1
|
Tucker MH, Kalamvoki M, Tilak K, Raje N, Sampath V. The immunogenetic basis of severe herpes simplex infections in neonates and children: a review. Pediatr Res 2025; 97:1370-1380. [PMID: 39827257 DOI: 10.1038/s41390-025-03830-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Human herpes simplex virus (HSV) is a double stranded DNA virus with two distinct types, HSV-1 and HSV-2. The global burden of HSV is high with an estimated 2/3 of the adult population seropositive for at least one of these types of HSV. HSV rarely causes life-threatening disease in immunocompetent children and adults. However, in neonates and children with a developmentally immature immune system it can cause disseminated disease and herpes simplex encephalitis (HSE). Recent studies in children and neonates suggest that genetic risk-factors can contribute to severe HSV phenotypes in neonates and children. In particular, genetic defects in the Toll Like Receptor 3 (TLR3) signaling pathway that attenuate the type I interferon response to HSV are being increasingly recognized in children with severe phenotypes of HSV. In this review, we discuss the epidemiology and immunological aspects of HSV disease in neonates and children and provide an in-depth review of the studies characterizing the role of inborn errors in the TLR3 pathway and other immune genes in HSV. We highlight the need for future research to identify the immunogenetic basis of severe or recurrent HSV disease in neonates and children. IMPACT: Review the epidemiology and phenotypes of herpes simplex virus (HSV) infection in neonates and children. Discuss the mechanisms of immunity against HSV highlighting the developmental vulnerability of neonates and infants to severe HSV disease. Explore in depth the genes and immune pathways that underlie genetic predisposition to severe HSV disease in neonates and children, and outline strategies for multi-disciplinary clinical evaluation of severe disease.
Collapse
Affiliation(s)
- Megan H Tucker
- Department of Neonatology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA.
| | - Maria Kalamvoki
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kedar Tilak
- Department of Neonatology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Nikita Raje
- Department of Allergy and Immunology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Venkatesh Sampath
- Department of Neonatology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA
| |
Collapse
|
2
|
Zhang SY, Casanova JL. Genetic defects of brain immunity in childhood herpes simplex encephalitis. Nature 2024; 635:563-573. [PMID: 39567785 PMCID: PMC11822754 DOI: 10.1038/s41586-024-08119-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/25/2024] [Indexed: 11/22/2024]
Abstract
Herpes simplex virus 1 (HSV-1) encephalitis (HSE) is the most common sporadic viral encephalitis in humans. It is life-threatening and has a first peak of incidence in childhood, during primary infection. Children with HSE are not particularly prone to other infections, including HSV-1 infections of tissues other than the brain. About 8-10% of childhood cases are due to monogenic inborn errors of 19 genes, two-thirds of which are recessive, and most of which display incomplete clinical penetrance. Childhood HSE can therefore be sporadic but genetic, enabling new diagnostic and therapeutic approaches. In this Review, we examine essential cellular and molecular mechanisms of cell-intrinsic antiviral immunity in the brain that are disrupted in individuals with HSE. These mechanisms include both known (such as mutations in the TLR3 pathway) and previously unknown (such as the TMEFF1 restriction factor) antiviral pathways, which may be dependent (for example, IFNAR1) or independent (for example, through RIPK3) of type I interferons. They operate in cortical or brainstem neurons, and underlie forebrain and brainstem infections, respectively. Conversely, the most severe inborn errors of leukocytes, including a complete lack of myeloid and/or lymphoid blood cells, do not underlie HSE. Thus congenital defects in intrinsic immunity in brain-resident neurons that underlie HSE broaden natural host defences against HSV-1 from the leukocytes of the immune system to other cells in the organism.
Collapse
Affiliation(s)
- Shen-Ying Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France.
- Paris Cité University, Imagine Institute, Paris, France.
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France.
- Paris Cité University, Imagine Institute, Paris, France.
- Howard Hughes Medical Institute, New York, NY, USA.
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France.
| |
Collapse
|
3
|
Huff HV, Wilson-Murphy M. Neuroinfectious Diseases in Children: Pathophysiology, Outcomes, and Global Challenges. Pediatr Neurol 2024; 151:53-64. [PMID: 38103523 DOI: 10.1016/j.pediatrneurol.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/22/2023] [Accepted: 09/24/2023] [Indexed: 12/19/2023]
Abstract
Pathogens with affinity for the central nervous system (CNS) in children are diverse in their mechanisms of infecting and attacking the brain. Infections can reach the CNS via hematogenous routes, transneurally thereby avoiding the blood-brain barrier, and across mucosal or skin surfaces. Once transmission occurs, pathogens can wreak havoc both by direct action on host cells and via an intricate interplay between the protective and pathologic actions of the host's immune system. Pathogen prevalence varies depending on region, and susceptibility differs based on epidemiologic factors such as age, immune status, and genetics. In addition, some infectious diseases are monophasic, whereas others may lie dormant for years, thereby causing a dynamic effect on outcomes. Outcomes in survivors are highly variable for each particular pathogen and depend on the vaccination and immune status of the patient as well as the speed by which the patient receives evidence-based treatments. Given pathogens cause communicable diseases that can cause morbidity and mortality on a population level when spread, the burden is often the greatest and the outcomes the worst in low-resource settings. Here we will focus on the most common infections with a propensity to affect a child's brain, the pathologic mechanisms by which they do so, and what is known about the developmental outcomes in children who are affected by these infections.
Collapse
Affiliation(s)
- Hanalise V Huff
- Department of Neurology, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
4
|
Zou Y, Kamoi K, Zong Y, Zhang J, Yang M, Ohno-Matsui K. Ocular Inflammation Post-Vaccination. Vaccines (Basel) 2023; 11:1626. [PMID: 37897028 PMCID: PMC10611055 DOI: 10.3390/vaccines11101626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
The association between vaccines and ocular disorders has attracted significant attention in scientific research. Numerous mainstream vaccines are associated with a range of uveitis types, including anterior, intermediate, and posterior uveitis. Additionally, they are associated with distinct ocular diseases such as multifocal choroiditis, Vogt-Koyanagi-Harada (VKH) disease, acute posterior multifocal placoid pigment epitheliopathy (APMPPE), and multiple evanescent white dot syndrome (MEWDS). These ocular conditions are often transient, with a vast majority of patients experiencing improvement after steroid intervention. To date, numerous cases of vaccine-induced uveitis have been reported. This study analyzed the correlation between antiviral vaccines, including the hepatitis B virus (HBV), human papillomavirus (HPV), measles-mumps-rubella (MMR), varicella zoster virus (VZV), and influenza vaccines, and different manifestations of uveitis. This is the first comprehensive study to offer a detailed analysis of uveitis types induced by antiviral vaccines. Through an extensive database search, we found a particularly strong link between influenza vaccines, followed by VZV and HPV vaccines. While anterior uveitis is common, conditions such as APMPPE, MEWDS, and VKH are particularly notable and merit careful consideration in clinical practice. Corticosteroid treatment was effective; however, half of the observed patients did not achieve full recovery, indicating potentially prolonged effects of the vaccine.
Collapse
Affiliation(s)
| | - Koju Kamoi
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; (Y.Z.); (Y.Z.); (J.Z.); (M.Y.); (K.O.-M.)
| | | | | | | | | |
Collapse
|
5
|
Liu Z, Garcia Reino EJ, Harschnitz O, Guo H, Chan YH, Khobrekar NV, Hasek ML, Dobbs K, Rinchai D, Materna M, Matuozzo D, Lee D, Bastard P, Chen J, Lee YS, Kim SK, Zhao S, Amin P, Lorenzo L, Seeleuthner Y, Chevalier R, Mazzola L, Gay C, Stephan JL, Milisavljevic B, Boucherit S, Rozenberg F, Perez de Diego R, Dix RD, Marr N, Béziat V, Cobat A, Aubart M, Abel L, Chabrier S, Smith GA, Notarangelo LD, Mocarski ES, Studer L, Casanova JL, Zhang SY. Encephalitis and poor neuronal death-mediated control of herpes simplex virus in human inherited RIPK3 deficiency. Sci Immunol 2023; 8:eade2860. [PMID: 37083451 PMCID: PMC10337828 DOI: 10.1126/sciimmunol.ade2860] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 03/30/2023] [Indexed: 04/22/2023]
Abstract
Inborn errors of TLR3-dependent type I IFN immunity in cortical neurons underlie forebrain herpes simplex virus-1 (HSV-1) encephalitis (HSE) due to uncontrolled viral growth and subsequent cell death. We report an otherwise healthy patient with HSE who was compound heterozygous for nonsense (R422*) and frameshift (P493fs9*) RIPK3 variants. Receptor-interacting protein kinase 3 (RIPK3) is a ubiquitous cytoplasmic kinase regulating cell death outcomes, including apoptosis and necroptosis. In vitro, the R422* and P493fs9* RIPK3 proteins impaired cellular apoptosis and necroptosis upon TLR3, TLR4, or TNFR1 stimulation and ZBP1/DAI-mediated necroptotic cell death after HSV-1 infection. The patient's fibroblasts displayed no detectable RIPK3 expression. After TNFR1 or TLR3 stimulation, the patient's cells did not undergo apoptosis or necroptosis. After HSV-1 infection, the cells supported excessive viral growth despite normal induction of antiviral IFN-β and IFN-stimulated genes (ISGs). This phenotype was, nevertheless, rescued by application of exogenous type I IFN. The patient's human pluripotent stem cell (hPSC)-derived cortical neurons displayed impaired cell death and enhanced viral growth after HSV-1 infection, as did isogenic RIPK3-knockout hPSC-derived cortical neurons. Inherited RIPK3 deficiency therefore confers a predisposition to HSE by impairing the cell death-dependent control of HSV-1 in cortical neurons but not their production of or response to type I IFNs.
Collapse
Affiliation(s)
- Zhiyong Liu
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Eduardo J Garcia Reino
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Oliver Harschnitz
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY, USA
- Human Technopole, Viale Rita Levi-Montalcini, Milan, Italy
| | - Hongyan Guo
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, GA, USA
- School of Medicine, Atlanta, GA, USA
- Louisiana State University Health Sciences Center at Shreveport (LSUHSC-S), Shreveport, LA, USA
| | - Yi-Hao Chan
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Noopur V Khobrekar
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Mary L Hasek
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Daniela Matuozzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Danyel Lee
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Jie Chen
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Yoon Seung Lee
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | | | - Shuxiang Zhao
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Param Amin
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Remi Chevalier
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Laure Mazzola
- Department of Pediatrics, Hôpital Nord, Saint-Etienne, Paris, France
| | - Claire Gay
- Department of Pediatrics, Hôpital Nord, Saint-Etienne, Paris, France
| | | | - Baptiste Milisavljevic
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Soraya Boucherit
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Flore Rozenberg
- Laboratory of Virology, Assistance Publique-Hôpitaux de Paris (AP-HP), Cochin Hospital, Paris, France
| | - Rebeca Perez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Richard D Dix
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, GA, USA
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nico Marr
- Research Branch, Sidra Medicine, Doha, Qatar
- Institute of Translational Immunology, Brandenburg Medical School, Brandenburg an der Havel, Germany
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Vivien Béziat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Aurelie Cobat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Mélodie Aubart
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Pediatric Neurology Department, Necker Hospital for Sick Children, APHP, Paris City University, Paris, France
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| | - Stephane Chabrier
- Department of Pediatrics, Hôpital Nord, Saint-Etienne, Paris, France
| | - Gregory A Smith
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Edward S Mocarski
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University, GA, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris City University, Imagine Institute, Paris, France
| |
Collapse
|
6
|
Lang R, Li H, Luo X, Liu C, Zhang Y, Guo S, Xu J, Bao C, Dong W, Yu Y. Expression and mechanisms of interferon-stimulated genes in viral infection of the central nervous system (CNS) and neurological diseases. Front Immunol 2022; 13:1008072. [PMID: 36325336 PMCID: PMC9618809 DOI: 10.3389/fimmu.2022.1008072] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/28/2022] [Indexed: 09/16/2023] Open
Abstract
Interferons (IFNs) bind to cell surface receptors and activate the expression of interferon-stimulated genes (ISGs) through intracellular signaling cascades. ISGs and their expression products have various biological functions, such as antiviral and immunomodulatory effects, and are essential effector molecules for IFN function. ISGs limit the invasion and replication of the virus in a cell-specific and region-specific manner in the central nervous system (CNS). In addition to participating in natural immunity against viral infections, studies have shown that ISGs are essential in the pathogenesis of CNS disorders such as neuroinflammation and neurodegenerative diseases. The aim of this review is to present a macroscopic overview of the characteristics of ISGs that restrict viral neural invasion and the expression of the ISGs underlying viral infection of CNS cells. Furthermore, we elucidate the characteristics of ISGs expression in neurological inflammation, neuropsychiatric disorders such as depression as well as neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Finally, we summarize several ISGs (ISG15, IFIT2, IFITM3) that have been studied more in recent years for their antiviral infection in the CNS and their research progress in neurological diseases.
Collapse
Affiliation(s)
- Rui Lang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huiting Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiaoqin Luo
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Cencen Liu
- Department of Pathology, People’s Hospital of Zhongjiang County, DeYang, China
| | - Yiwen Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - ShunYu Guo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingyi Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Changshun Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurological diseases and brain function laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
7
|
Abstract
Two of the most prevalent human viruses worldwide, herpes simplex virus type 1 and type 2 (HSV-1 and HSV-2, respectively), cause a variety of diseases, including cold sores, genital herpes, herpes stromal keratitis, meningitis and encephalitis. The intrinsic, innate and adaptive immune responses are key to control HSV, and the virus has developed mechanisms to evade them. The immune response can also contribute to pathogenesis, as observed in stromal keratitis and encephalitis. The fact that certain individuals are more prone than others to suffer severe disease upon HSV infection can be partially explained by the existence of genetic polymorphisms in humans. Like all herpesviruses, HSV has two replication cycles: lytic and latent. During lytic replication HSV produces infectious viral particles to infect other cells and organisms, while during latency there is limited gene expression and lack of infectious virus particles. HSV establishes latency in neurons and can cause disease both during primary infection and upon reactivation. The mechanisms leading to latency and reactivation and which are the viral and host factors controlling these processes are not completely understood. Here we review the HSV life cycle, the interaction of HSV with the immune system and three of the best-studied pathologies: Herpes stromal keratitis, herpes simplex encephalitis and genital herpes. We also discuss the potential association between HSV-1 infection and Alzheimer's disease.
Collapse
Affiliation(s)
- Shuyong Zhu
- Institute of Virology, Hannover Medical School, Cluster of Excellence RESIST (Exc 2155), Hannover Medical School, Hannover, Germany
| | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Cluster of Excellence RESIST (Exc 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Gao D, Ciancanelli MJ, Zhang P, Harschnitz O, Bondet V, Hasek M, Chen J, Mu X, Itan Y, Cobat A, Sancho-Shimizu V, Bigio B, Lorenzo L, Ciceri G, McAlpine J, Anguiano E, Jouanguy E, Chaussabel D, Meyts I, Diamond MS, Abel L, Hur S, Smith GA, Notarangelo L, Duffy D, Studer L, Casanova JL, Zhang SY. TLR3 controls constitutive IFN-β antiviral immunity in human fibroblasts and cortical neurons. J Clin Invest 2021; 131:134529. [PMID: 33393505 DOI: 10.1172/jci134529] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Human herpes simplex virus 1 (HSV-1) encephalitis can be caused by inborn errors of the TLR3 pathway, resulting in impairment of CNS cell-intrinsic antiviral immunity. Deficiencies of the TLR3 pathway impair cell-intrinsic immunity to vesicular stomatitis virus (VSV) and HSV-1 in fibroblasts, and to HSV-1 in cortical but not trigeminal neurons. The underlying molecular mechanism is thought to involve impaired IFN-α/β induction by the TLR3 recognition of dsRNA viral intermediates or by-products. However, we show here that human TLR3 controls constitutive levels of IFNB mRNA and secreted bioactive IFN-β protein, and thereby also controls constitutive mRNA levels for IFN-stimulated genes (ISGs) in fibroblasts. Tlr3-/- mouse embryonic fibroblasts also have lower basal ISG levels. Moreover, human TLR3 controls basal levels of IFN-β secretion and ISG mRNA in induced pluripotent stem cell-derived cortical neurons. Consistently, TLR3-deficient human fibroblasts and cortical neurons are vulnerable not only to both VSV and HSV-1, but also to several other families of viruses. The mechanism by which TLR3 restricts viral growth in human fibroblasts and cortical neurons in vitro and, by inference, by which the human CNS prevents infection by HSV-1 in vivo, is therefore based on the control of early viral infection by basal IFN-β immunity.
Collapse
Affiliation(s)
- Daxing Gao
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA.,Department of General Surgery, The First Affiliated Hospital of USTC, and.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Michael J Ciancanelli
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA.,Turnstone Biologics, New York, New York, USA
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Oliver Harschnitz
- The Center for Stem Cell Biology, and.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA
| | - Vincent Bondet
- Translational Immunology Laboratory, Pasteur Institute, Paris, France
| | - Mary Hasek
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jie Chen
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Xin Mu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, and.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France
| | - Vanessa Sancho-Shimizu
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France.,Department of Paediatric Infectious Diseases, Division of Medicine, Imperial College London, Norfolk Place, United Kingdom
| | - Benedetta Bigio
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France
| | - Gabriele Ciceri
- The Center for Stem Cell Biology, and.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA
| | - Jessica McAlpine
- The Center for Stem Cell Biology, and.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA
| | - Esperanza Anguiano
- Baylor Institute for Immunology Research/ANRS Center for Human Vaccines, INSERM U899, Dallas, Texas, USA
| | - Emmanuelle Jouanguy
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France
| | - Damien Chaussabel
- Baylor Institute for Immunology Research/ANRS Center for Human Vaccines, INSERM U899, Dallas, Texas, USA.,Benaroya Research Institute, Seattle, Washington, USA.,Sidra Medicine, Doha, Qatar
| | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium.,Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.,Precision Immunology Institute and Mindich Child Health and Development Institute at the Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France
| | - Sun Hur
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory A Smith
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Luigi Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Darragh Duffy
- Translational Immunology Laboratory, Pasteur Institute, Paris, France
| | - Lorenz Studer
- The Center for Stem Cell Biology, and.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France.,Pediatric Immunology-Hematology Unit, Necker Hospital for Sick Children, Paris, France.,Howard Hughes Medical Institute, New York, New York, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France
| |
Collapse
|
9
|
Rispoli F, Valencic E, Girardelli M, Pin A, Tesser A, Piscianz E, Boz V, Faletra F, Severini GM, Taddio A, Tommasini A. Immunity and Genetics at the Revolving Doors of Diagnostics in Primary Immunodeficiencies. Diagnostics (Basel) 2021; 11:532. [PMID: 33809703 PMCID: PMC8002250 DOI: 10.3390/diagnostics11030532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 12/14/2022] Open
Abstract
Primary immunodeficiencies (PIDs) are a large and growing group of disorders commonly associated with recurrent infections. However, nowadays, we know that PIDs often carry with them consequences related to organ or hematologic autoimmunity, autoinflammation, and lymphoproliferation in addition to simple susceptibility to pathogens. Alongside this conceptual development, there has been technical advancement, given by the new but already established diagnostic possibilities offered by new genetic testing (e.g., next-generation sequencing). Nevertheless, there is also the need to understand the large number of gene variants detected with these powerful methods. That means advancing beyond genetic results and resorting to the clinical phenotype and to immunological or alternative molecular tests that allow us to prove the causative role of a genetic variant of uncertain significance and/or better define the underlying pathophysiological mechanism. Furthermore, because of the rapid availability of results, laboratory immunoassays are still critical to diagnosing many PIDs, even in screening settings. Fundamental is the integration between different specialties and the development of multidisciplinary and flexible diagnostic workflows. This paper aims to tell these evolving aspects of immunodeficiencies, which are summarized in five key messages, through introducing and exemplifying five clinical cases, focusing on diseases that could benefit targeted therapy.
Collapse
Affiliation(s)
- Francesco Rispoli
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (F.R.); (V.B.); (A.T.); (A.T.)
| | - Erica Valencic
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| | - Martina Girardelli
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| | - Alessia Pin
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| | - Alessandra Tesser
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| | - Elisa Piscianz
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| | - Valentina Boz
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (F.R.); (V.B.); (A.T.); (A.T.)
| | - Flavio Faletra
- Department of Diagnostics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Giovanni Maria Severini
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| | - Andrea Taddio
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (F.R.); (V.B.); (A.T.); (A.T.)
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| | - Alberto Tommasini
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (F.R.); (V.B.); (A.T.); (A.T.)
- Department of Pediatrics, Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (M.G.); (A.P.); (A.T.); (E.P.); (G.M.S.)
| |
Collapse
|
10
|
Pignataro G, Cataldi M, Taglialatela M. Neurological risks and benefits of cytokine-based treatments in coronavirus disease 2019: from preclinical to clinical evidence. Br J Pharmacol 2021; 179:2149-2174. [PMID: 33512003 DOI: 10.1111/bph.15397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/15/2022] Open
Abstract
Immunodeficiency and hyperinflammation are responsible for the most frequent and life-threatening forms of coronavirus disease 2019 (COVID-19). Therefore, cytokine-based treatments targeting immuno-inflammatory mechanisms are currently undergoing clinical scrutiny in COVID-19-affected patients. In addition, COVID-19 patients also exhibit a wide range of neurological manifestations (neuro-COVID), which may also benefit from cytokine-based treatments. In fact, such drugs have shown some clinical efficacy also in neuroinflammatory diseases. On the other hand, anti-cytokine drugs are endowed with significant neurological risks, mainly attributable to their immunodepressant effects. Therefore, the aim of the present manuscript is to briefly describe the role of specific cytokines in neuroinflammation, to summarize the efficacy in preclinical models of neuroinflammatory diseases of drugs targeting these cytokines and to review the clinical data regarding the neurological effects of these drugs currently being investigated against COVID-19, in order to raise awareness about their potentially beneficial and/or detrimental neurological consequences.
Collapse
Affiliation(s)
- Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
11
|
Casanova JL, Abel L. Lethal Infectious Diseases as Inborn Errors of Immunity: Toward a Synthesis of the Germ and Genetic Theories. ANNUAL REVIEW OF PATHOLOGY 2021; 16:23-50. [PMID: 32289233 PMCID: PMC7923385 DOI: 10.1146/annurev-pathol-031920-101429] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It was first demonstrated in the late nineteenth century that human deaths from fever were typically due to infections. As the germ theory gained ground, it replaced the old, unproven theory that deaths from fever reflected a weak personal or even familial constitution. A new enigma emerged at the turn of the twentieth century, when it became apparent that only a small proportion of infected individuals die from primary infections with almost any given microbe. Classical genetics studies gradually revealed that severe infectious diseases could be driven by human genetic predisposition. This idea gained ground with the support of molecular genetics, in three successive, overlapping steps. First, many rare inborn errors of immunity were shown, from 1985 onward, to underlie multiple, recurrent infections with Mendelian inheritance. Second, a handful of rare and familial infections, also segregating as Mendelian traits but striking humans resistant to other infections, were deciphered molecularly beginning in 1996. Third, from 2007 onward, a growing number of rare or common sporadicinfections were shown to result from monogenic, but not Mendelian, inborn errors. A synthesis of the hitherto mutually exclusive germ and genetic theories is now in view.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA;
- Howard Hughes Medical Institute, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Paris University, Imagine Institute, 75015 Paris, France
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, 75015 Paris, France
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA;
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Paris University, Imagine Institute, 75015 Paris, France
| |
Collapse
|
12
|
Role of Melatonin on Virus-Induced Neuropathogenesis-A Concomitant Therapeutic Strategy to Understand SARS-CoV-2 Infection. Antioxidants (Basel) 2021; 10:antiox10010047. [PMID: 33401749 PMCID: PMC7823793 DOI: 10.3390/antiox10010047] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Viral infections may cause neurological disorders by directly inducing oxidative stress and interrupting immune system function, both of which contribute to neuronal death. Several reports have described the neurological manifestations in Covid-19 patients where, in severe cases of the infection, brain inflammation and encephalitis are common. Recently, extensive research-based studies have revealed and acknowledged the clinical and preventive roles of melatonin in some viral diseases. Melatonin has been shown to have antiviral properties against several viral infections which are accompanied by neurological symptoms. The beneficial properties of melatonin relate to its properties as a potent antioxidant, anti-inflammatory, and immunoregulatory molecule and its neuroprotective effects. In this review, what is known about the therapeutic role of melatonin in virus-induced neuropathogenesis is summarized and discussed.
Collapse
|
13
|
Devilliers MJ, Ben Hadj Salah W, Barreau E, Da Cunha E, M'Garrech M, Bénichou J, Labetoulle M, Rousseau A. [Ocular manifestations of viral diseases]. Rev Med Interne 2020; 42:401-410. [PMID: 33168354 PMCID: PMC7646372 DOI: 10.1016/j.revmed.2020.08.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/30/2020] [Indexed: 01/08/2023]
Abstract
Les infections virales peuvent toucher l’ensemble des tuniques oculaires et mettre en jeu la fonction visuelle à plus ou moins courte terme. Les kératites et kérato-uvéites liées au virus HSV-1 sont les atteintes les plus fréquentes. Les récurrences entraînent une opacification cornéenne irréversible qui en fait la première cause de cécité d’origine infectieuse dans les pays occidentaux, et justifient un traitement antiviral préventif au long cours. Le zona ophtalmique (10 à 20 % des zonas), peut s’accompagner d’atteintes oculaires sévères (kératites, kérato-uvéites), dont 30 % deviennent chroniques/récurrentes, et de douleurs post-zostériennes, redoutables dans le territoire trigéminé. Les rétinites nécrosantes liées aux herpesvirus (HSV, VZV, CMV), sont rares mais constituent des urgences fonctionnelles absolues nécessitant un traitement antiviral par voie intraveineuse et intravitréenne. Les conjonctivites à adénovirus constituent la première cause de conjonctivite infectieuse. Le plus souvent bénignes, elles sont extrêmement contagieuses et peuvent se compliquer de lésions cornéennes invalidantes persistant plusieurs mois, voire années. Certaines arboviroses s’accompagnent de manifestations oculaires inflammatoires. Dans le cas du Zika, les infections congénitales peuvent se compliquer d’atrophie maculaire et/ou optique. Les conjonctivites sont très fréquentes à la phase aiguë de la maladie à virus Ebola, dont 15 % des survivants présentent des atteintes inflammatoires chroniques sévères liées à la persistance du virus dans les tissus uvéaux. Enfin, dans le cadre de la COVID-19, les conjonctivites ne sont pas très fréquentes mais peuvent être inaugurales, voire au premier plan et sont associées à une excrétion virale lacrymale qui doit faire prendre toutes les précautions.
Collapse
Affiliation(s)
- M-J Devilliers
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France
| | - W Ben Hadj Salah
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France
| | - E Barreau
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France
| | - E Da Cunha
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France
| | - M M'Garrech
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France
| | - J Bénichou
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France
| | - M Labetoulle
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France; Département d'immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT) CEA, Fontenay-aux-Roses, France
| | - A Rousseau
- Service d'ophtalmologie, CHU de Bicêtre, Assistance publique-Hôpitaux de Paris, université Paris-Saclay, CRMR OPHTARA (maladies rares en ophtalmologie), 78, rue du Général-Leclerc, 94275 Le Kremlin-Bicêtre, France; Département d'immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT) CEA, Fontenay-aux-Roses, France.
| |
Collapse
|
14
|
Abstract
Herpes simplex virus 1 (HSV-1) can be responsible for life-threatening HSV encephalitis (HSE). The mortality rate of patients with HSE who do not receive antiviral treatment is 70%, with most survivors suffering from permanent neurological sequelae. The use of intravenous acyclovir together with improved diagnostic technologies such as PCR and magnetic resonance imaging has resulted in a reduction in the mortality rate to close to 20%. However, 70% of surviving patients still do not recover complete neurological functions. Thus, there is an urgent need to develop more effective treatments for a better clinical outcome. It is well recognized that cerebral damage resulting from HSE is caused by viral replication together with an overzealous inflammatory response. Both of these processes constitute potential targets for the development of innovative therapies against HSE. In this review, we discuss recent progress in therapy that may be used to ameliorate the outcome of patients with HSE, with a particular emphasis on immunomodulatory agents. Ideally, the administration of adjunctive immunomodulatory drugs should be initiated during the rise of the inflammatory response, and its duration should be limited in time to reduce undesired effects. This critical time frame should be optimized by the identification of reliable biomarkers of inflammation.
Collapse
|
15
|
Ivanova AM, Imomalieva KM, Narovlyansky AN, Izmest'eva AV, Sarymsakov AA, Bilalov EN, Ershov FI. [Ophthalmic drug films in the therapy of experimental herpetic keratoconjunctivitis.]. Vopr Virusol 2019; 64:238-245. [PMID: 32167689 DOI: 10.36233/0507-4088-2019-64-5-238-245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The complexity of the treatment of herpetic keratoconjunctivitis is due to the severity of the disease, complications, the transition to chronic relapsing forms and the insufficient effectiveness of the drugs used, which leads to a steady increase in the number of patients. The aim of the study was to evaluate the therapeutic efficacy of the eye drug films «GlazAvir» in the experimental model of acute herpetic eye infection in rabbits. RESEARCH OBJECTIVES to study the specific activity of «GlazAvir» and compare the long-term indicators of the level of manifestation of individual clinical signs of keratoconjunctivitis. MATERIAL AND METHODS In the work we used rabbits of the Chinchilla breed, the herpes simplex virus type 1 and the eye drug films «GlazAvir». A model of ophthalmic herpetic infection was formed by infection of rabbits with virus-containing material of a pre-scarified eye cornea against the background of local anesthesia. Аnimals were treated with the drug «GlazAvir» - 1 application per day for 7 days. Animals were observed daily for 15 days, then every 3 days until the 25th day of observation. The effectiveness of the drug was evaluated at the peak of the development of the pathological process. RESULTS AND DISCUSSION There was a decrease in mortality from 50 to 20%, and an increase in average life expectancy by 27.87%, compared with the control in animals treated with «GlazAvir». It was noted after activation of herpetic keratoconjunctivitis on the 2nd - 5th day, at the peak of the disease (6-9th day) a statistically significant decrease in the activity of the pathological process (p<0.05) by rabbits treated with the «GlazAvir» ophthalmic drug films. The tendency to normalization by the rabbits treated with the «GlazAvir» preparation was observed until the 14th day. CONCLUSION The data obtained indicate the pronounced effectiveness of the «GlazAvir» preparation in the treatment of experimental herpesvirus keratoconjunctivitis.
Collapse
Affiliation(s)
- A M Ivanova
- National Research Centre for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russia
| | | | - A N Narovlyansky
- National Research Centre for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russia
| | - A V Izmest'eva
- National Research Centre for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russia
| | - A A Sarymsakov
- Institute of Polymer Chemystry and Physics, Tashkent, 100128, Uzbekistan
| | - E N Bilalov
- Tashkent Medical Academy, Tashkent, 100109, Uzbekistan
| | - F I Ershov
- National Research Centre for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russia
| |
Collapse
|
16
|
Abstract
A large number of viruses belonging to various families are able to cause central nervous system (CNS) infections and contribute significantly to burden of disease in humans globally. Most viral CNS infections are benign and self-limiting, and most remain undiagnosed. However, some viruses can cause severe inflammation, leading to morbidity and mortality, and result in severe long-term residual damage and neurologic dysfunction in patients. The potential of viruses to cause CNS inflammation greatly varies depending on host factors, such as age, sex, and genetic background, as well as viral factors. Despite the need for protection against viral invasion and replication, the extent of the immune response in the CNS is carefully regulated to prevent excessive inflammation and tissue destruction leading to irretrievable loss of neurons. Direct cytopathology is for many virus infections a major cause of neurologic symptoms; however, the antiviral immune response can in some instances contribute substantially to pathology. This chapter highlights a selection of clinically important neurotropic viruses that infect the CNS and cause neurologic diseases such as meningitis, encephalitis, and myelitis in humans, with a focus on neuropathologic findings.
Collapse
Affiliation(s)
- Nikolaus Deigendesch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
17
|
Differentiated Human SH-SY5Y Cells Provide a Reductionist Model of Herpes Simplex Virus 1 Neurotropism. J Virol 2017; 91:JVI.00958-17. [PMID: 28956768 DOI: 10.1128/jvi.00958-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022] Open
Abstract
Neuron-virus interactions that occur during herpes simplex virus (HSV) infection are not fully understood. Neurons are the site of lifelong latency and are a crucial target for long-term suppressive therapy or viral clearance. A reproducible neuronal model of human origin would facilitate studies of HSV and other neurotropic viruses. Current neuronal models in the herpesvirus field vary widely and have caveats, including incomplete differentiation, nonhuman origins, or the use of dividing cells that have neuropotential but lack neuronal morphology. In this study, we used a robust approach to differentiate human SH-SY5Y neuroblastoma cells over 2.5 weeks, producing a uniform population of mature human neuronal cells. We demonstrate that terminally differentiated SH-SY5Y cells have neuronal morphology and express proteins with subcellular localization indicative of mature neurons. These neuronal cells are able to support a productive HSV-1 infection, with kinetics and overall titers similar to those seen in undifferentiated SH-SY5Y cells and the related SK-N-SH cell line. However, terminally differentiated, neuronal SH-SY5Y cells release significantly less extracellular HSV-1 by 24 h postinfection (hpi), suggesting a unique neuronal response to viral infection. With this model, we are able to distinguish differences in neuronal spread between two strains of HSV-1. We also show expression of the antiviral protein cyclic GMP-AMP synthase (cGAS) in neuronal SH-SY5Y cells, which is the first demonstration of the presence of this protein in nonepithelial cells. These data provide a model for studying neuron-virus interactions at the single-cell level as well as via bulk biochemistry and will be advantageous for the study of neurotropic viruses in vitroIMPORTANCE Herpes simplex virus (HSV) affects millions of people worldwide, causing painful oral and genital lesions, in addition to a multitude of more severe symptoms such as eye disease, neonatal infection, and, in rare cases, encephalitis. Presently, there is no cure available to treat those infected or prevent future transmission. Due to the ability of HSV to cause a persistent, lifelong infection in the peripheral nervous system, the virus remains within the host for life. To better understand the basis of virus-neuron interactions that allow HSV to persist within the host peripheral nervous system, improved neuronal models are required. Here we describe a cost-effective and scalable human neuronal model system that can be used to study many neurotropic viruses, such as HSV, Zika virus, dengue virus, and rabies virus.
Collapse
|
18
|
Varanasi SK, Donohoe D, Jaggi U, Rouse BT. Manipulating Glucose Metabolism during Different Stages of Viral Pathogenesis Can Have either Detrimental or Beneficial Effects. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1748-1761. [PMID: 28768727 PMCID: PMC5584583 DOI: 10.4049/jimmunol.1700472] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 12/16/2022]
Abstract
This report deals with physiological changes and their implication following ocular infection with HSV. This infection usually results in a blinding inflammatory reaction in the cornea, orchestrated mainly by proinflammatory CD4 T cells and constrained in severity by regulatory T cells. In the present report, we make the unexpected finding that blood glucose levels change significantly during the course of infection. Whereas levels remained normal during the early phase of infection when the virus was actively replicating in the cornea, they increased around 2-fold during the time when inflammatory responses to the virus was occurring. We could show that glucose levels influenced the extent of induction of the inflammatory T cell subset in vitro that mainly drives lesions, but not regulatory T cells. Additionally, if glucose utilization was limited in vivo as a consequence of therapy in the inflammatory phase with the drug 2-deoxy-glucose (2DG), lesions were diminished compared with untreated infected controls. In addition, lesions in 2DG-treated animals contained less proinflammatory effectors. Glucose metabolism also influenced the acute phase of infection when the replicating virus was present in the eye. Thus, therapy with 2DG to limit glucose utilization caused mice to become susceptible to the lethal effects of HSV infection, with the virus spreading to the brain causing encephalitis. Taken together, our results indicate that glucose metabolism changed during the course of HSV infection and that modulating glucose levels can influence the outcome of infection, being detrimental or beneficial according to the stage of viral pathogenesis.
Collapse
Affiliation(s)
- Siva Karthik Varanasi
- Department of Genome Science and Technology, College of Arts & Sciences, University of Tennessee, Knoxville, TN 37996
| | - Dallas Donohoe
- Department of Nutrition, College of Education, Health and Human Sciences, University of Tennessee, Knoxville, TN 37996; and
| | - Ujjaldeep Jaggi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996
| | - Barry T Rouse
- Department of Genome Science and Technology, College of Arts & Sciences, University of Tennessee, Knoxville, TN 37996;
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996
| |
Collapse
|
19
|
Abstract
The most common specimens from immunocompromised patients that are analyzed for detection of herpes simplex virus (HSV) or varicella-zoster virus (VZV) are from skin lesions. Many types of assays are applicable to these samples, but some, such as virus isolation and direct fluorescent antibody testing, are useful only in the early phases of the lesions. In contrast, nucleic acid (NA) detection methods, which generally have superior sensitivity and specificity, can be applied to skin lesions at any stage of progression. NA methods are also the best choice, and sometimes the only choice, for detecting HSV or VZV in blood, cerebrospinal fluid, aqueous or vitreous humor, and from mucosal surfaces. NA methods provide the best performance when reliability and speed (within 24 hours) are considered together. They readily distinguish the type of HSV detected or the source of VZV detected (wild type or vaccine strain). Nucleic acid detection methods are constantly being improved with respect to speed and ease of performance. Broader applications are under study, such as the use of quantitative results of viral load for prognosis and to assess the efficacy of antiviral therapy.
Collapse
|
20
|
Morinet F. TLR3 and skin features. Curr Res Transl Med 2016; 64:167. [PMID: 27939453 DOI: 10.1016/j.retram.2016.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- F Morinet
- Laboratoire de Virologie, Hôpital Saint-Louis, Université Paris 7 Denis-Diderot, AP-HP, Paris, France.
| |
Collapse
|
21
|
Von Willebrand Factor Gene Variants Associate with Herpes simplex Encephalitis. PLoS One 2016; 11:e0155832. [PMID: 27224245 PMCID: PMC4880288 DOI: 10.1371/journal.pone.0155832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/04/2016] [Indexed: 01/24/2023] Open
Abstract
Herpes simplex encephalitis (HSE) is a rare complication of Herpes simplex virus type-1 infection. It results in severe parenchymal damage in the brain. Although viral latency in neurons is very common in the population, it remains unclear why certain individuals develop HSE. Here we explore potential host genetic variants predisposing to HSE. In order to investigate this we used a rat HSE model comparing the HSE susceptible SHR (Spontaneously Hypertensive Rats) with the asymptomatic infection of BN (Brown Norway). Notably, both strains have HSV-1 spread to the CNS at four days after infection. A genome wide linkage analysis of 29 infected HXB/BXH RILs (recombinant inbred lines-generated from the prior two strains), displayed variable susceptibility to HSE enabling the definition of a significant QTL (quantitative trait locus) named Hse6 towards the end of chromosome 4 (160.89-174Mb) containing the Vwf (von Willebrand factor) gene. This was the only gene in the QTL with both cis-regulation in the brain and included several non-synonymous SNPs (single nucleotide polymorphism). Intriguingly, in human chromosome 12 several SNPs within the intronic region between exon 43 and 44 of the VWF gene were associated with human HSE pathogenesis. In particular, rs917859 is nominally associated with an odds ratio of 1.5 (95% CI 1.11-2.02; p-value = 0.008) after genotyping in 115 HSE cases and 428 controls. Although there are possibly several genetic and environmental factors involved in development of HSE, our study identifies variants of the VWF gene as candidates for susceptibility in experimental and human HSE.
Collapse
|
22
|
Innate immune interactions within the central nervous system modulate pathogenesis of viral infections. Curr Opin Immunol 2015; 36:47-53. [PMID: 26163762 DOI: 10.1016/j.coi.2015.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 11/23/2022]
Abstract
The innate immune system mediates protection against neurotropic viruses that replicate in the central nervous system (CNS). Virus infection within specific cells of the CNS triggers activation of several families of pattern recognition receptors including Toll-like receptors, retinoic acid-inducible gene I like receptors, nucleotide-binding oligomerization domain-like receptors, and cytosolic DNA sensors. In this review, we highlight recent advances in our understanding of how cell-intrinsic host defenses within the CNS modulate infection of different DNA and RNA viruses.
Collapse
|
23
|
Le-Trilling VTK, Trilling M. Attack, parry and riposte: molecular fencing between the innate immune system and human herpesviruses. ACTA ACUST UNITED AC 2015; 86:1-13. [DOI: 10.1111/tan.12594] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- V. T. K. Le-Trilling
- Institute for Virology; University Hospital Essen, University Duisburg-Essen; Essen Germany
| | - M. Trilling
- Institute for Virology; University Hospital Essen, University Duisburg-Essen; Essen Germany
| |
Collapse
|
24
|
Pleyer U, Chee SP. Current aspects on the management of viral uveitis in immunocompetent individuals. Clin Ophthalmol 2015; 9:1017-28. [PMID: 26089633 PMCID: PMC4467646 DOI: 10.2147/opth.s60394] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Viruses are a fundamental etiology of ocular inflammation, which may affect all structures of the organ. Advances in molecular diagnostics reveal an increasingly broader spectrum of virus-associated intraocular inflammation, including all members of the herpes family, rubella virus, and other more rare causes such as Epstein–Barr and chikungunya virus. In particular, viruses of the herpes family are important causes of anterior and posterior uveitis. Owing to their often fulminant clinical course and persistence in ocular tissues, a clear differential diagnosis between alpha- and beta-type herpes viruses is essential to guide acute and long-term treatment. Here, we review the epidemiology, clinical, and laboratory findings of virus-associated uveitis with emphasis on their therapy and management and include our own experience.
Collapse
Affiliation(s)
- Uwe Pleyer
- Augenklinik, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Soon-Phaik Chee
- Augenklinik, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany ; Ocular Inflammation and Immunology Service, Singapore National Eye Centre, Singapore ; Singapore Eye Research Institute, National University of Singapore, Singapore ; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore ; Duke-NUS Graduate Medical School Singapore, Singapore
| |
Collapse
|
25
|
Lafaille FG, Ciancanelli MJ, Studer L, Smith G, Notarangelo L, Casanova JL, Zhang SY. Deciphering Human Cell-Autonomous Anti-HSV-1 Immunity in the Central Nervous System. Front Immunol 2015; 6:208. [PMID: 26005444 PMCID: PMC4424875 DOI: 10.3389/fimmu.2015.00208] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/15/2015] [Indexed: 12/26/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) is a common virus that can rarely invade the human central nervous system (CNS), causing devastating encephalitis. The permissiveness to HSV-1 of the various relevant cell types of the CNS, neurons, astrocytes, oligodendrocytes, and microglia cells, as well as their response to viral infection, has been extensively studied in humans and other animals. Nevertheless, human CNS cell-based models of anti-HSV-1 immunity are of particular importance, as responses to any given neurotropic virus may differ between humans and other animals. Human CNS neuron cell lines as well as primary human CNS neurons, astrocytes, and microglia cells cultured/isolated from embryos or cadavers, have enabled the study of cell-autonomous anti-HSV-1 immunity in vitro. However, the paucity of biological samples and their lack of purity have hindered progress in the field, which furthermore suffers from the absence of testable primary human oligodendrocytes. Recently, the authors have established a human induced pluripotent stem cells (hiPSCs)-based model of anti-HSV-1 immunity in neurons, oligodendrocyte precursor cells, astrocytes, and neural stem cells, which has widened the scope of possible in vitro studies while permitting in-depth explorations. This mini-review summarizes the available data on human primary and iPSC-derived CNS cells for anti-HSV-1 immunity. The hiPSC-mediated study of anti-viral immunity in both healthy individuals and patients with viral encephalitis will be a powerful tool in dissecting the disease pathogenesis of CNS infections with HSV-1 and other neurotropic viruses.
Collapse
Affiliation(s)
- Fabien G Lafaille
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA
| | - Michael J Ciancanelli
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research , New York, NY , USA
| | - Gregory Smith
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine , Chicago, IL , USA
| | - Luigi Notarangelo
- Division of Immunology, Boston Children's Hospital and Harvard Medical School , Boston, MA , USA
| | - Jean-Laurent Casanova
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA ; Howard Hughes Medical Institute , New York, NY , USA ; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children , Paris , France ; Imagine Institute, Paris Descartes University , Paris , France ; Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children , Paris , France
| | - Shen-Ying Zhang
- Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University , New York, NY , USA ; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children , Paris , France ; Imagine Institute, Paris Descartes University , Paris , France
| |
Collapse
|
26
|
Increased Viral Dissemination in the Brain and Lethality in MCMV-Infected, Dicer-Deficient Neonates. Viruses 2015; 7:2308-20. [PMID: 25955106 PMCID: PMC4452907 DOI: 10.3390/v7052308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/23/2015] [Accepted: 04/30/2015] [Indexed: 12/15/2022] Open
Abstract
Among Herpesviruses, Human Cytomegalovirus (HCMV or HHV-5) represents a major threat during congenital or neonatal infections, which may lead to encephalitis with serious neurological consequences. However, as opposed to other less prevalent pathogens, the mechanisms and genetic susceptibility factors for CMV encephalitis are poorly understood. This lack of information considerably reduces the prognostic and/or therapeutic possibilities. To easily monitor the effects of genetic defects on brain dissemination following CMV infection we used a recently developed in vivo mouse model based on the neonatal inoculation of a MCMV genetically engineered to express Luciferase. Here, we further validate this protocol for live imaging, and demonstrate increased lethality associated with viral infection and encephalitis in mutant mice lacking Dicer activity. Our data indicate that miRNAs are important players in the control of MCMV pathogenesis and suggest that miRNA-based endothelial functions and integrity are crucial for CMV encephalitis.
Collapse
|
27
|
Cold sore susceptibility gene-1 genotypes affect the expression of herpes labialis in unrelated human subjects. Hum Genome Var 2014; 1:14024. [PMID: 27081513 PMCID: PMC4785534 DOI: 10.1038/hgv.2014.24] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 02/02/2023] Open
Abstract
Our group has recently described a gene on human chromosome 21, the Cold Sore Susceptibility Gene-1 (CSSG-1, also known as C21orf91), which may confer susceptibility to frequent cold sores in humans. We present here a genotype–phenotype analysis of CSSG-1 in a new, unrelated human population. Seven hundred fifty-eight human subjects were enrolled in a case/control Cold Sore Study. CSSG-1 genotyping, herpes simplex virus 1 (HSV1) serotyping, demographic and phenotypic data was available from 622 analyzed subjects. Six major alleles (H1–H6) were tested for associations with each of the self-reported phenotypes. The statistical analysis was adjusted for age, sex and ethnicity. Genotype–phenotype associations were analyzed from 388 HSV1-seropositive subjects. There were significant CSSG-1 haplotype effects on annual cold sore outbreaks (P=0.006), lifetime cold sores (P=0.012) and perceived cold sore severity (P=0.012). There were relatively consistent trends toward protection from frequent and severe cold sores among those with the H3 or H5/6 haplotypes, whereas those with H1, H2, and H4 haplotypes tended to have more frequent and more severe episodes. Different alleles of the newly described gene CSSG-1 affect the expression of cold sore phenotypes in this new, unrelated human population, confirming the findings of the previous family-based study.
Collapse
|
28
|
Mouse ENU Mutagenesis to Understand Immunity to Infection: Methods, Selected Examples, and Perspectives. Genes (Basel) 2014; 5:887-925. [PMID: 25268389 PMCID: PMC4276919 DOI: 10.3390/genes5040887] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 12/30/2022] Open
Abstract
Infectious diseases are responsible for over 25% of deaths globally, but many more individuals are exposed to deadly pathogens. The outcome of infection results from a set of diverse factors including pathogen virulence factors, the environment, and the genetic make-up of the host. The completion of the human reference genome sequence in 2004 along with technological advances have tremendously accelerated and renovated the tools to study the genetic etiology of infectious diseases in humans and its best characterized mammalian model, the mouse. Advancements in mouse genomic resources have accelerated genome-wide functional approaches, such as gene-driven and phenotype-driven mutagenesis, bringing to the fore the use of mouse models that reproduce accurately many aspects of the pathogenesis of human infectious diseases. Treatment with the mutagen N-ethyl-N-nitrosourea (ENU) has become the most popular phenotype-driven approach. Our team and others have employed mouse ENU mutagenesis to identify host genes that directly impact susceptibility to pathogens of global significance. In this review, we first describe the strategies and tools used in mouse genetics to understand immunity to infection with special emphasis on chemical mutagenesis of the mouse germ-line together with current strategies to efficiently identify functional mutations using next generation sequencing. Then, we highlight illustrative examples of genes, proteins, and cellular signatures that have been revealed by ENU screens and have been shown to be involved in susceptibility or resistance to infectious diseases caused by parasites, bacteria, and viruses.
Collapse
|
29
|
Mogensen TH. STAT3 and the Hyper-IgE syndrome: Clinical presentation, genetic origin, pathogenesis, novel findings and remaining uncertainties. JAKSTAT 2014; 2:e23435. [PMID: 24058807 PMCID: PMC3710320 DOI: 10.4161/jkst.23435] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 12/27/2012] [Accepted: 12/28/2012] [Indexed: 12/21/2022] Open
Abstract
During recent years a number of primary immunodeficiencies resulting from impaired function of JAK-STAT molecules have been described. One of these is the Hyper-IgE syndrome (HIES) characterized by elevated IgE levels, eczema, recurrent staphylococcal skin and pulmonary infections and pleiotropic somatic manifestations. In 2007 the genetic basis of HIES was revealed by identification of dominant negative STAT3 mutations in HIES patients. Subsequently impaired function of Tyk2 and DOCK8 have been implicated in milder forms of HIES. Since STAT3 acts as a central transcription factor downstream of multiple cytokine and growth factor receptors and thus regulates antimicrobial responses and cell survival, impaired STAT3 function results in immunodeficiency and in some cases tumorigenesis. However, as the immunological and molecular basis of HIES is being unraveled, important biological and immunological insight into JAK-STAT signaling is emerging that may have implications for our understanding of the pathogenesis and clinical management of patients with HIES.
Collapse
Affiliation(s)
- Trine H Mogensen
- Department of Infectious Diseases and International Centre for Immunodeficiency Diseases; Aarhus University Hospital Skejby; Aarhus, Denmark
| |
Collapse
|
30
|
Frans G, Meyts I, Picard C, Puel A, Zhang SY, Moens L, Wuyts G, Van der Werff Ten Bosch J, Casanova JL, Bossuyt X. Addressing diagnostic challenges in primary immunodeficiencies: Laboratory evaluation of Toll-like receptor- and NF-κB-mediated immune responses. Crit Rev Clin Lab Sci 2014; 51:112-23. [DOI: 10.3109/10408363.2014.881317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
31
|
Caignard G, Leiva-Torres GA, Leney-Greene M, Charbonneau B, Dumaine A, Fodil-Cornu N, Pyzik M, Cingolani P, Schwartzentruber J, Dupaul-Chicoine J, Guo H, Saleh M, Veillette A, Lathrop M, Blanchette M, Majewski J, Pearson A, Vidal SM. Genome-wide mouse mutagenesis reveals CD45-mediated T cell function as critical in protective immunity to HSV-1. PLoS Pathog 2013; 9:e1003637. [PMID: 24068938 PMCID: PMC3771889 DOI: 10.1371/journal.ppat.1003637] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/24/2013] [Indexed: 01/15/2023] Open
Abstract
Herpes simplex encephalitis (HSE) is a lethal neurological disease resulting from infection with Herpes Simplex Virus 1 (HSV-1). Loss-of-function mutations in the UNC93B1, TLR3, TRIF, TRAF3, and TBK1 genes have been associated with a human genetic predisposition to HSE, demonstrating the UNC93B-TLR3-type I IFN pathway as critical in protective immunity to HSV-1. However, the TLR3, UNC93B1, and TRIF mutations exhibit incomplete penetrance and represent only a minority of HSE cases, perhaps reflecting the effects of additional host genetic factors. In order to identify new host genes, proteins and signaling pathways involved in HSV-1 and HSE susceptibility, we have implemented the first genome-wide mutagenesis screen in an in vivo HSV-1 infectious model. One pedigree (named P43) segregated a susceptible trait with a fully penetrant phenotype. Genetic mapping and whole exome sequencing led to the identification of the causative nonsense mutation L3X in the Receptor-type tyrosine-protein phosphatase C gene (Ptprc(L3X)), which encodes for the tyrosine phosphatase CD45. Expression of MCP1, IL-6, MMP3, MMP8, and the ICP4 viral gene were significantly increased in the brain stems of infected Ptprc(L3X) mice accounting for hyper-inflammation and pathological damages caused by viral replication. Ptprc(L3X) mutation drastically affects the early stages of thymocytes development but also the final stage of B cell maturation. Transfer of total splenocytes from heterozygous littermates into Ptprc(L3X) mice resulted in a complete HSV-1 protective effect. Furthermore, T cells were the only cell population to fully restore resistance to HSV-1 in the mutants, an effect that required both the CD4⁺ and CD8⁺ T cells and could be attributed to function of CD4⁺ T helper 1 (Th1) cells in CD8⁺ T cell recruitment to the site of infection. Altogether, these results revealed the CD45-mediated T cell function as potentially critical for infection and viral spread to the brain, and also for subsequent HSE development.
Collapse
Affiliation(s)
- Grégory Caignard
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | | | - Michael Leney-Greene
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Benoit Charbonneau
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Anne Dumaine
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Nassima Fodil-Cornu
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Michal Pyzik
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
| | - Pablo Cingolani
- School of Computer Science and McGill Centre for Bioinformatics, McGill University, Montréal, Quebec, Canada
| | | | | | - Huaijian Guo
- Laboratory of Molecular Oncology, Clinical Research Institute of Montréal, Montréal, Quebec, Canada
| | - Maya Saleh
- Departments of Biochemistry and Medicine, McGill University, Montréal, Quebec, Canada
| | - André Veillette
- Laboratory of Molecular Oncology, Clinical Research Institute of Montréal, Montréal, Quebec, Canada
| | - Marc Lathrop
- McGill University and Genome Québec Innovation Centre, Montréal, Quebec, Canada
| | - Mathieu Blanchette
- School of Computer Science and McGill Centre for Bioinformatics, McGill University, Montréal, Quebec, Canada
| | - Jacek Majewski
- McGill University and Genome Québec Innovation Centre, Montréal, Quebec, Canada
| | - Angela Pearson
- INRS-Institut Armand-Frappier, Université du Québec, Laval, Quebec, Canada
| | - Silvia M. Vidal
- Departments of Human Genetics and Medicine, McGill University, Montréal, Quebec, Canada
- * E-mail:
| |
Collapse
|
32
|
Hafezi W, Hoerr V. In vivo visualization of encephalitic lesions in herpes simplex virus type 1 (HSV-1) infected mice by magnetic resonance imaging (MRI). Methods Mol Biol 2013; 1064:253-65. [PMID: 23996263 DOI: 10.1007/978-1-62703-601-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Herpes simplex encephalitis (HSE) is one of the most severe viral infections affecting the temporal lobes of the brain. Despite the improvements in diagnosis and antiviral drug treatment, one third of all patients fail to respond to therapy or subsequently suffer neurological relapse and develop long term neurological damage. Magnetic resonance imaging (MRI) is among the appropriate noninvasive tools for early diagnosis of viral central nervous system (CNS) infections. In this chapter we introduce a mouse model for HSE and describe a MRI protocol to characterize the pathogenesis of HSE over time.
Collapse
Affiliation(s)
- Wali Hafezi
- Institute of Medical Microbiology Clinical Virology, University Hospital Münster, Münster, Germany
| | | |
Collapse
|
33
|
Primary immunodeficiencies: a rapidly evolving story. J Allergy Clin Immunol 2013; 131:314-23. [PMID: 23374262 DOI: 10.1016/j.jaci.2012.11.051] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/06/2012] [Accepted: 11/29/2012] [Indexed: 12/28/2022]
Abstract
The characterization of primary immunodeficiencies (PIDs) in human subjects is crucial for a better understanding of the biology of the immune response. New achievements in this field have been possible in light of collaborative studies; attention paid to new phenotypes, infectious and otherwise; improved immunologic techniques; and use of exome sequencing technology. The International Union of Immunological Societies Expert Committee on PIDs recently reported on the updated classification of PIDs. However, new PIDs are being discovered at an ever-increasing rate. A series of 19 novel primary defects of immunity that have been discovered after release of the International Union of Immunological Societies report are discussed here. These new findings highlight the molecular pathways that are associated with clinical phenotypes and suggest potential therapies for affected patients.
Collapse
|
34
|
Murphy AA, Rosato PC, Parker ZM, Khalenkov A, Leib DA. Synergistic control of herpes simplex virus pathogenesis by IRF-3, and IRF-7 revealed through non-invasive bioluminescence imaging. Virology 2013; 444:71-9. [PMID: 23777662 DOI: 10.1016/j.virol.2013.05.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/20/2013] [Accepted: 05/22/2013] [Indexed: 12/13/2022]
Abstract
Interferon regulatory factors IRF-3 and IRF-7 are central to the establishment of the innate antiviral response. This study examines HSV-1 pathogenesis in IRF-3(-/-), IRF-7(-/-) and double-deleted IRF3/7(-/-) (DKO) mice. Bioluminescence imaging of infection revealed that DKO mice developed visceral infection following corneal inoculation, along with increased viral burdens in all tissues relative to single knockout mice. While all DKO mice synchronously reached endpoint criteria 5 days post infection, the IRF-7(-/-) mice survived longer, indicating that although IRF-7 is dominant, IRF-3 also plays a role in controlling disease. Higher levels of systemic pro-inflammatory cytokines were found in IRF7(-/-) and DKO mice relative to wild-type and IRF-3(-/-) mice, and IL-6 and G-CSF, indicative of sepsis, were increased in the DKO mice relative to wild-type or single-knockout mice. In addition to controlling viral replication, IRF-3 and -7 therefore play coordinating roles in modulation of inflammation during HSV infection.
Collapse
Affiliation(s)
- Aisling A Murphy
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Drive, HB 7556, Lebanon, NH 03756, USA
| | | | | | | | | |
Collapse
|
35
|
Casanova JL, Abel L. The genetic theory of infectious diseases: a brief history and selected illustrations. Annu Rev Genomics Hum Genet 2013; 14:215-43. [PMID: 23724903 PMCID: PMC4980761 DOI: 10.1146/annurev-genom-091212-153448] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Until the mid-nineteenth century, life expectancy at birth averaged 20 years worldwide, owing mostly to childhood fevers. The germ theory of diseases then gradually overcame the belief that diseases were intrinsic. However, around the turn of the twentieth century, asymptomatic infection was discovered to be much more common than clinical disease. Paradoxically, this observation barely challenged the newly developed notion that infectious diseases were fundamentally extrinsic. Moreover, interindividual variability in the course of infection was typically explained by the emerging immunological (or somatic) theory of infectious diseases, best illustrated by the impact of vaccination. This powerful explanation is, however, best applicable to reactivation and secondary infections, particularly in adults; it can less easily account for interindividual variability in the course of primary infection during childhood. Population and clinical geneticists soon proposed a complementary hypothesis, a germline genetic theory of infectious diseases. Over the past century, this idea has gained some support, particularly among clinicians and geneticists, but has also encountered resistance, particularly among microbiologists and immunologists. We present here the genetic theory of infectious diseases and briefly discuss its history and the challenges encountered during its emergence in the context of the apparently competing but actually complementary microbiological and immunological theories. We also illustrate its recent achievements by highlighting inborn errors of immunity underlying eight life-threatening infectious diseases of children and young adults. Finally, we consider the far-reaching biological and clinical implications of the ongoing human genetic dissection of severe infectious diseases.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065;
| | | |
Collapse
|
36
|
Rapid host immune response and viral dynamics in herpes simplex virus-2 infection. Nat Med 2013; 19:280-90. [PMID: 23467247 DOI: 10.1038/nm.3103] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 01/10/2013] [Indexed: 02/07/2023]
Abstract
Herpes simplex virus-2 (HSV-2) is periodically shed throughout the human genital tract. Although a high viral load correlates with the development of genital ulcers, shedding also commonly occurs even when ulcers are absent, allowing for silent transmission during coitus and contributing to high seroprevalence of HSV-2 worldwide. Frequent viral reactivation occurs within ganglia despite diverse and complementary host and viral mechanisms that predispose toward latency, suggesting that viral replication may be constantly occurring in a small minority of neurons at these sites. Within genital mucosa, the in vivo expansion and clearance rates of HSV-2 are extremely rapid. Resident dendritic cells and memory HSV-2 specific T cells persist at prior sites of genital tract reactivation and, in conjunction with prompt innate recognition of infected cells, lead to rapid containment of infected cells. The fact that immune responses usually control viral replication in genital skin before lesions develop provides hope that enhancing such responses could lead to effective vaccines and immunotherapies.
Collapse
|
37
|
Dreyfus DH. Herpesviruses and the microbiome. J Allergy Clin Immunol 2013; 132:1278-86. [PMID: 23611298 DOI: 10.1016/j.jaci.2013.02.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 02/07/2013] [Accepted: 02/25/2013] [Indexed: 12/15/2022]
Abstract
The focus of this article will be to examine the role of common herpesviruses as a component of the microbiome of atopic patients and to review clinical observations suggesting that atopic patients might be predisposed to more severe and atypical herpes-related illness because their immune response is biased toward a TH2 cytokine profile. Human populations are infected with 8 herpesviruses, including herpes simplex virus HSV1 and HSV2 (also termed HHV1 and HHV2), varicella zoster virus (VZV or HHV3), EBV (HHV4), cytomegalovirus (HHV5), HHV6, HHV7, and Kaposi sarcoma-associated herpesvirus (termed KSV or HHV8). Herpesviruses are highly adapted to lifelong infection of their human hosts and thus can be considered a component of the human "microbiome" in addition to their role in illness triggered by primary infection. HSV1 and HSV2 infection and reactivation can present with more severe cutaneous symptoms termed eczema herpeticum in the atopic population, similar to the more severe eczema vaccinatum, and drug reaction with eosinophilia and systemic symptoms syndrome (DRESS) is associated with reactivation of HSV6 and possibly other herpesviruses in both atopic and nonatopic patients. In this review evidence is reviewed that primary infection with herpesviruses may have an atypical presentation in the atopic patient and conversely that childhood infection might alter the atopic phenotype. Reactivation of latent herpesviruses can directly alter host cytokine profiles through viral expression of cytokine-like proteins, such as IL-10 (EBV) or IL-6 (cytomegalovirus and HHV8), viral encoded and secreted siRNA and microRNAs, and modulation of expression of host transcription pathways, such as nuclear factor κB. Physicians caring for allergic and atopic populations should be aware of common and uncommon presentations of herpes-related disease in atopic patients to provide accurate diagnosis and avoid unnecessary laboratory testing or incorrect diagnosis of other conditions, such as drug allergy or autoimmune disease. Antiviral therapy and vaccines should be administered promptly when indicated clinically.
Collapse
Affiliation(s)
- David H Dreyfus
- Department of Pediatrics, Clinical Faculty, Yale School of Medicine, New Haven, and the Center for Allergy, Asthma, and Immunology, Waterbury, Conn.
| |
Collapse
|
38
|
Abstract
Herpes simplex encephalitis (HSE) is a rare but severe complication of frequent and mostly benign infection with herpes simplex virus (HSV). Although rapid and sensitive diagnosis tools and active antiviral drugs are available, HSE morbidity/mortality levels remain unsatisfactory. Molecular and cellular determinants of HSE are incompletely understood. The rarity and severity of the disease have suggested an increased susceptibility of some subjects to HSV infection. Numerous experimental studies have investigated the respective role of host and viral factors in HSE. The results of these studies have illustrated the major role of the innate immune response, in particular interferons (IFNs), in limiting access of the virus into and/or virus replication in the central nervous system (CNS). In a few children with HSE, specific defects of the immune innate response have been identified, which impair the IFN-α/β and IFN-λ production of fibroblasts and/or neurons infected with HSV and render these cells more permissive to infection. The mutations affect proteins involved in the IFN pathway induced by stimulation of the TLR3 receptor. The patients' susceptibility to infection is restricted to HSV CNS invasion, underlining the major role of TLR3 in CNS protection against viral infection. The incomplete clinical penetrance of these molecular defects suggests that other factors (age, infectious dose) are involved in HSE. Whether pathogenesis of adult HSE is similar has not been investigated.
Collapse
Affiliation(s)
- F Rozenberg
- Service de virologie, pôle biologie pharmacie pathologie, hôpital Cochin, bâtiment Jean-Dausset, 27, rue du Faubourg-St-Jacques, 75679 Paris cedex 14, France.
| |
Collapse
|
39
|
|
40
|
Borysiewicz E, Doppalapudi S, Kirschman LT, Konat GW. TLR3 ligation protects human astrocytes against oxidative stress. J Neuroimmunol 2012; 255:54-9. [PMID: 23245579 DOI: 10.1016/j.jneuroim.2012.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/11/2012] [Accepted: 11/21/2012] [Indexed: 11/17/2022]
Abstract
Astrocytic Toll-like receptor 3 (TLR3) plays an important role not only in antiviral response but also in regeneration/healing of the CNS. The present study was undertaken to determine whether the neuroprotective effects of TLR3 signaling also include antioxidative protection. TLR3 ligation in human astrocytes induced protracted resistance of the cells to H(2)O(2) toxicity. Similar resistance was induced by conditioned medium from TLR3-ligated astrocytes indicating the involvement of paracrine signaling mechanisms. Out of 13 major antioxidative genes only the gene encoding superoxide dismutase 2 (SOD2) was postligationally upregulated suggesting that SOD2 is the major enzyme responsible for this protection.
Collapse
Affiliation(s)
- E Borysiewicz
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, 1 Medical Center Dr., Morgantown, WV 26506, USA
| | | | | | | |
Collapse
|
41
|
Sancho-Shimizu V, Perez de Diego R, Jouanguy E, Zhang SY, Casanova JL. Inborn errors of anti-viral interferon immunity in humans. Curr Opin Virol 2012; 1:487-96. [PMID: 22347990 DOI: 10.1016/j.coviro.2011.10.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The three types of interferon (IFNs) are essential for immunity against at least some viruses in the mouse model of experimental infections, type I IFNs displaying the broadest and strongest anti-viral activity. Consistently, human genetic studies have shown that type II IFN is largely redundant for immunity against viruses in the course of natural infections. The precise contributions of human type I and III IFNs remain undefined. However, various inborn errors of anti-viral IFN immunity have been described, which can result in either broad or narrow immunological and viral phenotypes. The broad disorders impair the response to (STAT1, TYK2) or the production of at least type I and type III IFNs following multiple stimuli (NEMO), resulting in multiple viral infections at various sites, including herpes simplex encephalitis (HSE). The narrow disorders impair exclusively (TLR3) or mostly (UNC-93B, TRIF, TRAF3) the TLR3-dependent induction of type I and III IFNs, leading to HSE in apparently otherwise healthy individuals. These recent discoveries highlight the importance of human type I and III IFNs in protective immunity against viruses, including the TLR3-IFN pathway in protection against HSE.
Collapse
Affiliation(s)
- Vanessa Sancho-Shimizu
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale, U980, Necker Medical School, Paris 75015, France
| | | | | | | | | |
Collapse
|
42
|
Abdelmagid N, Bereczky-Veress B, Guerreiro-Cacais AO, Bergman P, Luhr KM, Bergström T, Sköldenberg B, Piehl F, Olsson T, Diez M. The calcitonin receptor gene is a candidate for regulation of susceptibility to herpes simplex type 1 neuronal infection leading to encephalitis in rat. PLoS Pathog 2012; 8:e1002753. [PMID: 22761571 PMCID: PMC3386237 DOI: 10.1371/journal.ppat.1002753] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 05/01/2012] [Indexed: 11/24/2022] Open
Abstract
Herpes simplex encephalitis (HSE) is a fatal infection of the central nervous system (CNS) predominantly caused by Herpes simplex virus type 1. Factors regulating the susceptibility to HSE are still largely unknown. To identify host gene(s) regulating HSE susceptibility we performed a genome-wide linkage scan in an intercross between the susceptible DA and the resistant PVG rat. We found one major quantitative trait locus (QTL), Hse1, on rat chromosome 4 (confidence interval 24.3–31 Mb; LOD score 29.5) governing disease susceptibility. Fine mapping of Hse1 using recombinants, haplotype mapping and sequencing, as well as expression analysis of all genes in the interval identified the calcitonin receptor gene (Calcr) as the main candidate, which also is supported by functional studies. Thus, using unbiased genetic approach variability in Calcr was identified as potentially critical for infection and viral spread to the CNS and subsequent HSE development. Herpes simplex encephalitis (HSE) is a rare, but severe infection of the central nervous system (CNS) caused by Herpes simplex virus type 1. We have previously characterized a model for HSE in the inbred DA rat which resembles human HSE. Interestingly the inbred PVG rat is completely resistant to the disease and displays reduced or no uptake of viral particles into the peripheral and central nerve compartments respectively. To identify the gene(s) regulating HSE pathogenesis, we crossed the susceptible DA and the resistant PVG.A rats for two generations and infected 239 rats of the F2 (DAxPVG.A) cohort with HSV-1. A genome-wide linkage scan demonstrated one strong quantitative trait locus (QTL), Hse1, on rat chromosome 4 regulating disease susceptibility. Fine mapping, haplotype mapping, sequencing and expression analysis of the genes in the Hse1 interval collectively support the underlying genetic variation to be located in, or adjacent to the calcitonin receptor gene (Calcr). Further support for a role of CalcR in regulating HSV-1 replication and propagation is provided by strain-dependent differences in the calcitonin receptor protein tissue localization and in functional studies. Using an unbiased genetic mapping approach this study identifies Calcr as a candidate for regulating susceptibility to HSE.
Collapse
Affiliation(s)
- Nada Abdelmagid
- Department of Clinical Neuroscience, Neuroimmunology Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hafezi W, Lorentzen EU, Eing BR, Müller M, King NJC, Klupp B, Mettenleiter TC, Kühn JE. Entry of herpes simplex virus type 1 (HSV-1) into the distal axons of trigeminal neurons favors the onset of nonproductive, silent infection. PLoS Pathog 2012; 8:e1002679. [PMID: 22589716 PMCID: PMC3349744 DOI: 10.1371/journal.ppat.1002679] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 03/21/2012] [Indexed: 12/12/2022] Open
Abstract
Following productive, lytic infection in epithelia, herpes simplex virus type 1 (HSV-1) establishes a lifelong latent infection in sensory neurons that is interrupted by episodes of reactivation. In order to better understand what triggers this lytic/latent decision in neurons, we set up an organotypic model based on chicken embryonic trigeminal ganglia explants (TGEs) in a double chamber system. Adding HSV-1 to the ganglion compartment (GC) resulted in a productive infection in the explants. By contrast, selective application of the virus to distal axons led to a largely nonproductive infection that was characterized by the poor expression of lytic genes and the presence of high levels of the 2.0-kb major latency-associated transcript (LAT) RNA. Treatment of the explants with the immediate-early (IE) gene transcriptional inducer hexamethylene bisacetamide, and simultaneous co-infection of the GC with HSV-1, herpes simplex virus type 2 (HSV-2) or pseudorabies virus (PrV) helper virus significantly enhanced the ability of HSV-1 to productively infect sensory neurons upon axonal entry. Helper-virus-induced transactivation of HSV-1 IE gene expression in axonally-infected TGEs in the absence of de novo protein synthesis was dependent on the presence of functional tegument protein VP16 in HSV-1 helper virus particles. After the establishment of a LAT-positive silent infection in TGEs, HSV-1 was refractory to transactivation by superinfection of the GC with HSV-1 but not with HSV-2 and PrV helper virus. In conclusion, the site of entry appears to be a critical determinant in the lytic/latent decision in sensory neurons. HSV-1 entry into distal axons results in an insufficient transactivation of IE gene expression and favors the establishment of a nonproductive, silent infection in trigeminal neurons. Upon primary infection of the oronasal mucosa, herpes simplex virus type 1 (HSV-1) rapidly reaches the ganglia of the peripheral nervous system via axonal transport and establishes lifelong latency in surviving neurons. Central to the establishment of latency is the ability of HSV-1 to reliably switch from productive, lytic spread in epithelia to nonproductive, latent infection in sensory neurons. It is not fully understood what specifically disposes incoming particles of a highly cytopathogenic, fast-replicating alphaherpesvirus to nonproductive, latent infection in sensory neurons. The present study shows that selective entry of HSV-1 into the distal axons of trigeminal neurons strongly favors the establishment of a nonproductive, latent infection, whereas nonselective infection of neurons still enables HSV-1 to induce lytic gene expression. Our data support a model of latency establishment in which the site of entry is an important determinant of the lytic/latent decision in the infected neuron. Productive infection of the neuron ensues if particles enter the soma of the neuron directly. In contrast, previous retrograde axonal transport of incoming viral particles creates a distinct scenario that abrogates VP16-dependent transactivation of immediate-early gene expression and precludes the expression of lytic genes to an extent sufficient to prevent the initiation of massive productive infection of trigeminal neurons.
Collapse
Affiliation(s)
- Wali Hafezi
- University Hospital Münster, Institute of Medical Microbiology - Clinical Virology, Münster, Germany
- Interdisciplinary Center of Clinical Research (IZKF), Münster, Germany
| | - Eva U. Lorentzen
- University Hospital Münster, Institute of Medical Microbiology - Clinical Virology, Münster, Germany
| | - Bodo R. Eing
- University Hospital Münster, Institute of Medical Microbiology - Clinical Virology, Münster, Germany
| | - Marcus Müller
- University Hospital Bonn, Department of Neurology, Bonn, Germany
| | - Nicholas J. C. King
- University of Sydney, Sydney Medical School, Department of Pathology, Bosch Institute for Medical Research, New South Wales, Australia
| | - Barbara Klupp
- Friedrich-Loeffler-Institut, Institute of Molecular Biology, Greifswald-Insel Riems, Germany
| | - Thomas C. Mettenleiter
- Friedrich-Loeffler-Institut, Institute of Molecular Biology, Greifswald-Insel Riems, Germany
| | - Joachim E. Kühn
- University Hospital Münster, Institute of Medical Microbiology - Clinical Virology, Münster, Germany
- Interdisciplinary Center of Clinical Research (IZKF), Münster, Germany
- * E-mail:
| |
Collapse
|
44
|
Arkwright PD, Gennery AR. Ten warning signs of primary immunodeficiency: a new paradigm is needed for the 21st century. Ann N Y Acad Sci 2012; 1238:7-14. [PMID: 22129048 DOI: 10.1111/j.1749-6632.2011.06206.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The 10 warning signs of primary immunodeficiency are being promoted as a screening tool for use by both the general public and physicians. A recent study, however, shows that except for family history, need for intravenous antibiotics and failure to thrive, the 10 warning signs are not a useful screen of primary immunodeficiency diseases (PIDs). Over the last few decades, there has been a revolution in our understanding of PID. The 10 warning signs do not take into account the fact that PIDs now include diseases that present with sporadic infections, autoimmunity, autoinflammation, and malignancy. This review focuses on the advances in our understanding of PID, the current limitations of the 10 warning signs, and recommendations to ensure that patients with PID are diagnosed in a timely fashion in the future.
Collapse
Affiliation(s)
- Peter D Arkwright
- Department of Paediatric Allergy and Immunology, Royal Manchester Children's Hospital, University of Manchester, United Kingdom.
| | | |
Collapse
|
45
|
The C terminus of the large tegument protein pUL36 contains multiple capsid binding sites that function differently during assembly and cell entry of herpes simplex virus. J Virol 2012; 86:3682-700. [PMID: 22258258 DOI: 10.1128/jvi.06432-11] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The largest tegument protein of herpes simplex virus type 1 (HSV1), pUL36, is a multivalent cross-linker between the viral capsids and the tegument and associated membrane proteins during assembly that upon subsequent cell entry releases the incoming capsids from the outer tegument and viral envelope. Here we show that pUL36 was recruited to cytosolic progeny capsids that later colocalized with membrane proteins of herpes simplex virus type 1 (HSV1) and the trans-Golgi network. During cell entry, pUL36 dissociated from viral membrane proteins but remained associated with cytosolic capsids until arrival at the nucleus. HSV1 UL36 mutants lacking C-terminal portions of increasing size expressed truncated pUL36 but could not form plaques. Cytosolic capsids of mutants lacking the C-terminal 735 of the 3,164 amino acid residues accumulated in the cytosol but did not recruit pUL36 or associate with membranes. In contrast, pUL36 lacking only the 167 C-terminal residues bound to cytosolic capsids and subsequently colocalized with viral and host membrane proteins. Progeny virions fused with neighboring cells, but incoming capsids did not retain pUL36, nor could they target the nucleus or initiate HSV1 gene expression. Our data suggest that residues 2430 to 2893 of HSV1 pUL36, containing one binding site for the capsid protein pUL25, are sufficient to recruit pUL36 onto cytosolic capsids during assembly for secondary envelopment, whereas the 167 residues of the very C terminus with the second pUL25 binding site are crucial to maintain pUL36 on incoming capsids during cell entry. Capsids lacking pUL36 are targeted neither to membranes for virus assembly nor to nuclear pores for genome uncoating.
Collapse
|
46
|
Wang Y, Swiecki M, McCartney SA, Colonna M. dsRNA sensors and plasmacytoid dendritic cells in host defense and autoimmunity. Immunol Rev 2011; 243:74-90. [PMID: 21884168 DOI: 10.1111/j.1600-065x.2011.01049.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The innate immune system detects viruses through molecular sensors that trigger the production of type I interferons (IFN-I) and inflammatory cytokines. As viruses vary tremendously in size, structure, genomic composition, and tissue tropism, multiple sensors are required to detect their presence in various cell types and tissues. In this review, we summarize current knowledge of the diversity, specificity, and signaling pathways downstream of viral sensors and ask whether two distinct sensors that recognize the same viral component are complementary, compensatory, or simply redundant. We also discuss why viral sensors are differentially distributed in distinct cell types and whether a particular cell type dominates the IFN-I response during viral infection. Finally, we review evidence suggesting that inappropriate signaling through viral sensors may induce autoimmunity. The picture emerging from these studies is that disparate viral sensors in different cell types form a dynamic and integrated molecular network that can be exploited for improving vaccination and therapeutic strategies for infectious and autoimmune diseases.
Collapse
Affiliation(s)
- Yaming Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
47
|
|
48
|
Kriesel JD, Jones BB, Matsunami N, Patel MK, St Pierre CA, Kurt-Jones EA, Finberg RW, Leppert M, Hobbs MR. C21orf91 genotypes correlate with herpes simplex labialis (cold sore) frequency: description of a cold sore susceptibility gene. J Infect Dis 2011; 204:1654-62. [PMID: 22039568 PMCID: PMC3203230 DOI: 10.1093/infdis/jir633] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 01/21/2011] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Herpes simplex virus type 1 (HSV-1) infects >70% of the United States population. We identified a 3-megabase region on human chromosome 21 containing 6 candidate genes associated with herpes simplex labialis (HSL, "cold sores"). METHODS We conducted single nucleotide polymorphism (SNP) scans of the chromosome 21 region to define which of 6 possible candidate genes were associated with cold sore frequency. We obtained the annual HSL frequency for 355 HSV-1 seropositive individuals and determined the individual genotypes by SNPlex for linkage analysis and parental transmission disequilibrium testing (ParenTDT). RESULTS Two-point linkage analysis showed positive linkage between cold sore frequency and 2 SNPs within the C21orf91 region, 1 of which is nonsynonymous. ParenTDT analysis revealed a strong association between another C21orf91 SNP, predicted to lie in the 3' untranslated region, and frequent HSL (P = .0047). C21orf 91 is a predicted open reading frame of unknown function that encodes a cytosolic protein. CONCLUSIONS We evaluated candidate genes in the cold sore susceptibility region using fine mapping with 45 SNP markers. 2 complementary techniques identified C21orf91 as a gene of interest for susceptibility to HSL. We propose that C21orf91 be designated the Cold Sore Susceptibility Gene-1 (CSSG1).
Collapse
Affiliation(s)
- John D Kriesel
- Division of Infectious Diseases, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, 84132, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Type I interferons: diversity of sources, production pathways and effects on immune responses. Curr Opin Virol 2011; 1:463-75. [PMID: 22440910 DOI: 10.1016/j.coviro.2011.10.026] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 10/24/2011] [Accepted: 10/27/2011] [Indexed: 12/24/2022]
Abstract
Type I interferons (IFN-I) were first described over 50 years ago as factors produced by cells that interfere with virus replication and promote an antiviral state. Innate and adaptive immune responses to viruses are also greatly influenced by IFN-I. In this article we discuss the diversity of cellular sources of IFN-I and the pathways leading to IFN-I production during viral infections. Finally, we discuss the effects of IFN-I on cells of the immune system with emphasis on dendritic cells.
Collapse
|
50
|
Sancho-Shimizu V, Pérez de Diego R, Lorenzo L, Halwani R, Alangari A, Israelsson E, Fabrega S, Cardon A, Maluenda J, Tatematsu M, Mahvelati F, Herman M, Ciancanelli M, Guo Y, AlSum Z, Alkhamis N, Al-Makadma AS, Ghadiri A, Boucherit S, Plancoulaine S, Picard C, Rozenberg F, Tardieu M, Lebon P, Jouanguy E, Rezaei N, Seya T, Matsumoto M, Chaussabel D, Puel A, Zhang SY, Abel L, Al-Muhsen S, Casanova JL. Herpes simplex encephalitis in children with autosomal recessive and dominant TRIF deficiency. J Clin Invest 2011; 121:4889-902. [PMID: 22105173 DOI: 10.1172/jci59259] [Citation(s) in RCA: 222] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 10/06/2011] [Indexed: 12/13/2022] Open
Abstract
Herpes simplex encephalitis (HSE) is the most common sporadic viral encephalitis of childhood. Autosomal recessive (AR) UNC-93B and TLR3 deficiencies and autosomal dominant (AD) TLR3 and TRAF3 deficiencies underlie HSE in some children. We report here unrelated HSE children with AR or AD TRIF deficiency. The AR form of the disease was found to be due to a homozygous nonsense mutation that resulted in a complete absence of the TRIF protein. Both the TLR3- and the TRIF-dependent TLR4 signaling pathways were abolished. The AD form of disease was found to be due to a heterozygous missense mutation, resulting in a dysfunctional protein. In this form of the disease, the TLR3 signaling pathway was impaired, whereas the TRIF-dependent TLR4 pathway was unaffected. Both patients, however, showed reduced capacity to respond to stimulation of the DExD/H-box helicases pathway. To date, the TRIF-deficient patients with HSE described herein have suffered from no other infections. Moreover, as observed in patients with other genetic etiologies of HSE, clinical penetrance was found to be incomplete, as some HSV-1-infected TRIF-deficient relatives have not developed HSE. Our results provide what we believe to be the first description of human TRIF deficiency and a new genetic etiology for HSE. They suggest that the TRIF-dependent TLR4 and DExD/H-box helicase pathways are largely redundant in host defense. They further demonstrate the importance of TRIF for the TLR3-dependent production of antiviral IFNs in the CNS during primary infection with HSV-1 in childhood.
Collapse
Affiliation(s)
- Vanessa Sancho-Shimizu
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale, Necker Medical School, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|