1
|
Yang HW, Hu T, Ait-Ali T. Lawsonia intracellularis regulates nuclear factor-κB signalling pathway during infection. PLoS One 2024; 19:e0310804. [PMID: 39325775 PMCID: PMC11426430 DOI: 10.1371/journal.pone.0310804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Lawsonia intracellularis is the etiological agent of proliferative enteropathy (PE) in pigs, horses and wide range of mammals. Little is known about the role of innate immune response during L. intracellularis infection. In this study, we investigated the nuclear factor-κB (NF-κB)-regulated immune response against infection of a clinical strain Dkp23 and a live-attenuated Enterisol vaccine strain in PK-15 cells. We found that expression of NF-κB target genes TNF-α, IFN-γ, IL-6 and IL-8 were modulated during the course of infection. At 5 dpi, there was a significant increase in p65 NF-κB activation, including protein nuclear translocation and phosphorylation, synchronous with the induction of IL-6, IFN-γ and IL-8 expression in L. intracellularis infected cells, especially for Enterisol vaccine strain-infected cells. This result suggests that NF-κB signalling level is induced when L. intracellularis bacterial load peaks at 5 dpi. The induction of pro-inflammatory cytokines expression is consistent with the decreased viability of L. intracellularis-infected cells especially that of the vaccine strain. There were no significant changes in NF-κB signalling between vaccine and Dkp23 infection in PK-15 cells, except for moderate levels of differences in NF-κB target genes expression which might be a reflection of differences in intracellular bacterial load. Overall, the data presented here indicate a correlation between the induction of NF-κB signalling and the L. intracellularis bacterial load in PK-15 cells.
Collapse
Affiliation(s)
- Huan W Yang
- Department of Biochemistry, The University of Illinois Champaign-Urbana, Champaign, IL, United States of America
| | - Tuanjun Hu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | | |
Collapse
|
2
|
Batselova HM, Velikova TV. Ambispective epidemiological observational study of varicella-zoster virus infection: An 18 year-single-center Bulgarian experience. World J Virol 2024; 13:92525. [PMID: 39323443 PMCID: PMC11401008 DOI: 10.5501/wjv.v13.i3.92525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/20/2024] [Accepted: 07/04/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Varicella (chickenpox) and herpes zoster (shingles) are outcomes of varicella-zoster virus (VZV) infection, and understanding their incidence trends is vital for public health planning. AIM To conduct an ambispective epidemiological study by analyzing the main epidemiological characteristics of VZV infection during an 18 year-period (2000-2018). METHODS We used descriptive and epidemiological methods to characterize chickenpox in Bulgaria, the city of Plovdiv and the region for a period of 18 years (2000-2018). RESULTS The average incidence of varicella-zoster infection for the period 2000-2018 in the Plovdiv region was estimated at 449.58‰. The highest relative share of the infection was assessed in the month of January at 13.6%, and the lowest in the months of August and September at 2.9% (both months). The age group most affected by the infection was 1-4 years, followed by 5-9 years. This corresponds to the so-called "pro-epidemic population" - a phenomenon typical for airborne infections, confirming their mass impact on the perpetuation of VZV infection. CONCLUSION Our findings reveal significant insights into VZV epidemiology, including age-specific incidence rates, clinical manifestations, and vaccination impact. This comprehensive analysis contributes to the broader understanding of VZV infection dynamics and may inform evidence-based preventive measures.
Collapse
Affiliation(s)
- Hristiana M Batselova
- Department of Epidemiology and Disaster Medicine, Medical University, Plovdiv, University Hospital “St George”, Plovdiv 4000, Bulgaria
| | | |
Collapse
|
3
|
Bo Y, Zhong X, Xiang Y, Ren Q, Hao P. Death Caused by Disseminated Herpes Zoster in a Patient with Multiple Myeloma: A Case Report and Literature Review. Clin Cosmet Investig Dermatol 2024; 17:941-951. [PMID: 38707610 PMCID: PMC11066659 DOI: 10.2147/ccid.s464039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024]
Abstract
We report a fatal case of disseminated herpes zoster in a patient with multiple myeloma, illustrating the severe risks immunocompromised individuals face from viral infections. By combining a detailed case report with an extensive literature review, the paper seeks to shed light on the underlying susceptibility factors for varicella-zoster virus infection in multiple myeloma patients. We further evaluate effective prophylactic protocols for herpes zoster, aiming to equip clinicians with improved therapeutic strategies. The case underscores the critical need for vigilant clinical assessments and tailored patient management to mitigate infection risks and enhance patient outcomes.
Collapse
Affiliation(s)
- Yang Bo
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiaojing Zhong
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Yanping Xiang
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Qingjun Ren
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Pingsheng Hao
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
4
|
Ferrara G, Longobardi C, Sgadari MF, Restucci B, Iovane G, Ciarcia R, Pagnini U, Montagnaro S. Apoptosis is mediated by FeHV-1 through the intrinsic pathway and interacts with the autophagic process. Virol J 2023; 20:295. [PMID: 38087282 PMCID: PMC10716993 DOI: 10.1186/s12985-023-02267-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/09/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Although FeHV-1 is a primary feline pathogen, little is known about its interactions with host cells. Its relationship with several cellular pathways has recently been described, whereas its interplay with the apoptotic process, unlike other herpesviruses, has not yet been clarified. The aim of this work was to evaluate whether FeHV-1 induces apoptosis in its permissive cells, as well as the pathway involved and the effects of induction and inhibition of apoptosis on viral replication. METHODS Monolayers of CRFK cells were infected at different times with different viral doses. A cytofluorimetric approach allowed the quantification of cells in early and late apoptosis. All infections and related controls were also subjected to Western blot analysis to assess the expression of apoptotic markers (caspase 3-8-9, Bcl-2, Bcl-xL, NF-κB). An inhibitor (Z-VAD-FMK) and an inducer (ionomycin) were used to evaluate the role of apoptosis in viral replication. Finally, the expression of autophagy markers during the apoptosis inhibition/induction and the expression of apoptosis markers during autophagy inhibition/induction were evaluated to highlight any crosstalk between the two pathways. RESULTS FeHV-1 triggered apoptosis in a time- and dose-dependent manner. Caspase 3 cleavage was evident 48 h after infection, indicating the completeness of the process at this stage. While caspase 8 was not involved, caspase 9 cleavage started 24 h post-infection. The expression of other mitochondrial damage markers also changed, suggesting that apoptosis was induced via the intrinsic pathway. NF- κB was up-regulated at 12 h, followed by a gradual decrease in levels up to 72 h. The effects of apoptosis inhibitors and inducers on viral replication and autophagy were also investigated. Inhibition of caspases resulted in an increase in viral glycoprotein expression, higher titers, and enhanced autophagy, whereas induction of apoptosis resulted in a decrease in viral protein expression, lower viral titer, and attenuated autophagy. On the other hand, the induction of autophagy reduced the cleavage of caspase 3. CONCLUSIONS In this study, we established how FeHV-1 induces the apoptotic process, contributing to the understanding of the relationship between FeHV-1 and this pathway.
Collapse
Affiliation(s)
- Gianmarco Ferrara
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy.
| | - Consiglia Longobardi
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Maria Francesca Sgadari
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Brunella Restucci
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Giuseppe Iovane
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Ugo Pagnini
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Serena Montagnaro
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| |
Collapse
|
5
|
Arvin AM. Creating the "Dew Drop on a Rose Petal": the Molecular Pathogenesis of Varicella-Zoster Virus Skin Lesions. Microbiol Mol Biol Rev 2023; 87:e0011622. [PMID: 37354037 PMCID: PMC10521358 DOI: 10.1128/mmbr.00116-22] [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] [Indexed: 06/26/2023] Open
Abstract
Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella (chicken pox) as the primary infection in a susceptible host. Varicella is very contagious through its transmission by direct contact with vesicular skin lesions that contain high titers of infectious virus and respiratory droplets. While the clinical manifestations of primary VZV infection are well recognized, defining the molecular mechanisms that drive VZV pathogenesis in the naive host before adaptive antiviral immunity is induced has been a challenge due to species specificity. This review focuses on advances made in identifying the differentiated human host cells targeted by VZV to cause varicella, the processes involved in viral takeover of these heterogenous cell types, and the host cell countermeasures that typically culminate in a benign illness. This work has revealed many unexpected and multifaceted mechanisms used by VZV to achieve its high prevalence and persistence in the human population.
Collapse
Affiliation(s)
- Ann M. Arvin
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Sen N, Arvin AM. Modulation of Host Cell Signaling Pathways by Varicella-Zoster Virus. Curr Top Microbiol Immunol 2023; 438:75-84. [PMID: 35624345 DOI: 10.1007/82_2021_251] [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] [Indexed: 11/28/2022]
Abstract
Host-pathogen interactions involve complex inside-out and outside-in signal transmission through critical cellular networks that dictate disease outcomes. The phosphoinositide 3-kinase (PI3K)/Akt pathway is a pivotal junction that regulates several cell functions, and phospho-Akt (pAkt) is often found to be constitutively active in cancer cells, similar to phospho-STAT3. In this chapter, we discuss the regulation of PI3K/Akt pathway in VZV infected cells and of other pathways including p53 which, unlike pAkt and pSTAT3, directs cells towards apoptosis. The fine spatio-temporal balance of activation of pro- and anti-apoptotic factors during VZV infection likely provides an optimum environment for the virus to replicate and cause disease in the human host.
Collapse
Affiliation(s)
- Nandini Sen
- Departments of Pediatrics and Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Ann M Arvin
- Departments of Pediatrics and Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
7
|
Tommasi C, Breuer J. The Biology of Varicella-Zoster Virus Replication in the Skin. Viruses 2022; 14:982. [PMID: 35632723 PMCID: PMC9147561 DOI: 10.3390/v14050982] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023] Open
Abstract
The replication of varicella-zoster virus (VZV) in skin is critical to its pathogenesis and spread. Primary infection causes chickenpox, which is characterised by centrally distributed skin blistering lesions that are rich in infectious virus. Cell-free virus in the cutaneous blistering lesions not only spreads to cause further cases, but infects sensory nerve endings, leading to the establishment of lifelong latency in sensory and autonomic ganglia. The reactivation of virus to cause herpes zoster is again characterised by localised painful skin blistering rash containing infectious virus. The development of in vitro and in vivo models of VZV skin replication has revealed aspects of VZV replication and pathogenesis in this important target organ and improved our understanding of the vaccine strain vOKa attenuation. In this review, we outline the current knowledge on VZV interaction with host signalling pathways, the viral association with proteins associated with epidermal terminal differentiation, and how these interconnect with the VZV life cycle to facilitate viral replication and shedding.
Collapse
Affiliation(s)
- Cristina Tommasi
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Judith Breuer
- Department of Infection, Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
8
|
Romero N, Favoreel HW. Pseudorabies Virus Infection Triggers NF-κB Activation via the DNA Damage Response but Actively Inhibits NF-κB-Dependent Gene Expression. J Virol 2021; 95:e0166621. [PMID: 34613805 PMCID: PMC8610585 DOI: 10.1128/jvi.01666-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
The nuclear factor kappa B (NF-κB) pathway is known to integrate signaling associated with very diverse intra- and extracellular stressors, including virus infections, and triggers a powerful (proinflammatory) response through the expression of NF-κB-regulated genes. Typically, the NF-κB pathway collects and transduces threatening signals at the cell surface or in the cytoplasm leading to nuclear import of activated NF-κB transcription factors. In the current work, we demonstrate that the swine alphaherpesvirus pseudorabies virus (PRV) induces a peculiar mode of NF-κB activation known as "inside-out" NF-κB activation. We show that PRV triggers the DNA damage response (DDR) and that this DDR response drives NF-κB activation since inhibition of the nuclear ataxia telangiectasia-mutated (ATM) kinase, a chief controller of DDR, abolished PRV-induced NF-κB activation. Initiation of the DDR-NF-κB signaling axis requires viral protein synthesis but occurs before active viral genome replication. In addition, the initiation of the DDR-NF-κB signaling axis is followed by a virus-induced complete shutoff of NF-κB-dependent gene expression that depends on viral DNA replication. In summary, the results presented in this study reveal that PRV infection triggers a noncanonical DDR-NF-κB activation signaling axis and that the virus actively inhibits the (potentially antiviral) consequences of this pathway, by inhibiting NF-κB-dependent gene expression. IMPORTANCE The NF-κB signaling pathway plays a critical role in coordination of innate immune responses that are of vital importance in the control of infections. The current report generates new insights into the interaction of the alphaherpesvirus pseudorabies virus (PRV) with the NF-κB pathway, as they reveal that (i) PRV infection leads to NF-κB activation via a peculiar "inside-out" nucleus-to-cytoplasm signal that is triggered via the DNA damage response (DDR), (ii) the DDR-NF-κB signaling axis requires expression of viral proteins but is initiated before active PRV replication, and (iii) late viral factor(s) allow PRV to actively and efficiently inhibit NF-κB-dependent (proinflammatory) gene expression. These data suggest that activation of the DDR-NF-κB during PRV infection is host driven and that its potential antiviral consequences are actively inhibited by the virus.
Collapse
Affiliation(s)
- Nicolás Romero
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Herman W. Favoreel
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
9
|
Human Herpesvirus 6A Tegument Protein U14 Induces NF-κB Signaling by Interacting with p65. J Virol 2021; 95:e0126921. [PMID: 34549982 DOI: 10.1128/jvi.01269-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viral infection induces host cells to mount a variety of immune responses, which may either limit viral propagation or create conditions conducive to virus replication in some instances. In this regard, activation of the NF-κB transcription factor is known to modulate virus replication. Human herpesvirus 6A (HHV-6A), which belongs to the Betaherpesvirinae subfamily, is frequently found in patients with neuroinflammatory diseases, although its role in disease pathogenesis has not been elucidated. In this study, we found that the HHV-6A-encoded U14 protein activates NF-κB signaling following interaction with the NF-κB complex protein, p65. Through induction of nuclear translocation of p65, U14 increases the expression of interleukin-6 (IL-6), IL-8, and monocyte chemoattractant protein 1 transcripts. We also demonstrated that activation of NF-κB signaling is important for HHV-6A replication, since inhibition of this pathway reduced virus protein accumulation and viral genome copy number. Taken together, our results suggest that HHV-6A infection activates the NF-κB pathway and promotes viral gene expression via late gene products, including U14. IMPORTANCE Human herpesvirus 6A (HHV-6A) is frequently found in patients with neuro-inflammation, although its role in the pathogenesis of this disease has not been elucidated. Most viral infections activate the NF-κB pathway, which causes the transactivation of various genes, including those encoding proinflammatory cytokines. Our results indicate that HHV-6A U14 activates the NF-κB pathway, leading to upregulation of proinflammatory cytokines. We also found that activation of the NF-κB transcription factor is important for efficient viral replication. This study provides new insight into HHV-6A U14 function in host cell signaling and identifies potential cellular targets involved in HHV-6A pathogenesis and replication.
Collapse
|
10
|
Abstract
Purpose of review Varicella zoster virus (VZV) is a highly contagious, neurotropic alpha herpes virus that causes varicella (chickenpox). VZV establishes lifelong latency in the sensory ganglia from which it can reactivate to induce herpes zoster (HZ), a painful disease that primarily affects older individuals and those who are immune-suppressed. Given that VZV infection is highly specific to humans, developing a reliable in vivo model that recapitulates the hallmarks of VZV infection has been challenging. Simian Varicella Virus (SVV) infection in nonhuman primates reproduces the cardinal features of VZV infections in humans and allows the study of varicella virus pathogenesis in the natural host. In this review, we summarize our current knowledge about genomic and virion structure of varicelloviruses as well as viral pathogenesis and antiviral immune responses during acute infection, latency and reactivation. We also examine the immune evasion mechanisms developed by varicelloviruses to escape the host immune responses and the current vaccines available for protecting individuals against chickenpox and herpes zoster. Recent findings Data from recent studies suggest that infected T cells are important for viral dissemination to the cutaneous sites of infection as well as site of latency and that a viral latency-associated transcript might play a role in the transition from lytic infection to latency and then reactivation. Summary Recent studies have provided exciting insights into mechanisms of varicelloviruses pathogenesis such as the critical role of T cells in VZV/SVV dissemination from the respiratory mucosa to the skin and the sensory ganglia; the ability of VZV/SVV to interfere with host defense; and the identification of VLT transcripts in latently infected ganglia. However, our understanding of these phenomena remains poorly understood. Therefore, it is critical that we continue to investigate host-pathogen interactions during varicelloviruses infection. These studies will lead to a deeper understanding of VZV biology as well as novel aspects of cell biology.
Collapse
|
11
|
Abstract
Prophylactic and therapeutic vaccines for the alphaherpesviruses including varicella zoster virus (VZV) and herpes simplex virus types 1 and 2 have been the focus of enormous preclinical and clinical research. A live viral vaccine for prevention of chickenpox and a subunit therapeutic vaccine to prevent zoster are highly successful. In contrast, progress towards the development of effective prophylactic or therapeutic vaccines against HSV-1 and HSV-2 has met with limited success. This review provides an overview of the successes and failures, the different types of immune responses elicited by various vaccine modalities, and the need to reconsider the preclinical models and immune correlates of protection against HSV.
Collapse
Affiliation(s)
- Clare Burn Aschner
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Betsy C. Herald
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
12
|
Gerada C, Campbell TM, Kennedy JJ, McSharry BP, Steain M, Slobedman B, Abendroth A. Manipulation of the Innate Immune Response by Varicella Zoster Virus. Front Immunol 2020; 11:1. [PMID: 32038653 PMCID: PMC6992605 DOI: 10.3389/fimmu.2020.00001] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022] Open
Abstract
Varicella zoster virus (VZV) is the causative agent of chickenpox (varicella) and shingles (herpes zoster). VZV and other members of the herpesvirus family are distinguished by their ability to establish a latent infection, with the potential to reactivate and spread virus to other susceptible individuals. This lifelong relationship continually subjects VZV to the host immune system and as such VZV has evolved a plethora of strategies to evade and manipulate the immune response. This review will focus on our current understanding of the innate anti-viral control mechanisms faced by VZV. We will also discuss the diverse array of strategies employed by VZV to regulate these innate immune responses and highlight new knowledge on the interactions between VZV and human innate immune cells.
Collapse
Affiliation(s)
- Chelsea Gerada
- Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Tessa M Campbell
- Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Jarrod J Kennedy
- Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Brian P McSharry
- Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Megan Steain
- Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Barry Slobedman
- Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Allison Abendroth
- Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Wang TY, Yang YL, Feng C, Sun MX, Peng JM, Tian ZJ, Tang YD, Cai XH. Pseudorabies Virus UL24 Abrogates Tumor Necrosis Factor Alpha-Induced NF-κB Activation by Degrading P65. Viruses 2020; 12:v12010051. [PMID: 31906441 PMCID: PMC7020041 DOI: 10.3390/v12010051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 12/17/2022] Open
Abstract
The transcription factor NF-κB plays a critical role in diverse biological processes. The NF-κB pathway can be activated by incoming pathogens and then stimulates both innate and adaptive immunity. However, many viruses have evolved corresponding strategies to balance NF-κB activation to benefit their replication. Pseudorabies virus (PRV) is an economically important pathogen that belongs to the alphaherpesvirus group. There is little information about PRV infection and NF-κB regulation. This study demonstrates for the first time that the UL24 protein could abrogate tumor necrosis factor alpha (TNF-α)-mediated NF-κB activation. An overexpression assay indicated that UL24 inhibits this pathway at or downstream of P65. Furthermore, co-immunoprecipitation analysis demonstrated that UL24 selectively interacts with P65. We demonstrated that UL24 could significantly degrade P65 by the proteasome pathway. For the first time, PRV UL24 was shown to play an important role in NF-κB evasion during PRV infection. This study expands our understanding that PRV can utilize its encoded protein UL24 to evade NF-κB signaling.
Collapse
Affiliation(s)
- Tong-Yun Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China; (T.-Y.W.); (Y.-L.Y.); (M.-X.S.); (J.-M.P.)
| | - Yue-Lin Yang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China; (T.-Y.W.); (Y.-L.Y.); (M.-X.S.); (J.-M.P.)
| | - Cong Feng
- Guangdong Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou 510000, China;
| | - Ming-Xia Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China; (T.-Y.W.); (Y.-L.Y.); (M.-X.S.); (J.-M.P.)
| | - Jin-Mei Peng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China; (T.-Y.W.); (Y.-L.Y.); (M.-X.S.); (J.-M.P.)
| | - Zhi-Jun Tian
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China; (T.-Y.W.); (Y.-L.Y.); (M.-X.S.); (J.-M.P.)
- Correspondence: (Z.-J.T.); (Y.-D.T.); (X.-H.C.); Tel.: +86-18249466512 (Y.-D.T.); +86-135-0451-2466 (X.-H.C.)
| | - Yan-Dong Tang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China; (T.-Y.W.); (Y.-L.Y.); (M.-X.S.); (J.-M.P.)
- Correspondence: (Z.-J.T.); (Y.-D.T.); (X.-H.C.); Tel.: +86-18249466512 (Y.-D.T.); +86-135-0451-2466 (X.-H.C.)
| | - Xue-Hui Cai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China; (T.-Y.W.); (Y.-L.Y.); (M.-X.S.); (J.-M.P.)
- Correspondence: (Z.-J.T.); (Y.-D.T.); (X.-H.C.); Tel.: +86-18249466512 (Y.-D.T.); +86-135-0451-2466 (X.-H.C.)
| |
Collapse
|
14
|
Interferon Gamma Inhibits Varicella-Zoster Virus Replication in a Cell Line-Dependent Manner. J Virol 2019; 93:JVI.00257-19. [PMID: 30918075 DOI: 10.1128/jvi.00257-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023] Open
Abstract
The major immediate early 62 (IE62) protein of varicella-zoster virus (VZV) is delivered to newly infected cell nuclei, where it initiates VZV replication by transactivating viral immediate early (IE), early (E), and late (L) genes. Interferon gamma (IFN-γ) is a potent cytokine produced following primary VZV infection. Furthermore, VZV reactivation correlates with a decline in IFN-γ-producing immune cells. Our results showed that treatment with 20 ng/ml of IFN-γ completely reduced intracellular VZV yield in A549 lung epithelial cells, MRC-5 lung fibroblasts, and ARPE-19 retinal epithelial cells at 4 days post-VZV infection. However, IFN-γ reduced virus yield only 2-fold in MeWo melanoma cells compared to that of untreated cells. IFN-β significantly inhibited VZV replication in both ARPE-19 and MeWo cells. In luciferase assays with VZV open reading frame 61 (ORF61) promoter reporter plasmid, IFN-γ abrogated the transactivation activity of IE62 by 95%, 97%, and 89% in A549, ARPE-19, and MRC-5 cells, respectively. However, IFN-γ abrogated IE62's transactivation activity by 16% in MeWo cells, indicating that IFN-γ inhibits VZV replication as well as IE62-mediated transactivation in a cell line-dependent manner. The expression of VZV IE62 and ORF63 suppressed by IFN-γ was restored by JAK1 inhibitor treatment, indicating that the inhibition of VZV replication is mediated by JAK/STAT1 signaling. In the presence of IFN-γ, knockdown of interferon response factor 1 (IRF1) increased VZV replication. Ectopic expression of IRF1 reduced VZV yields 4,000-fold in MRC-5 and ARPE-19 cells but 3-fold in MeWo cells. These results suggest that IFN-γ blocks VZV replication by inhibiting IE62 function in a cell line-dependent manner.IMPORTANCE Our results showed that IFN-γ significantly inhibited VZV replication in a cell line-dependent manner. IFN-γ inhibited VZV gene expression after the immediate early stage of infection and abrogated IE62-mediated transactivation. These results suggest that IFN-γ blocks VZV replication by inhibiting IE62 function in a cell line-dependent manner. Understanding the mechanisms by which IFN-γ plays a role in VZV gene programming may be important in determining the tissue restriction of VZV.
Collapse
|
15
|
Sharma R, Chakraborty T, Buadi FK, Beam E, Pureza VS, Pagani-Estevez GL, Flanagan EP. Clinical Reasoning: A 56-year-old woman with acute vertigo and diplopia. Neurology 2018; 90:748-752. [PMID: 29661894 DOI: 10.1212/wnl.0000000000005337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Rishi Sharma
- From the Department of Neurology (R.S., T.C., G.L.P.-E., E.P.F.) and Divisions of Hematology (F.K.B.), Infectious Disease (E.B.), and Hospital Internal Medicine, Department of Medicine (V.S.P.), Mayo Clinic, Rochester, MN; and College of Biological Sciences (R.S.), University of Minnesota, Minneapolis
| | - Tia Chakraborty
- From the Department of Neurology (R.S., T.C., G.L.P.-E., E.P.F.) and Divisions of Hematology (F.K.B.), Infectious Disease (E.B.), and Hospital Internal Medicine, Department of Medicine (V.S.P.), Mayo Clinic, Rochester, MN; and College of Biological Sciences (R.S.), University of Minnesota, Minneapolis
| | - Francis K Buadi
- From the Department of Neurology (R.S., T.C., G.L.P.-E., E.P.F.) and Divisions of Hematology (F.K.B.), Infectious Disease (E.B.), and Hospital Internal Medicine, Department of Medicine (V.S.P.), Mayo Clinic, Rochester, MN; and College of Biological Sciences (R.S.), University of Minnesota, Minneapolis
| | - Elena Beam
- From the Department of Neurology (R.S., T.C., G.L.P.-E., E.P.F.) and Divisions of Hematology (F.K.B.), Infectious Disease (E.B.), and Hospital Internal Medicine, Department of Medicine (V.S.P.), Mayo Clinic, Rochester, MN; and College of Biological Sciences (R.S.), University of Minnesota, Minneapolis
| | - Vincent S Pureza
- From the Department of Neurology (R.S., T.C., G.L.P.-E., E.P.F.) and Divisions of Hematology (F.K.B.), Infectious Disease (E.B.), and Hospital Internal Medicine, Department of Medicine (V.S.P.), Mayo Clinic, Rochester, MN; and College of Biological Sciences (R.S.), University of Minnesota, Minneapolis
| | - Gabriel L Pagani-Estevez
- From the Department of Neurology (R.S., T.C., G.L.P.-E., E.P.F.) and Divisions of Hematology (F.K.B.), Infectious Disease (E.B.), and Hospital Internal Medicine, Department of Medicine (V.S.P.), Mayo Clinic, Rochester, MN; and College of Biological Sciences (R.S.), University of Minnesota, Minneapolis
| | - Eoin P Flanagan
- From the Department of Neurology (R.S., T.C., G.L.P.-E., E.P.F.) and Divisions of Hematology (F.K.B.), Infectious Disease (E.B.), and Hospital Internal Medicine, Department of Medicine (V.S.P.), Mayo Clinic, Rochester, MN; and College of Biological Sciences (R.S.), University of Minnesota, Minneapolis.
| |
Collapse
|
16
|
Sarkar S, Balasuriya UBR, Horohov DW, Chambers TM. Equine herpesvirus-1 infection disrupts interferon regulatory factor-3 (IRF-3) signaling pathways in equine endothelial cells. Vet Immunol Immunopathol 2016; 173:1-9. [PMID: 27090619 DOI: 10.1016/j.vetimm.2016.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 03/08/2016] [Accepted: 03/10/2016] [Indexed: 12/25/2022]
Abstract
Equine herpesvirus-1 (EHV-1) is a major respiratory viral pathogen of horses, causing upper respiratory tract disease, abortion, neonatal death, and neurological disease that may lead to paralysis and death. EHV-1 replicates initially in the respiratory epithelium and then spreads systemically to endothelial cells lining the small blood vessels in the uterus and spinal cord leading to abortion and EHM in horses. Like other herpesviruses, EHV-1 employs a variety of mechanisms for immune evasion including suppression of type-I interferon (IFN) production in equine endothelial cells (EECs). Previously we have shown that the neuropathogenic T953 strain of EHV-1 inhibits type-I IFN production in EECs and this is mediated by a viral late gene product. But the mechanism of inhibition was not known. Here we show that T953 strain infection of EECs induced degradation of endogenous IRF-3 protein. This in turn interfered with the activation of IRF-3 signaling pathways. EHV-1 infection caused the activation of the NF-κB signaling pathways, suggesting that inhibition of type-I IFN production is probably due to interference in IRF-3 and not NF-κB signal transduction.
Collapse
Affiliation(s)
- Sanjay Sarkar
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA.
| | - Udeni B R Balasuriya
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA
| | - David W Horohov
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA
| | - Thomas M Chambers
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA
| |
Collapse
|
17
|
Modulation of host CD59 expression by varicella-zoster virus in human xenografts in vivo. Virology 2016; 491:96-105. [PMID: 26891237 DOI: 10.1016/j.virol.2016.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 01/13/2016] [Accepted: 01/26/2016] [Indexed: 01/06/2023]
Abstract
Varicella-zoster virus (VZV) is the causative agent of both chickenpox (varicella) and shingles (zoster). VZV survives host defenses, even with an intact immune system, and disseminates in the host before causing disease. To date, several diverse immunomodulatory strategies used by VZV to undermine host immunity have been identified; however, few studies have addressed the complement evasion strategies used by this virus. Here, we show that expression of CD59, which is a key member of host regulators of complement activation (RCA), is significantly upregulated in response to VZV infection in human T cells and dorsal root ganglia (DRG) but not in human skin xenografts in SCID-hu mice in vivo. This is the first report demonstrating that VZV infection upregulates host CD59 expression in a tissue-specific manner in vivo, which may aid VZV in complement evasion and pathogenesis.
Collapse
|
18
|
Lee JY, Lim SH, Lee MY, Kim H, Sinn DH, Gwak GY, Choi MS, Lee JH, Jung CW, Jang JH, Kim WS, Kim SJ, Kim K. Hepatitis B reactivation in multiple myeloma patients with resolved hepatitis B undergoing chemotherapy. Liver Int 2015; 35:2363-9. [PMID: 25832927 DOI: 10.1111/liv.12838] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/23/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Despite increasing reports of hepatitis B virus (HBV) reactivation in multiple myeloma (MM), HBV reactivation in patients with resolved hepatitis B [hepatitis B surface antigen (HBsAg)-negative/anti-hepatitis B core antigen antibody (anti-HBc)-positive] is still poorly characterized. The aim of this study was to clarify its frequency and risk factors. METHODS A total of 230 MM patients with resolved hepatitis B were retrospectively reviewed for HBV reactivation and biochemical flare. RESULTS During a median 2.4 years of follow-up, HBV reactivation was diagnosed in 12 patients (5.2%). The cumulative rates of HBV reactivation at 2 years and 5 years were 5% and 8% respectively. A baseline anti-HBs-negative status (P = 0.033) and high-dose therapy/autologous stem-cell transplantation [HDT/ASCT (P = 0.025)] were significant risk factors that were positively associated with HBV reactivation. In subgroup analysis of patients treated with HDT/ASCT (n = 127), a baseline anti-HBs-negative status was the only significant risk factor for HBV reactivation (hazard ratio, 4.64; 95% CI, 1.47-14.7; P = 0.009). DISCUSSION These data show that evaluation of anti-HBc is needed for MM patients, and suggest that monitoring of HBV DNA should be considered for patients with resolved hepatitis B undergoing HDT/ASCT, especially those who are anti-HBs-negative.
Collapse
Affiliation(s)
- Ji Yun Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Hee Lim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min-Young Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Haesu Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong Hyun Sinn
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Geum-Youn Gwak
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Moon Seok Choi
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Hyeok Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chul Won Jung
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jun Ho Jang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Seog Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kihyun Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Sarkar S, Balasuriya UBR, Horohov DW, Chambers TM. Equine herpesvirus-1 suppresses type-I interferon induction in equine endothelial cells. Vet Immunol Immunopathol 2015; 167:122-9. [PMID: 26275803 DOI: 10.1016/j.vetimm.2015.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/23/2015] [Accepted: 07/30/2015] [Indexed: 12/31/2022]
Abstract
Equine herpesvirus-1 (EHV-1) is one of the most common and important respiratory viral pathogens of horses. EHV-1 in horses replicates initially in the respiratory epithelium and then spreads systematically to endothelial cells lining the small blood vessels in the uterus and spinal cord, and highly pathogenic virus strains can produce aborted fetuses or myeloencephalopathy. Like other herpes viruses, EHV-1 employs a variety of mechanisms for immune evasion. Some herpes viruses down-regulate the type-I interferon (IFN) response to infection, but such activity has not been described for EHV-1. Here, in an in vitro system utilizing an established equine endothelial cell line, we studied the temporal effect on IFN-β responses following infection with the neuropathogenic T953 strain of EHV-1. Results show that after an early induction of IFN-β, the virus actively shut down further production of IFN-β and this was correlated with expression of the viral late genes. Expression of the IFN response factor viperin, a marker of host cell type-I IFN responses, was also suppressed by T953 virus infection. EHV-1-mediated suppression of host type-I IFN responses may play an important role in EHV-1 pathogenesis and the mechanism of this, presumably involving a viral late gene product, warrants investigation.
Collapse
Affiliation(s)
- Sanjay Sarkar
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA.
| | - Udeni B R Balasuriya
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA
| | - David W Horohov
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA
| | - Thomas M Chambers
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546-0099, USA
| |
Collapse
|
20
|
The ORF61 Protein Encoded by Simian Varicella Virus and Varicella-Zoster Virus Inhibits NF-κB Signaling by Interfering with IκBα Degradation. J Virol 2015; 89:8687-700. [PMID: 26085158 DOI: 10.1128/jvi.01149-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/12/2015] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Varicella-zoster virus (VZV) causes chickenpox upon primary infection and establishes latency in ganglia. Reactivation from latency causes herpes zoster, which may be complicated by postherpetic neuralgia. Innate immunity mediated by interferon and proinflammatory cytokines represents the first line of immune defense upon infection and reactivation. VZV is known to interfere with multiple innate immune signaling pathways, including the central transcription factor NF-κB. However, the role of these inhibitory mechanisms in vivo is unknown. Simian varicella virus (SVV) infection of rhesus macaques recapitulates key aspects of VZV pathogenesis, and this model thus permits examination of the role of immune evasion mechanisms in vivo. Here, we compare SVV and VZV with respect to interference with NF-κB activation. We demonstrate that both viruses prevent ubiquitination of the NF-κB inhibitor IκBα, whereas SVV additionally prevents IκBα phosphorylation. We show that the ORF61 proteins of VZV and SVV are sufficient to prevent IκBα ubiquitination upon ectopic expression. We further demonstrate that SVV ORF61 interacts with β-TrCP, a subunit of the SCF ubiquitin ligase complex that mediates the degradation of IκBα. This interaction seems to inactivate SCF-mediated protein degradation in general, since the unrelated β-TrCP target Snail is also stabilized by ORF61. In addition to ORF61, SVV seems to encode additional inhibitors of the NF-κB pathway, since SVV with ORF61 deleted still prevented IκBα phosphorylation and degradation. Taken together, our data demonstrate that SVV interferes with tumor necrosis factor alpha (TNF-α)-induced NF-κB activation at multiple levels, which is consistent with the importance of these countermechanisms for varicella virus infection. IMPORTANCE The role of innate immunity during the establishment of primary infection, latency, and reactivation by varicella-zoster virus (VZV) is incompletely understood. Since infection of rhesus macaques by simian varicella virus (SVV) is used as an animal model of VZV infection, we characterized the molecular mechanism by which SVV interferes with innate immune activation. Specifically, we studied how SVV prevents activation of the transcription factor NF-κB, a central factor in eliciting proinflammatory responses. The identification of molecular mechanisms that counteract innate immunity might ultimately lead to better vaccines and treatments for VZV, since overcoming these mechanisms, either by small-molecule inhibition or by genetic modification of vaccine strains, is expected to reduce the pathogenic potential of VZV. Moreover, using SVV infection of rhesus macaques, it will be possible to study how increasing the vulnerability of varicella viruses to innate immunity will impact viral pathogenesis.
Collapse
|
21
|
Duncan CJ, Hambleton S. Varicella zoster virus immunity: A primer. J Infect 2015; 71 Suppl 1:S47-53. [DOI: 10.1016/j.jinf.2015.04.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2015] [Indexed: 01/22/2023]
|
22
|
Herpes simplex virus 1 protein kinase US3 hyperphosphorylates p65/RelA and dampens NF-κB activation. J Virol 2014; 88:7941-51. [PMID: 24807716 DOI: 10.1128/jvi.03394-13] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nuclear factor κB (NF-κB) plays important roles in innate immune responses by regulating the expression of a large number of target genes involved in the immune and inflammatory response, apoptosis, cell proliferation, differentiation, and survival. To survive in the host cells, viruses have evolved multiple strategies to evade and subvert the host immune response. Herpes simplex virus 1 (HSV-1) bears a large DNA genome, with the capacity to encode many different viral proteins to counteract the host immune responses. In the present study, we demonstrated that HSV-1 protein kinase US3 significantly inhibited NF-κB activation and decreased the expression of inflammatory chemokine interleukin-8 (IL-8). US3 was also shown to hyperphosphorylate p65 at serine 75 and block its nuclear translocation. Two US3 mutants, K220M and D305A, still interacted with p65; however, they could not hyperphosphorylate p65, indicating that the kinase activity of US3 was indispensable for the function. The attenuation of NF-κB activation by HSV-1 US3 protein kinase may represent a critical adaptation to enable virus persistence within the host. Importance: This study demonstrated that HSV-1 protein kinase US3 significantly inhibited NF-κB activation and decreased the expression of inflammatory chemokine interleukin-8 (IL-8). US3 hyperphosphorylated p65 at serine 75 to inhibit NF-κB activation. The kinase activity of US3 was indispensable for its hyperphosphorylation of p65 and abrogation of the nuclear translocation of p65. The present study elaborated a novel mechanism of HSV-1 US3 to evade the host innate immunity.
Collapse
|
23
|
Cellular transcriptome analysis reveals differential expression of pro- and antiapoptosis genes by varicella-zoster virus-infected neurons and fibroblasts. J Virol 2014; 88:7674-7. [PMID: 24741086 DOI: 10.1128/jvi.00500-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transcriptional changes following varicella-zoster virus (VZV) infection of cultured human neurons derived from embryonic stem cells were compared to those in VZV-infected human foreskin fibroblasts. Transcription of 340 neuronal genes significantly altered by VZV infection included 223 transcript changes unique to neurons. Strikingly, genes inhibiting apoptosis were upregulated in neurons, while proapoptotic gene transcription was increased in fibroblasts. These data are a basis for discovery of differences in virus-host interactions between these VZV targets.
Collapse
|
24
|
Zerboni L, Sen N, Oliver SL, Arvin AM. Molecular mechanisms of varicella zoster virus pathogenesis. Nat Rev Microbiol 2014; 12:197-210. [PMID: 24509782 PMCID: PMC4066823 DOI: 10.1038/nrmicro3215] [Citation(s) in RCA: 298] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Varicella zoster virus (VZV) is the causative agent of varicella (chickenpox) and zoster (shingles). Investigating VZV pathogenesis is challenging as VZV is a human-specific virus and infection does not occur, or is highly restricted, in other species. However, the use of human tissue xenografts in mice with severe combined immunodeficiency (SCID) enables the analysis of VZV infection in differentiated human cells in their typical tissue microenvironment. Xenografts of human skin, dorsal root ganglia or foetal thymus that contains T cells can be infected with mutant viruses or in the presence of inhibitors of viral or cellular functions to assess the molecular mechanisms of VZV-host interactions. In this Review, we discuss how these models have improved our understanding of VZV pathogenesis.
Collapse
Affiliation(s)
- Leigh Zerboni
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Nandini Sen
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Stefan L Oliver
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Ann M Arvin
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
25
|
Jones M, Dry IR, Frampton D, Singh M, Kanda RK, Yee MB, Kellam P, Hollinshead M, Kinchington PR, O'Toole EA, Breuer J. RNA-seq analysis of host and viral gene expression highlights interaction between varicella zoster virus and keratinocyte differentiation. PLoS Pathog 2014; 10:e1003896. [PMID: 24497829 PMCID: PMC3907375 DOI: 10.1371/journal.ppat.1003896] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 12/09/2013] [Indexed: 12/27/2022] Open
Abstract
Varicella zoster virus (VZV) is the etiological agent of chickenpox and shingles, diseases characterized by epidermal skin blistering. Using a calcium-induced keratinocyte differentiation model we investigated the interaction between epidermal differentiation and VZV infection. RNA-seq analysis showed that VZV infection has a profound effect on differentiating keratinocytes, altering the normal process of epidermal gene expression to generate a signature that resembles patterns of gene expression seen in both heritable and acquired skin-blistering disorders. Further investigation by real-time PCR, protein analysis and electron microscopy revealed that VZV specifically reduced expression of specific suprabasal cytokeratins and desmosomal proteins, leading to disruption of epidermal structure and function. These changes were accompanied by an upregulation of kallikreins and serine proteases. Taken together VZV infection promotes blistering and desquamation of the epidermis, both of which are necessary to the viral spread and pathogenesis. At the same time, analysis of the viral transcriptome provided evidence that VZV gene expression was significantly increased following calcium treatment of keratinocytes. Using reporter viruses and immunohistochemistry we confirmed that VZV gene and protein expression in skin is linked with cellular differentiation. These studies highlight the intimate host-pathogen interaction following VZV infection of skin and provide insight into the mechanisms by which VZV remodels the epidermal environment to promote its own replication and spread. Varicella zoster virus (VZV) causes chickenpox and shingles, which are characterised by the formation of fluid-filled skin lesions. Infectious viral particles present in these lesions are critical for airborne spread to cause chickenpox in non-immune contacts and for infection of nerve ganglia via nerve endings in the skin, a pre-requisite for shingles. Several VZV proteins, although dispensable in laboratory cell-culture, are essential for VZV infection of skin, a finding thought to relate to VZV interaction with a process known as epidermal differentiation. In this, the specialised keratinocyte cells of the outer layer of skin, the epidermis, are continually shed to be replaced by differentiating keratinocytes, which migrate up from lower layers. How VZV interaction with epidermal differentiation leads to the formation of fluid-filled lesions remains unclear. We show using a keratinocyte model of epidermal differentiation that VZV infection alters epidermal differentiation, generating a specific pattern of changes in that is characteristic of blistering and skin shedding diseases. We also identified that the differentiation status of the keratinocytes influences the replication pattern of the viral gene and protein expression, with both increasing as the VZV particles traverses to the uppermost layers of the skin. The findings provide new insights into VZV-host cell interactions.
Collapse
Affiliation(s)
- Meleri Jones
- Division of Infection and Immunity, University College London, London, United Kingdom
- * E-mail:
| | - Inga R. Dry
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dan Frampton
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Manuraj Singh
- Centre for Cutaneous Research, QMUL, London, United Kingdom
| | - Ravinder K. Kanda
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Michael B. Yee
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Paul Kellam
- Division of Infection and Immunity, University College London, London, United Kingdom
- Virus Genomics Team, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Michael Hollinshead
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Paul R. Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | | - Judith Breuer
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
26
|
Yamakura M, Tsuda K, Ugai T, Sugihara H, Nisihida Y, Takeuchi M, Matsue K. High frequency of varicella zoster virus reactivation associated with the use of arsenic trioxide in patients with acute promyelocytic leukemia. Acta Haematol 2013; 131:76-7. [PMID: 24081111 DOI: 10.1159/000353126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/29/2013] [Indexed: 11/19/2022]
Affiliation(s)
- Masayuki Yamakura
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Varicella zoster virus (VZV) is a highly successful human pathogen, which is never completely eliminated from the host. VZV causes two clinically distinct diseases, varicella (chickenpox) during primary infection and herpes zoster (shingles) following virus reactivation from latency. Throughout its lifecycle the virus encounters the innate and adaptive immune response, and in order to prevent eradication it has developed many mechanisms to evade and overcome these responses. This review will provide a comprehensive overview of the host immune response to VZV infection, during the multiple stages of the virus lifecycle and at key sites of VZV infection. We will also briefly describe some of the strategies employed by the virus to overcome the host immune response and the ongoing challenges in further elucidating the interplay between VZV and the host immune response in an attempt to lead to better therapies and a ‘second generation’ vaccine for VZV disease.
Collapse
Affiliation(s)
- Megan Steain
- Discipline of Infectious Diseases & Immunology, The University of Sydney, NSW, Australia
- Centre for Virus Research, Westmead Millennium Institute, NSW, Australia
| | - Barry Slobedman
- Discipline of Infectious Diseases & Immunology, The University of Sydney, NSW, Australia
- Centre for Virus Research, Westmead Millennium Institute, NSW, Australia
| | - Allison Abendroth
- Discipline of Infectious Diseases & Immunology, The University of Sydney, NSW, Australia
| |
Collapse
|
28
|
Chen LJ, Dong XY, Zhao MQ, Shen HY, Wang JY, Pei JJ, Liu WJ, Luo YW, Ju CM, Chen JD. Classical swine fever virus failed to activate nuclear factor-kappa b signaling pathway both in vitro and in vivo. Virol J 2012. [PMID: 23186553 PMCID: PMC3565942 DOI: 10.1186/1743-422x-9-293] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Classical swine fever virus (CSFV) is the cause of CSF which is a severe disease of pigs, leading to heavy economic losses in many regions of the world. Nuclear factor-kappa B (NF-κB) is a critical regulator of innate and adaptive immunity, and commonly activated upon viral infection. In our previous study, we found that CSFV could suppress the maturation and modulate the functions of monocyte-derived dendritic cells (Mo-DCs) without activating NF-κB pathway. To further prove the effects of CSFV on the NF-κB signaling pathway, we investigated the activity of NF-κB after CSFV infection in vivo and in vitro. METHODS Attenuated Thiverval strain and virulent wild-type GXW-07 strain were used as challenge viruses in this study. Porcine kidney 15 (PK-15) cells were cultured in vitro and peripheral blood mononuclear cells (PBMCs) were isolated from the blood of CSFV-infected pigs. DNA binding of NF-κB was measured by electrophoretic mobility shift assays (EMSA), NF-κB p65 translocation was detected using immunofluorescent staining, and p65/RelA and IκBα expression was measured by Western Blotting. RESULTS Infection of cells with CSFV in vitro and in vivo showed that compared with tumor necrosis factor alpha (TNF-α) stimulated cells, there was no distinct DNA binding band of NF-κB, and no significant translocation of p65/RelA from the cytoplasm to the nucleus was observed, which might have been due to the apparent lack of IkBa degradation. CONCLUSIONS CSFV infection had no effect on the NF-κB signaling pathway, indicating that CSFV could evade host activation of NF-κB during infection.
Collapse
Affiliation(s)
- Li-Jun Chen
- College of Veterinary Medicine, South China Agricultural University, 483 Wu Shan Road, Guangzhou, Tian He District 510642, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Signal transducer and activator of transcription 3 (STAT3) and survivin induction by varicella-zoster virus promote replication and skin pathogenesis. Proc Natl Acad Sci U S A 2011; 109:600-5. [PMID: 22190485 DOI: 10.1073/pnas.1114232109] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Varicella-zoster virus (VZV) is a human α-herpesvirus that causes varicella (chickenpox) during primary infection and zoster (shingles) upon reactivation. Like other viruses, VZV must subvert the intrinsic antiviral defenses of differentiated human cells to produce progeny virions. Accordingly, VZV inhibits the activation of the cellular transcription factors IFN regulatory factor 3 (IRF3) and signal transducers and activators of transcription 1 (STAT1), thereby downregulating antiviral factors, including IFNs. Conversely, in this study, we found that VZV triggers STAT3 phosphorylation in cells infected in vitro and in human skin xenografts in SCID mice in vivo and that STAT3 activation induces the anti-apoptotic protein survivin. Small-molecule inhibitors of STAT3 phosphorylation and survivin restrict VZV replication in vitro, and VZV infection of skin xenografts in vivo is markedly impaired by the administration of the phospho-STAT3 inhibitor S3I-201. STAT3 and survivin are required for malignant transformation caused by γ-herpesviruses, such as Kaposi's sarcoma virus. We show that STAT3 activation is also critical for VZV, a nononcogenic herpesvirus, via a survivin-dependent mechanism. Furthermore, STAT3 activation is critical for the life cycle of the virus because VZV skin infection is necessary for viral transmission and persistence in the human population. Therefore, we conclude that takeover of this major cell-signaling pathway is necessary, independent of cell transformation, for herpesvirus pathogenesis and that STAT3 activation and up-regulation of survivin is a common mechanism important for the pathogenesis of lytic as well as tumorigenic herpesviruses.
Collapse
|
30
|
Varicella-zoster virus inhibition of the NF-κB pathway during infection of human dendritic cells: role for open reading frame 61 as a modulator of NF-κB activity. J Virol 2011; 86:1193-202. [PMID: 22090112 DOI: 10.1128/jvi.06400-11] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Dendritic cells (DC) are antigen-presenting cells essential for initiating primary immune responses and therefore an ideal target for viral immune evasion. Varicella-zoster virus (VZV) can productively infect immature human DCs and impair their function as immune effectors by inhibiting their maturation, as evidenced by the expression modulation of functionally important cell surface immune molecules CD80, CD86, CD83, and major histocompatibility complex I. The NF-κB pathway largely regulates the expression of these immune molecules, and therefore we sought to determine whether VZV infection of DCs modulates the NF-κB pathway. Nuclear localization of NF-κB p50 and p65 indicates pathway activation; however, immunofluorescence studies revealed cytoplasmic retention of these NF-κB subunits in VZV-infected DCs. Western blotting revealed phosphorylation of the inhibitor of κBα (IκBα) in VZV-infected DCs, indicating that the pathway is active at this point. We conclude that VZV infection of DC inhibits the NF-κB pathway following protein phosphorylation but before the translocation of NF-κB subunits into the nucleus. An NF-κB reporter assay identified VZV open reading frame 61 (ORF61) as an inhibitor of tumor necrosis factor alpha-induced NF-κB reporter activity. Mutational analysis of ORF61 identified the E3 ubiquitin ligase domain as a region required for NF-κB pathway inhibition. In summary, we provide evidence that VZV inhibits the NF-κB signaling pathway in human DCs and that the E3 ubiquitin ligase domain of ORF61 is required to modulate this pathway. Thus, this work identifies a mechanism by which VZV modulates host immune function.
Collapse
|
31
|
Wang L, Oliver SL, Sommer M, Rajamani J, Reichelt M, Arvin AM. Disruption of PML nuclear bodies is mediated by ORF61 SUMO-interacting motifs and required for varicella-zoster virus pathogenesis in skin. PLoS Pathog 2011; 7:e1002157. [PMID: 21901090 PMCID: PMC3161977 DOI: 10.1371/journal.ppat.1002157] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 05/23/2011] [Indexed: 12/15/2022] Open
Abstract
Promyelocytic leukemia protein (PML) has antiviral functions and many viruses encode gene products that disrupt PML nuclear bodies (PML NBs). However, evidence of the relevance of PML NB modification for viral pathogenesis is limited and little is known about viral gene functions required for PML NB disruption in infected cells in vivo. Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes cutaneous lesions during primary and recurrent infection. Here we show that VZV disrupts PML NBs in infected cells in human skin xenografts in SCID mice and that the disruption is achieved by open reading frame 61 (ORF61) protein via its SUMO-interacting motifs (SIMs). Three conserved SIMs mediated ORF61 binding to SUMO1 and were required for ORF61 association with and disruption of PML NBs. Mutation of the ORF61 SIMs in the VZV genome showed that these motifs were necessary for PML NB dispersal in VZV-infected cells in vitro. In vivo, PML NBs were highly abundant, especially in basal layer cells of uninfected skin, whereas their frequency was significantly decreased in VZV-infected cells. In contrast, mutation of the ORF61 SIMs reduced ORF61 association with PML NBs, most PML NBs remained intact and importantly, viral replication in skin was severely impaired. The ORF61 SIM mutant virus failed to cause the typical VZV lesions that penetrate across the basement membrane into the dermis and viral spread in the epidermis was limited. These experiments indicate that VZV pathogenesis in skin depends upon the ORF61-mediated disruption of PML NBs and that the ORF61 SUMO-binding function is necessary for this effect. More broadly, our study elucidates the importance of PML NBs for the innate control of a viral pathogen during infection of differentiated cells within their tissue microenvironment in vivo and the requirement for a viral protein with SUMO-binding capacity to counteract this intrinsic barrier. PML nuclear bodies (PML NBs) are spherical nuclear structures that are present in most human and animal cells. These bodies contribute to anti-viral defense and therefore many viruses have developed strategies to disrupt them. This interaction has been demonstrated for a number of viruses in cultured cells but little is known about these processes in differentiated cells within human tissues. Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes chicken pox and shingle lesions in skin. Here we show that VZV disrupts PML NBs in epidermal and dermal cells in skin tissues implanted subcutaneously in immunodeficient mice. We found that PML NB dispersal is mediated by VZV ORF61 protein and is required for VZV cell to cell spread and lesion formation in skin. The ability of ORF61 to disrupt PML NBs depends on its capacity to bind to SUMO1 protein, which is conjugated to PML and other proteins within PML NBs. To our knowledge, our study provides the first evidence of PML NB modification through the SUMO-binding function of a viral protein, VZV ORF61, and the importance of this molecular mechanism for virus-induced PML NB disruption in differentiated cells infected within their tissue microenvironment in vivo.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Line, Tumor
- Cell Proliferation
- Cloning, Molecular
- Genes, Viral
- Herpesvirus 3, Human/genetics
- Herpesvirus 3, Human/pathogenicity
- Herpesvirus 3, Human/physiology
- Humans
- Intranuclear Inclusion Bodies/metabolism
- Intranuclear Inclusion Bodies/virology
- Leukemia, Promyelocytic, Acute
- Mice
- Mice, SCID
- Models, Animal
- Mutagenesis
- Plasmids/genetics
- Protein Interaction Domains and Motifs/genetics
- SUMO-1 Protein/genetics
- SUMO-1 Protein/metabolism
- Skin/virology
- Up-Regulation
- Viral Proteins/genetics
- Viral Proteins/metabolism
- Virus Replication
Collapse
Affiliation(s)
- Li Wang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, United States of America.
| | | | | | | | | | | |
Collapse
|
32
|
Chen LJ, Dong XY, Shen HY, Zhao MQ, Ju CM, Yi L, Zhang XT, Kang YM, Chen JD. Classical swine fever virus suppresses maturation and modulates functions of monocyte-derived dendritic cells without activating nuclear factor kappa B. Res Vet Sci 2011; 93:529-37. [PMID: 21764089 DOI: 10.1016/j.rvsc.2011.06.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/21/2011] [Accepted: 06/24/2011] [Indexed: 11/25/2022]
Abstract
Classical swine fever virus (CSFV) compromises the host immune system, causing the severe disease of pigs. Dendritic cells (DCs) are the most potent inducers of immune responses. In the present study, we investigated the functional properties of porcine monocyte-derived DCs (Mo-DCs) affected by CSFV. Results showed that the expression of surface markers of DCs such as major histocompatibility complex class II (MHC-II), CD80, CD83 and CD86 were unimpaired, but an obviously increased expression of CD172a in DCs was noticed 48 h after CSFV infection. The expression profiles of cytokines were detected in cultured Mo-DCs after various treatments for 48 h by Q-RT-PCR. The findings suggested that CSFV infection significantly increased the mRNA expression of IL-10 and TNF-α, and inhibited IL-12 expression, with little effect on IFN-α and IFN-γ expression. We further demonstrated that CSFV was incapable of activating the nuclear factor kappa B (NF-κB) in infected DCs, which was characterized by an unvaried DNA binding activity of NF-κB, the lack of translocation of p65/RelA from the cytoplasm to the nucleus and the stabilization of p65/RelA expression. Furthermore, Western blot analysis indicated that the inactivation of NF-κB was due to the failure of IκBα degradation. The data demonstrated that CSFV could be replicated in DCs and CSFV infection could modulate the secretion of crucial co-stimulatory molecules and cytokines which down-regulated maturation of DCs, without activating NF-κB in DCs. Thus, the results suggested a possible mechanism for CSFV evasion of innate host defenses, providing the basis for understanding molecular pathways in CSFV pathogenesis.
Collapse
Affiliation(s)
- Li-Jun Chen
- College of Veterinary Medicine, South China Agricultural University, 483 Wu Shan Road, Tian He District, Guangzhou 510642, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors plays a central part in the host response to infection by microbial pathogens, by orchestrating the innate and acquired host immune responses. The NF-κB proteins are activated by diverse signalling pathways that originate from many different cellular receptors and sensors. Many successful pathogens have acquired sophisticated mechanisms to regulate the NF-κB signalling pathways by deploying subversive proteins or hijacking the host signalling molecules. Here, we describe the mechanisms by which viruses and bacteria micromanage the host NF-κB signalling circuitry to favour the continued survival of the pathogen.
Collapse
Affiliation(s)
- Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100266, Gainesville, Florida, USA
| | | |
Collapse
|
34
|
Vandevenne P, Lebrun M, El Mjiyad N, Ote I, Di Valentin E, Habraken Y, Dortu E, Piette J, Sadzot-Delvaux C. The varicella-zoster virus ORF47 kinase interferes with host innate immune response by inhibiting the activation of IRF3. PLoS One 2011; 6:e16870. [PMID: 21347389 PMCID: PMC3036730 DOI: 10.1371/journal.pone.0016870] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 01/17/2011] [Indexed: 12/24/2022] Open
Abstract
The innate immune response constitutes the first line of host defence that limits viral spread and plays an important role in the activation of adaptive immune response. Viral components are recognized by specific host pathogen recognition receptors triggering the activation of IRF3. IRF3, along with NF-κB, is a key regulator of IFN-β expression. Until now, the role of IRF3 in the activation of the innate immune response during Varicella-Zoster Virus (VZV) infection has been poorly studied. In this work, we demonstrated for the first time that VZV rapidly induces an atypical phosphorylation of IRF3 that is inhibitory since it prevents subsequent IRF3 homodimerization and induction of target genes. Using a mutant virus unable to express the viral kinase ORF47p, we demonstrated that (i) IRF3 slower-migrating form disappears; (ii) IRF3 is phosphorylated on serine 396 again and recovers the ability to form homodimers; (iii) amounts of IRF3 target genes such as IFN-β and ISG15 mRNA are greater than in cells infected with the wild-type virus; and (iv) IRF3 physically interacts with ORF47p. These data led us to hypothesize that the viral kinase ORF47p is involved in the atypical phosphorylation of IRF3 during VZV infection, which prevents its homodimerization and subsequent induction of target genes such as IFN-β and ISG15.
Collapse
Affiliation(s)
- Patricia Vandevenne
- GIGA-Research, Laboratory of Virology and Immunology, University of Liege, Liege, Belgium
| | - Marielle Lebrun
- GIGA-Research, Laboratory of Virology and Immunology, University of Liege, Liege, Belgium
| | - Nadia El Mjiyad
- Laboratory of Molecular Oncology (LOM), Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Isabelle Ote
- GIGA-Research, Laboratory of Virology and Immunology, University of Liege, Liege, Belgium
| | - Emmanuel Di Valentin
- GIGA-Research, Laboratory of Virology and Immunology, University of Liege, Liege, Belgium
| | - Yvette Habraken
- GIGA-Research, Laboratory of Virology and Immunology, University of Liege, Liege, Belgium
| | - Estelle Dortu
- Department of Pathology, University of Liege, Liege, Belgium
| | - Jacques Piette
- GIGA-Research, Laboratory of Virology and Immunology, University of Liege, Liege, Belgium
| | - Catherine Sadzot-Delvaux
- GIGA-Research, Laboratory of Virology and Immunology, University of Liege, Liege, Belgium
- * E-mail:
| |
Collapse
|
35
|
Varicella-zoster virus neurotropism in SCID mouse-human dorsal root ganglia xenografts. Curr Top Microbiol Immunol 2010; 342:255-76. [PMID: 20225014 DOI: 10.1007/82_2009_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Varicella-zoster virus (VZV) is a neurotropic human alphaherpesvirus and the causative agent of varicella and herpes zoster. VZV reactivation from latency in sensory nerve ganglia is a direct consequence of VZV neurotropism. Investigation of VZV neuropathogenesis by infection of human dorsal root ganglion xenografts in immunocompromised (SCID) mice has provided a novel system in which to examine VZV neurotropism. Experimental infection with recombinant VZV mutants with targeted deletions or mutations of specific genes or regulatory elements provides an opportunity to assess gene candidates that may mediate neurotropism and neurovirulence. The SCID mouse-human DRG xenograft model may aid in the development of clinical strategies in the management of herpes zoster as well as in the development of "second generation" neuroattenuated vaccines.
Collapse
|
36
|
Varicella-zoster virus immediate-early protein 62 blocks interferon regulatory factor 3 (IRF3) phosphorylation at key serine residues: a novel mechanism of IRF3 inhibition among herpesviruses. J Virol 2010; 84:9240-53. [PMID: 20631144 DOI: 10.1128/jvi.01147-10] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Varicella-zoster virus (VZV) is an alphaherpesvirus that is restricted to humans. VZV infection of differentiated cells within the host and establishment of latency likely require evasion of innate immunity and limited secretion of antiviral cytokines. Since interferons (IFNs) severely limit VZV replication, we examined the ability of VZV to modulate the induction of the type I IFN response in primary human embryonic lung fibroblasts (HELF). IFN-beta production was not detected, and transcription of two interferon response factor 3 (IRF3)-dependent interferon-stimulated genes (ISGs), ISG54 and ISG56, in response to poly(I:C) stimulation was downregulated in VZV-infected HELF. Inhibition of IRF3 function did not require VZV replication; the viral immediate-early protein 62 (IE62) alone was sufficient to produce this effect. IE62 blocked TBK1-mediated IFN-beta secretion and IRF3 function, as shown in an IFN-stimulated response element (ISRE)-luciferase reporter assay. However, IRF3 function was preserved if constitutively active IRF3 (IRF3-5D) was expressed in VZV-infected or IE62-transfected cells, indicating that VZV interferes with IRF3 phosphorylation. IE62-mediated inhibition was mapped to blocking phosphorylation of at least three serine residues on IRF3. However, IE62 binding to TBK1 or IRF3 was not detected and IE62 did not perturb TBK1-IRF3 complex formation. IE62-mediated inhibition of IRF3 function was maintained even if IE62 transactivator activity was disrupted. Thus, IE62 has two critical but discrete roles following VZV entry: to induce expression of VZV genes and to disarm the IFN-dependent antiviral defense through a novel mechanism that prevents IRF3 phosphorylation.
Collapse
|
37
|
Vandevenne P, Sadzot-Delvaux C, Piette J. Innate immune response and viral interference strategies developed by human herpesviruses. Biochem Pharmacol 2010; 80:1955-72. [PMID: 20620129 DOI: 10.1016/j.bcp.2010.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 07/01/2010] [Accepted: 07/01/2010] [Indexed: 12/24/2022]
Abstract
Viruses are by far the most abundant parasites on earth and they have been found to infect animals, plants and bacteria. However, different types of viruses can only infect a limited range of hosts and many are species-specific. Herpesviruses constitute a large family of DNA viruses that cause diseases in animals, including humans and that are known to undergo lytic or latent infections. Consequently, they developed numerous strategies to counteract host antiviral responses to escape immune surveillance. Innate immune response constitutes the first line of host defence that limits the viral spread and also plays an important role in the activation of adaptive immune response. Viral components are recognized by specific host Pathogen Recognition Receptors (PRRs) which trigger the activation of IRF3, NF-κB and AP-1, three regulators of IFN-β expression. IFN-β is responsible for the induction of Interferon-Stimulated Genes (ISGs) that encode antiviral effectors important to limit the viral spread and to establish an antiviral state as well in the infected cells as in the neighbouring non-infected cells. In this review, we will summarize how host cells recognize viral components and activate downstream signalling pathways leading to the production of IFN-β and ISGs. We will also review the most recent findings in Herpesviruses-encoded proteins involved in host immune evasion.
Collapse
Affiliation(s)
- Patricia Vandevenne
- Laboratory of Virology and Immunology, GIGA-Research B34, University of Liège, B-4000 Liège, Belgium
| | | | | |
Collapse
|
38
|
Varicella-zoster virus T cell tropism and the pathogenesis of skin infection. Curr Top Microbiol Immunol 2010; 342:189-209. [PMID: 20397071 DOI: 10.1007/82_2010_29] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Varicella-zoster virus (VZV) is a medically important human alphaherpesvirus that causes varicella and zoster. VZV initiates primary infection by inoculation of the respiratory mucosa. In the course of primary infection, VZV establishes a life-long persistence in sensory ganglia; VZV reactivation from latency may result in zoster in healthy and immunocompromised patients. The VZV genome has at least 70 known or predicted open reading frames (ORFs), but understanding how these gene products function in virulence is difficult because VZV is a highly human-specific pathogen. We have addressed this obstacle by investigating VZV infection of human tissue xenografts in the severe combined immunodeficiency mouse model. In studies relevant to the pathogenesis of primary VZV infection, we have examined VZV infection of human T cell (thymus/liver) and skin xenografts. This work supports a new paradigm for VZV pathogenesis in which VZV T cell tropism provides a mechanism for delivering the virus to skin. We have also shown that VZV-infected T cells transfer VZV to neurons in sensory ganglia. The construction of infectious VZV recombinants that have deletions or targeted mutations of viral genes or their promoters and the evaluation of VZV mutants in T cell and skin xenografts has revealed determinants of VZV virulence that are important for T cell and skin tropism in vivo.
Collapse
|
39
|
Abstract
The capacity of varicella zoster virus (VZV) to cause varicella (chickenpox) relies upon multiple steps, beginning with inoculation of the host at mucosal sites with infectious virus in respiratory droplets. Despite the presence of a powerful immune defense system, this virus is able to disseminate from the site of initial infection to multiple sites, resulting in the emergence of distinctive cutaneous vesiculopustular lesions. Most recently, it has been proposed that the steps leading to cutaneous infection include VZV infecting human tonsillar CD4(+) T cells that express skin homing markers that allow them to transport VZV directly from the lymph node to the skin during the primary viremia. It has also been proposed that dendritic cells (DC) of the respiratory mucosa may be among the first cells to encounter VZV and these cells may transport virus to the draining lymph node. These various virus-host cell interactions would all need to occur in the face of an intact host immune response for the virus to successfully cause disease. Significantly, following primary exposure to VZV, there is a prolonged incubation period before emergence of skin lesions, during which time the adaptive immune response is delayed. For these reasons, it has been proposed that VZV must encode functions which benefit the virus by evading the immune response. This chapter will review the diverse array of immunomodulatory mechanisms identified to date that VZV has evolved to at least transiently limit immune recognition.
Collapse
|
40
|
Li H, Zhan T, Li C, Liu M, Wang QK. Repression of MHC class I transcription by HPV16E7 through interaction with a putative RXRbeta motif and NF-kappaB cytoplasmic sequestration. Biochem Biophys Res Commun 2009; 388:383-8. [PMID: 19665994 DOI: 10.1016/j.bbrc.2009.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Accepted: 08/04/2009] [Indexed: 11/17/2022]
Abstract
Down-regulation of transcription of the MHC class I genes in HPV16 tumorigenic cells is partly due to HPV16E7 associated with the MHC class I promoter and repressed chromatin activation. In this study, we further demonstrated that HPV16E7 is physically associated with a putative RXRbeta binding motif (GGTCA) of the proximal promoter of the MHC class I genes by using reporter transcriptional assays and chromatin immunoprecipitation assays. Our data also provide evidence that HPV16E7 inhibits TNF-alpha-induced up-regulation of MHC class I transcription by impaired nuclear translocation of NF-kappaB. More importantly, CaSki tumor cells treated with TSA and transfected with the constitutively active mutant form of IKK-alpha (which can activate NF-kappaB directly) showed a maximal level of up-regulation of MHC-I expression. Taken together, our results suggest that HPV16E7 may employ two independent mechanisms to ensure that either the constitutive or inducible transcription of MHC class I genes is down-regulated.
Collapse
Affiliation(s)
- Hui Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | | | | | | | | |
Collapse
|
41
|
Guven A, Kesik V, Deveci MS, Ugurel MS, Ozturk H, Koseoglu V. Post varicella hepatic actinomycosis in a 5-year-old girl mimicking acute abdomen. Eur J Pediatr 2008; 167:1199-201. [PMID: 18066595 DOI: 10.1007/s00431-007-0639-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 11/07/2007] [Indexed: 12/15/2022]
Abstract
Actinomycosis is an indolent, slowly progressive infection caused by gram-positive, anaerobic or microaerophilic bacteria. Hepatic involvement is rare and generally secondary to abdominal or thoracic actinomycosis. Hepatic actinomycosis in children may mimic a wide variety of diseases and thus make the diagnosis much more challenging. Here, we report a 5-year-old girl with apparently primary hepatic actinomycosis mimicking acute abdomen 2 weeks after varicella. The diagnosis was made by ultrasonic guided fine needle aspiration biopsy of a hypoechoic lesion of 3.5 cm diameter in the liver showing sulfur granules surrounded by neutrophils. Hepatic actinomycosis should be taken into account when evaluating acute abdomen symptoms in children.
Collapse
Affiliation(s)
- Ahmet Guven
- Department of Pediatric Surgery, Gulhane Military Medical Academy, Etlik/Ankara, 06017, Turkey.
| | | | | | | | | | | |
Collapse
|
42
|
Desloges N, Rahaus M, Wolff MH. The phosphorylation profile of protein kinase A substrates is modulated during Varicella-zoster virus infection. Med Microbiol Immunol 2007; 197:353-60. [PMID: 18066594 DOI: 10.1007/s00430-007-0068-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Indexed: 12/15/2022]
Abstract
The cAMP-dependent protein kinase A (PKA) is a key enzyme for many cellular mechanisms. In this study, we investigated the importance of this kinase for the replication of the alphaherpesvirus Varicella-zoster virus (VZV). We report that the expression of the catalytic subunit of PKA was strongly increased at the beginning of the viral cycle. The presence of a peptide inhibitor of PKA had no consequence on viral replication in a melanoma cell line whereas in fibroblasts, it resulted in a drastic decrease of replication. An overall analysis of PKA substrates phosphorylation patterns during VZV replication showed that the phosphorylation of PKA substrates was modulated. These results were completed by investigating the accumulation and phosphorylation patterns of the PKA target cAMP response element binding protein (CREB). This transcription factor remained available throughout the VZV replication, but its phosphorylation decreased in the early phase of infection before it rose later on. These results indicate that the PKA signalling plays a cell-type dependent role for VZV replication and that the infection resulted in a regulated CREB-dependent gene expression.
Collapse
Affiliation(s)
- Nathalie Desloges
- Institute of Microbiology and Virology, Private University Witten/Herdecke gGmbH, Stockumer Str. 10, 58448, Witten, Germany.
| | | | | |
Collapse
|
43
|
Desloges N, Schubert C, Wolff MH, Rahaus M. Varicella-zoster virus infection induces the secretion of interleukin-8. Med Microbiol Immunol 2007; 197:277-84. [PMID: 17909856 DOI: 10.1007/s00430-007-0060-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Indexed: 10/22/2022]
Abstract
Interleukin-8 (IL-8) is an important mediator in neutrophil-mediated acute inflammation but has also a wide range of actions on various cells types. We demonstrated that infection of melanoma cells and fibroblasts with cell-associated varicella-zoster virus (VZV) and infection of a T cell line with cell-free VZV resulted in an induction of IL-8 secretion in vitro. The inhibition of the VZV replication with a drug interfering with its DNA replication had no effect on the IL-8 release. Since the IL-8 promoter contains binding sites for NF-kappaB and AP-1, melanoma cells and the T cell line were treated with inhibitors of NF-kappaB, JNK/SAPK or p38/MAPK prior to infection. In melanoma cells, the JNK/SAPK pathway was shown to be important for the IL-8 secretion during the VZV replication, whereas in the T cell line, not only the JNK/SAPK but also the p38/MAPK pathways were required for IL-8 secretion. The neutralisation of the IL-8 bioactivity had no significant consequence on the VZV replication, suggesting that IL-8 acts neither as a proviral nor as an antiviral cytokine during the VZV replication in vitro.
Collapse
Affiliation(s)
- Nathalie Desloges
- Institute of Microbiology and Virology, Private University Witten/Herdecke gGmbH, Stockumer Strasse 10, Witten, Germany
| | | | | | | |
Collapse
|
44
|
El Mjiyad N, Bontems S, Gloire G, Horion J, Vandevenne P, Dejardin E, Piette J, Sadzot-Delvaux C. Varicella-zoster virus modulates NF-kappaB recruitment on selected cellular promoters. J Virol 2007; 81:13092-104. [PMID: 17855547 PMCID: PMC2169121 DOI: 10.1128/jvi.01378-07] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intercellular adhesion molecule 1 (ICAM-1) expression is down-regulated in the center of cutaneous varicella lesions despite the expression of proinflammatory cytokines such as gamma interferon and tumor necrosis factor alpha (TNF-alpha). To study the molecular basis of this down-regulation, the ICAM-1 induction of TNF-alpha was analyzed in varicella-zoster virus (VZV)-infected melanoma cells (MeWo), leading to the following observations: (i) VZV inhibits the stimulation of icam-1 mRNA synthesis; (ii) despite VZV-induced nuclear translocation of p65, p52, and c-Rel, p50 does not translocate in response to TNF-alpha; (iii) the nuclear p65 present in VZV-infected cells is no longer associated with p50 and is unable to bind the proximal NF-kappaB site of the icam-1 promoter, despite an increased acetylation and accessibility of the promoter in response to TNF-alpha; and (iv) VZV induces the nuclear accumulation of the NF-kappaB inhibitor p100. VZV also inhibits icam-1 stimulation of TNF-alpha by strongly reducing NF-kappaB nuclear translocation in MRC5 fibroblasts. Taken together, these data show that VZV interferes with several aspects of the immune response by inhibiting NF-kappaB binding and the expression of target genes. Targeting NF-kappaB activation, which plays a central role in innate and adaptive immune responses, leads to obvious advantages for the virus, particularly in melanocytes, which are a site of viral replication in the skin.
Collapse
Affiliation(s)
- Nadia El Mjiyad
- GIGA-Research, Virology and Immunology Unit, GIGA B34, University of Liège, B-4000 Liège, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Tong Y, Qian J, Li Y, Meng H, Jin J. The high incidence of varicella herpes zoster with the use of bortezomib in 10 patients. Am J Hematol 2007; 82:403-4. [PMID: 17133426 DOI: 10.1002/ajh.20838] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Bortezomib, a proteasome inhibitor, has been used for patients with refractory and relapsed multiple myeloma, lymphoma and leukemia. We used bortezomib in ten refractory or relapsed patients (seven of multiple myeloma, two of lymphoma and one of acute myeloblastic leukemia). Six out of ten (60%) patients developed varicella herpes zoster after the complete of one cycle of bortezomib. The incidence of varicella herpes zoster was higher than reported in the literature. It may be due to immunosuppression caused by the combination of high-dose dexamethasone or other drugs. We considered that prophylactic antiviral medication could be used in predisposed patients to reduce the incidence of varicella herpes zoster.
Collapse
Affiliation(s)
- Yin Tong
- Department of Hematology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | |
Collapse
|
46
|
Venkatachari NJ, Majumder B, Ayyavoo V. Human immunodeficiency virus (HIV) type 1 Vpr induces differential regulation of T cell costimulatory molecules: Direct effect of Vpr on T cell activation and immune function. Virology 2007; 358:347-56. [PMID: 17023015 DOI: 10.1016/j.virol.2006.08.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Revised: 07/18/2006] [Accepted: 08/22/2006] [Indexed: 10/24/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) viral proteins disrupt the normal host cellular immune pathways thus exploiting the cellular machinery for replication, survival and to escape host immune attack. Here we evaluated the direct effects of HIV-1 Vpr-mediated immune modulation of infected T cells. Vpr specifically downregulated the expression of CD28 and increased the expression of CTLA-4, whereas no significant difference in the expression of CD25 and HLA-DR was observed. Interferon gamma (IFN-gamma) production in T cells was evaluated as a measure of the downstream effector functions. Results indicate that Vpr significantly inhibited IFN-gamma production and this may, in part, due to Vpr's ability to inhibit the nuclear translocation of NF-kappaB, and its transcriptional regulation. Together these results support that HIV-1 Vpr selectively dysregulates the immune functions at multiple levels and exerts its inhibitory effects in the presence of other viral proteins.
Collapse
Affiliation(s)
- Narasimhan J Venkatachari
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, 130 Desoto Street, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
47
|
Ramilo O, Allman W, Chung W, Mejias A, Ardura M, Glaser C, Wittkowski KM, Piqueras B, Banchereau J, Palucka AK, Chaussabel D. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood 2006; 109:2066-77. [PMID: 17105821 PMCID: PMC1801073 DOI: 10.1182/blood-2006-02-002477] [Citation(s) in RCA: 372] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Each infectious agent represents a unique combination of pathogen-associated molecular patterns that interact with specific pattern-recognition receptors expressed on immune cells. Therefore, we surmised that the blood immune cells of individuals with different infections might bear discriminative transcriptional signatures. Gene expression profiles were obtained for 131 peripheral blood samples from pediatric patients with acute infections caused by influenza A virus, Gram-negative (Escherichia coli) or Gram-positive (Staphylococcus aureus and Streptococcus pneumoniae) bacteria. Thirty-five genes were identified that best discriminate patients with influenza A virus infection from patients with either E coli or S pneumoniae infection. These genes classified with 95% accuracy (35 of 37 samples) an independent set of patients with either influenza A, E coli, or S pneumoniae infection. A different signature discriminated patients with E coli versus S aureus infections with 85% accuracy (34 of 40). Furthermore, distinctive gene expression patterns were observed in patients presenting with respiratory infections of different etiologies. Thus, microarray analyses of patient peripheral blood leukocytes might assist in the differential diagnosis of infectious diseases.
Collapse
Affiliation(s)
- Octavio Ramilo
- Baylor National Institute of Allergy and Infections Diseases (NIAID) Cooperative Center for Translational Research on Human Immunology and Biodefense and Baylor Institute for Immunology Research, Dallas, TX 75204, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|