) Efstathios S. Giotis1†, Rebecca C. Robey1†, Natalie G. Skinner2, Christopher D. Tomlinson3, Stephen Goodbourn4 and Michael A. Skinner1* Abstract Viruses that infect birds pose major threats—to the global supply of chicken, the major, universally-acceptable meat, and as zoonotic agents (e.g. avian influenza viruses H5N1 and H7N9). Controlling these viruses in birds as well as understanding their emergence into, and transmission amongst, humans will require considerable ingenuity and understanding of how different species defend themselves. The type I interferon-coordinated response constitutes the major antiviral innate defence. Although interferon was discovered in chicken cells, details of the response, par- ticularly the identity of hundreds of stimulated genes, are far better described in mammals. Viruses induce interferon- stimulated genes but they also regulate the expression of many hundreds of cellular metabolic and structural genes to facilitate their replication. This study focusses on the potentially anti-viral genes by identifying those induced just by interferon in primary chick embryo fibroblasts. Three transcriptomic technologies were exploited: RNA-seq, a classi- cal 3′-biased chicken microarray and a high density, “sense target”, whole transcriptome chicken microarray, with each recognising 120–150 regulated genes (curated for duplication and incorrect assignment of some microarray probe- sets). Overall, the results are considered robust because 128 of the compiled, curated list of 193 regulated genes were detected by two, or more, of the technologies.
    [Show full text]