1
|
Ramachandran PS, Wilson MR, Catho G, Blanchard-Rohner G, Schiess N, Cohrs RJ, Boutolleau D, Burrel S, Yoshikawa T, Wapniarski A, Heusel EH, Carpenter JE, Jackson W, Ford BA, Grose C. Meningitis Caused by the Live Varicella Vaccine Virus: Metagenomic Next Generation Sequencing, Immunology Exome Sequencing and Cytokine Multiplex Profiling. Viruses 2021; 13:2286. [PMID: 34835092 PMCID: PMC8620440 DOI: 10.3390/v13112286] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/21/2022] Open
Abstract
Varicella vaccine meningitis is an uncommon delayed adverse event of vaccination. Varicella vaccine meningitis has been diagnosed in 12 children, of whom 3 were immunocompromised. We now report two additional cases of vaccine meningitis in twice-immunized immunocompetent children and we perform further testing on a prior third case. We used three methods to diagnose or investigate cases of varicella vaccine meningitis, none of which have been used previously on this disease. These include metagenomic next-generation sequencing and cytokine multiplex profiling of cerebrospinal fluid and immunology exome analysis of white blood cells. In one new case, the diagnosis was confirmed by metagenomic next-generation sequencing of cerebrospinal fluid. Both varicella vaccine virus and human herpesvirus 7 DNA were detected. We performed cytokine multiplex profiling on the cerebrospinal fluid of two cases and found ten elevated biomarkers: interferon gamma, interleukins IL-1RA, IL-6, IL-8, IL-10, IL-17F, chemokines CXCL-9, CXCL-10, CCL-2, and G-CSF. In a second new case, we performed immunology exome sequencing on a panel of 356 genes, but no errors were found. After a review of all 14 cases, we concluded that (i) there is no common explanation for this adverse event, but (ii) ingestion of an oral corticosteroid burst 3-4 weeks before onset of vaccine meningitis may be a risk factor in some cases.
Collapse
Affiliation(s)
- Prashanth S. Ramachandran
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94110, USA; (P.S.R.); (M.R.W.); (A.W.)
| | - Michael R. Wilson
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94110, USA; (P.S.R.); (M.R.W.); (A.W.)
| | - Gaud Catho
- Division of Pediatric Infectious Diseases, Geneva University Hospitals, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland;
| | - Geraldine Blanchard-Rohner
- Pediatric Immunology and Vaccinology Unit, Division of General Pediatrics, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, University of Geneva, 1205 Geneva, Switzerland;
| | - Nicoline Schiess
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| | - Randall J. Cohrs
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - David Boutolleau
- Virology Department, National Reference Center for Herpesviruses, Pitie-Salpetriere Hospital, Sorbonne University, 75013 Paris, France; (D.B.); (S.B.)
| | - Sonia Burrel
- Virology Department, National Reference Center for Herpesviruses, Pitie-Salpetriere Hospital, Sorbonne University, 75013 Paris, France; (D.B.); (S.B.)
| | - Tetsushi Yoshikawa
- Department of Pediatrics, Fujita Health University School of Medicine, Aichi, Toyoake 470-1192, Japan;
| | - Anne Wapniarski
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94110, USA; (P.S.R.); (M.R.W.); (A.W.)
| | - Ethan H. Heusel
- Division of Infectious Diseases/Virology, Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA; (E.H.H.); (J.E.C.); (W.J.)
| | - John E. Carpenter
- Division of Infectious Diseases/Virology, Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA; (E.H.H.); (J.E.C.); (W.J.)
| | - Wallen Jackson
- Division of Infectious Diseases/Virology, Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA; (E.H.H.); (J.E.C.); (W.J.)
| | - Bradley A. Ford
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA;
| | - Charles Grose
- Division of Infectious Diseases/Virology, Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA; (E.H.H.); (J.E.C.); (W.J.)
| |
Collapse
|
2
|
Kennedy PGE, Mogensen TH, Cohrs RJ. Recent Issues in Varicella-Zoster Virus Latency. Viruses 2021; 13:v13102018. [PMID: 34696448 PMCID: PMC8540691 DOI: 10.3390/v13102018] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 12/16/2022] Open
Abstract
Varicella-zoster virus (VZV) is a human herpes virus which causes varicella (chicken pox) as a primary infection, and, following a variable period of latency in neurons in the peripheral ganglia, may reactivate to cause herpes zoster (shingles) as well as a variety of neurological syndromes. In this overview we consider some recent issues in alphaherpesvirus latency with special focus on VZV ganglionic latency. A key question is the nature and extent of viral gene transcription during viral latency. While it is known that this is highly restricted, it is only recently that the very high degree of that restriction has been clarified, with both VZV gene 63-encoded transcripts and discovery of a novel VZV transcript (VLT) that maps antisense to the viral transactivator gene 61. It has also emerged in recent years that there is significant epigenetic regulation of VZV gene transcription, and the mechanisms underlying this are complex and being unraveled. The last few years has also seen an increased interest in the immunological aspects of VZV latency and reactivation, in particular from the perspective of inborn errors of host immunity that predispose to different VZV reactivation syndromes.
Collapse
Affiliation(s)
- Peter G. E. Kennedy
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow G61 1QH, UK
- Correspondence:
| | - Trine H. Mogensen
- Department of Infectious Diseases, Aarhus University Hospital, 8000 Aarhus, Denmark;
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Randall J. Cohrs
- Department of Neurology, University of Colorado School of Medicine, 80045 Aurora, CO, USA
| |
Collapse
|
3
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
4
|
Sutherland JP, Steain M, Buckland ME, Rodriguez M, Cunningham AL, Slobedman B, Abendroth A. Persistence of a T Cell Infiltrate in Human Ganglia Years After Herpes Zoster and During Post-herpetic Neuralgia. Front Microbiol 2019; 10:2117. [PMID: 31572325 PMCID: PMC6749866 DOI: 10.3389/fmicb.2019.02117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/28/2019] [Indexed: 12/14/2022] Open
Abstract
Varicella-zoster virus (VZV) is a human herpesvirus which causes varicella (chicken pox) during primary infection, establishes latency in sensory ganglia, and can reactivate from this site to cause herpes zoster (HZ) (shingles). A major complication of HZ is a severe and often debilitating pain called post-herpetic neuralgia (PHN) which persists long after the resolution of the HZ-associated rash. The underlying cause of PHN is not known, although it has been postulated that it may be a consequence of immune cell mediated damage. However, the nature of virus-immune cell interactions within ganglia during PHN is unknown. We obtained rare formalin fixed paraffin embedded sections cut from surgically excised ganglia from a PHN-affected patient years following HZ rash resolution. VZV DNA was readily detected by qPCR and regions of immune infiltration were detected by hematoxylin and eosin staining. Immunostaining using a range of antibodies against immune cell subsets revealed an immune cell response comprising of CD4+ and CD8+ T cells and CD20+ B cells. This study explores the immune cell repertoire present in ganglia during PHN and provides evidence for an ongoing immune cell inflammation years after HZ.
Collapse
Affiliation(s)
- Jeremy P Sutherland
- Emergency Department, Westmead Hospital, The University of Sydney, Sydney, NSW, Australia
| | - Megan Steain
- Discipline of Infectious Diseases and Immunology, The University of Sydney, Sydney, NSW, Australia
| | - Michael E Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, The University of Sydney, Sydney, NSW, Australia
| | - Michael Rodriguez
- Department of Pathology, The University of Sydney, Sydney, NSW, Australia
| | - Anthony L Cunningham
- Centre for Virus Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Barry Slobedman
- Discipline of Infectious Diseases and Immunology, The University of Sydney, Sydney, NSW, Australia
| | - Allison Abendroth
- Discipline of Infectious Diseases and Immunology, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Abstract
Varicella zoster virus (VZV) is a ubiquitous, exclusively human alphaherpesvirus. Primary infection usually results in varicella (chickenpox), after which VZV becomes latent in ganglionic neurons along the entire neuraxis. As VZV-specific cell-mediated immunity declines in elderly and immunocompromised individuals, VZV reactivates and causes herpes zoster (shingles), frequently complicated by postherpetic neuralgia. VZV reactivation also produces multiple serious neurological and ocular diseases, such as cranial nerve palsies, meningoencephalitis, myelopathy, and VZV vasculopathy, including giant cell arteritis, with or without associated rash. Herein, we review the clinical, laboratory, imaging, and pathological features of neurological complications of VZV reactivation as well as diagnostic tests to verify VZV infection of the nervous system. Updates on the physical state of VZV DNA and viral gene expression in latently infected ganglia, neuronal, and primate models to study varicella pathogenesis and immunity are presented along with innovations in the immunization of elderly individuals to prevent VZV reactivation.
Collapse
Affiliation(s)
- Don Gilden
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, 12700, USA; Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 12800, USA
| | - Maria Nagel
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, 12700, USA
| | - Randall Cohrs
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, 12700, USA; Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 12800, USA
| | - Ravi Mahalingam
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, 12700, USA
| | - Nicholas Baird
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, 12700, USA
| |
Collapse
|
6
|
Abstract
Among the human herpes viruses, three are neurotropic and capable of producing severe neurological abnormalities: herpes simplex virus type 1 and 2 (HSV-1 and HSV-2) and varicella-zoster virus (VZV). Both the acute, primary infection and the reactivation from the site of latent infection, the dorsal sensory ganglia, are associated with severe human morbidity and mortality. The peripheral nervous system is one of the major loci affected by these viruses. The present review details the virology and molecular biology underlying the human infection. This is followed by detailed description of the symtomatology, clinical presentation, diagnosis, course, therapy, and prognosis of disorders of the peripheral nervous system caused by these viruses.
Collapse
Affiliation(s)
- Israel Steiner
- Department of Neurology, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel.
| |
Collapse
|
7
|
Abreu Velez AM, Smoller BR, Gao W, Grossniklaus HE, Jiao Z, Arias LF, Dudley SC, Howard MS. Varicella-zoster virus (VZV) and alpha 1 antitrypsin: a fatal outcome in a patient affected by endemic pemphigus foliaceus. Int J Dermatol 2012; 51:809-16. [PMID: 22715825 DOI: 10.1111/j.1365-4632.2011.05296.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Herpes virus infections are well known infectious complications of pemphigus and bullous pemphigoid. We describe pathologic findings utilizing autopsy tissue from several organs from a patient affected by a new variant of endemic pemphigus in El Bagre, Colombia, South America. CASE REPORT We describe a patient by a new variant of endemic pemphigus foliaceus from El Bagre that was receiving high-dosage immunosuppressants when hospitalized and died suddenly following contact with a second patient affected by chicken pox. MATERIALS AND METHODS We performed studies utilizing hematoxylin and eosin, immunohistochemistry, and direct immunofluorescence techniques on tissues from several organs. RESULTS We detected the presence of varicella zoster virus, as well as strong positivity for α-1 antitrypsin in the heart, kidneys, spleen, liver, skin, brain, lungs, pancreas, small and large intestines, and skeletal muscle. In regard to structural damage in the kidney and heart, we believe the observed damage is associated with the presence of autoantibodies to these organs, since both of them are rich in plakins and El Bagre-EPF patients present significant antibodies to plakin molecules. CONCLUSION In patients with endemic pemphigus foliaceus, we recommend complete isolation of the patient when receiving high dosages of systemic immunosuppressive agents. We further suggest the clinical possibility of a synergistic, fatal interaction between active pemphigus foliaceus, varicella zoster virus, herpes simplex virus, immunosuppressive agents, and a systemic activation of α-1 antitrypsin. Thus, we suggest adequate bed spacing, barrier nursing, and preventative testing for α-1 antitrypsin activation are warranted in these patients to address these complications.
Collapse
|
8
|
Gilden D, Mahalingam R, Nagel MA, Pugazhenthi S, Cohrs RJ. Review: The neurobiology of varicella zoster virus infection. Neuropathol Appl Neurobiol 2011; 37:441-63. [PMID: 21342215 PMCID: PMC3176736 DOI: 10.1111/j.1365-2990.2011.01167.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Varicella zoster virus (VZV) is a neurotropic herpesvirus that infects nearly all humans. Primary infection usually causes chickenpox (varicella), after which virus becomes latent in cranial nerve ganglia, dorsal root ganglia and autonomic ganglia along the entire neuraxis. Although VZV cannot be isolated from human ganglia, nucleic acid hybridization and, later, polymerase chain reaction proved that VZV is latent in ganglia. Declining VZV-specific host immunity decades after primary infection allows virus to reactivate spontaneously, resulting in shingles (zoster) characterized by pain and rash restricted to one to three dermatomes. Multiple other serious neurological and ocular disorders also result from VZV reactivation. This review summarizes the current state of knowledge of the clinical and pathological complications of neurological and ocular disease produced by VZV reactivation, molecular aspects of VZV latency, VZV virology and VZV-specific immunity, the role of apoptosis in VZV-induced cell death and the development of an animal model provided by simian varicella virus infection of monkeys.
Collapse
Affiliation(s)
- D Gilden
- Department of Neurology, University of Colorado School of Medicine, USA.
| | | | | | | | | |
Collapse
|
9
|
Abstract
Primary infection by varicella zoster virus (VZV) typically results in childhood chickenpox, at which time latency is established in the neurons of the cranial nerve, dorsal root and autonomic ganglia along the entire neuraxis. During latency, the histone-associated virus genome assumes a circular episomal configuration from which transcription is epigenetically regulated. The lack of an animal model in which VZV latency and reactivation can be studied, along with the difficulty in obtaining high-titer cell-free virus, has limited much of our understanding of VZV latency to descriptive studies of ganglia removed at autopsy and analogy to HSV-1, the prototype alphaherpesvirus. However, the lack of miRNA, detectable latency-associated transcript and T-cell surveillance during VZV latency highlight basic differences between the two neurotropic herpesviruses. This article focuses on VZV latency: establishment, maintenance and reactivation. Comparisons are made with HSV-1, with specific attention to differences that make these viruses unique human pathogens.
Collapse
Affiliation(s)
| | - Aamir Shahzad
- Department for Biomolecular Structural Chemistry Max F. Perutz Laboratories, University of Vienna, Austria
| | - Randall J Cohrs
- Author for correspondence: University of Colorado Denver Medical School, Aurora, CO, USA, Tel.: +1 303 742 4325
| |
Collapse
|
10
|
Mahalingam R, Traina-Dorge V, Wellish M, Deharo E, Singletary ML, Ribka EP, Sanford R, Gilden D. Latent simian varicella virus reactivates in monkeys treated with tacrolimus with or without exposure to irradiation. J Neurovirol 2011; 16:342-54. [PMID: 20822371 DOI: 10.3109/13550284.2010.513031] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Simian varicella virus (SVV) infection of primates resembles human varicella-zoster virus (VZV) infection. After primary infection, SVV becomes latent in ganglia and reactivates after immunosuppression or social and environmental stress. Herein, natural SVV infection was established in 5 cynomolgus macaques (cynos) and 10 African green (AG) monkeys. Four cynos were treated with the immunosuppressant tacrolimus (80 to 300 μg/kg/day) for 4 months and 1 was untreated (group 1). Four AG monkeys were exposed to a single dose (200 cGy) of x-irradiation (group 2), and 4 other AG monkeys were irradiated and treated with tacrolimus for 4 months (group 3); the remaining 2 AG monkeys were untreated. Zoster rash developed 1 to 2 weeks after tacrolimus treatment in 3 of 4 monkeys in group 1, 6 weeks after irradiation in 1 of 4 monkeys in group 2, and 1 to 2 weeks after irradiation in all 4 monkeys in group 3. All monkeys were euthanized 1 to 4 months after immunosuppression. SVV antigens were detected immunohistochemically in skin biopsies as well as in lungs of most monkeys. Low copy number SVV DNA was detected in ganglia from all three groups of monkeys, including controls. RNA specific for SVV ORFs 61, 63, and 9 was detected in ganglia from one immunosuppressed monkey in group 1. SVV antigens were detected in multiple ganglia from all immunosuppressed monkeys in every group, but not in controls. These results indicate that tacrolimus treatment produced reactivation in more monkeys than irradiation and tacrolimus and irradiation increased the frequency of SVV reactivation as compared to either treatment alone.
Collapse
Affiliation(s)
- Ravi Mahalingam
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Varicella-zoster virus neurotropism in SCID mouse-human dorsal root ganglia xenografts. Curr Top Microbiol Immunol 2010; 342:255-76. [PMID: 20225014 DOI: 10.1007/82_2009_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Varicella-zoster virus (VZV) is a neurotropic human alphaherpesvirus and the causative agent of varicella and herpes zoster. VZV reactivation from latency in sensory nerve ganglia is a direct consequence of VZV neurotropism. Investigation of VZV neuropathogenesis by infection of human dorsal root ganglion xenografts in immunocompromised (SCID) mice has provided a novel system in which to examine VZV neurotropism. Experimental infection with recombinant VZV mutants with targeted deletions or mutations of specific genes or regulatory elements provides an opportunity to assess gene candidates that may mediate neurotropism and neurovirulence. The SCID mouse-human DRG xenograft model may aid in the development of clinical strategies in the management of herpes zoster as well as in the development of "second generation" neuroattenuated vaccines.
Collapse
|
12
|
Abstract
Varicella zoster virus (VZV) infection results in the establishment of latency in human sensory neurons. Reactivation of VZV leads to herpes zoster which can be followed by persistent neuropathic pain, termed post-herpetic neuralgia (PHN). Humans are the only natural host for VZV, and the strict species specificity of the virus has restricted the development of an animal model of infection which mimics all phases of disease. In order to elucidate the mechanisms which control the establishment of latency and reactivation as well as the effect of VZV replication on neuronal function, in vitro models of neuronal infection have been developed. Currently these models involve culturing and infecting dissociated human fetal neurons, with or without their supporting cells, an intact explant fetal dorsal root ganglia (DRG) model, neuroblastoma cell lines and rodent neuronal cell models. Each of these models has distinct advantages as well as disadvantages, and all have contributed towards our understanding of VZV neuronal infection. However, as yet none have been able to recapitulate the full virus lifecycle from primary infection to latency through to reactivation. The development of such a model will be a crucial step towards advancing our understanding of the mechanisms involved in VZV replication in neuronal cells, and the design of new therapies to combat VZV-related disease.
Collapse
|
13
|
Kennedy PGE, Cohrs RJ. Varicella-zoster virus human ganglionic latency: a current summary. J Neurovirol 2010; 16:411-8. [PMID: 20874010 DOI: 10.1007/bf03210846] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Varicella-zoster virus (VZV) is a ubiquitous human herpes virus typically acquired in childhood when it causes varicella (chickenpox), following which the virus establishes a latent infection in trigeminal and dorsal root ganglia that lasts for the life of the individual. VZV subsequently reactivates, spontaneously or after specific triggering factors, to cause herpes zoster (shingles), which may be complicated by postherpetic neuralgia and several other neurological complications including vasculopathy. Our understanding of VZV latency lags behind our knowledge of herpes simplex virus type 1 (HSV-1) latency primarily due to the difficulty in propagating the virus to high titers in a cell-free state, and the lack of a suitable small-animal model for studying virus latency and reactivation. It is now established beyond doubt that latent VZV is predominantly located in human ganglionic neurons. Virus gene transcription during latency is epigenetically regulated, and appears to be restricted to expression of at least six genes, with expression of gene 63 being the hallmark of latency. However, viral gene transcription may be more extensive than previously thought. There is also evidence for several VZV genes being expressed at the protein level, including VZV gene 63-encoded protein, but recent evidence suggests that this may not be a common event. The nature and extent of the chronic inflammatory response in latently infected ganglia is also of current interest. There remain several questions concerning the VZV latency process that still need to be resolved unambiguously and it is likely that this will require the use of newly developed molecular technologies, such as GeXPS multiplex polymerase chain reaction (PCR) for virus transcriptional analysis and ChIP-seq to study the epigenetic of latent virus genome ( Liu et al, 2010 , BMC Biol 8: 56).
Collapse
Affiliation(s)
- Peter G E Kennedy
- Department of Neurology, Glasgow University, Southern General Hospital, Glasgow, Scotland, UK.
| | | |
Collapse
|
14
|
Nagel M, Mahalingam R, Cohrs R, Gilden D. Virus vasculopathy and stroke: an under-recognized cause and treatment target. Infect Disord Drug Targets 2010; 10:105-111. [PMID: 20166970 PMCID: PMC2909030 DOI: 10.2174/187152610790963537] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 10/07/2009] [Indexed: 05/28/2023]
Abstract
While arteriosclerotic disease and hypertension, with or without diabetes, are the most common causes of stroke, viruses may also produce transient ischemic attacks and stroke. The three most-well studied viruses in this respect are varicella zoster virus (VZV), cytomegalovirus (CMV) and human immunodeficiency virus (HIV), all of which are potentially treatable with antiviral agents. Productive VZV infection in cerebral arteries after reactivation (zoster) or primary infection (varicella) has been documented as a cause of ischemic and hemorrhagic stroke, aneurysms with subarachnoid and intracerebral hemorrhage, arterial ectasia and as a co-factor in cerebral arterial dissection. CMV has been suggested to play a role in the pathogenesis of arteriosclerotic plaques in cerebral arteries. HIV patients have a small but definite increased incidence of stroke which may be due to either HIV infection or opportunistic VZV infection in these immunocompromised individuals. Importantly, many described cases of vasculopathy in HIV-infected patients were not studied for the presence of anti-VZV IgG antibody in CSF, a sensitive indicator of VZV vasculopathy. Unlike the well-documented role of VZV in vasculopathy, evidence for a causal link between HIV or CMV and stroke remains indirect and awaits further studies demonstrating productive HIV and CMV infection of cerebral arteries in stroke patients. Nonetheless, all three viruses have been implicated in stroke and should be considered in clinical diagnoses.
Collapse
Affiliation(s)
- M.A. Nagel
- Department of Neurology, University of Colorado Denver School of Medicine, 12700 East 19 Avenue, Box B182, Denver, CO 80045, USA
| | - R. Mahalingam
- Department of Neurology, University of Colorado Denver School of Medicine, 12700 East 19 Avenue, Box B182, Denver, CO 80045, USA
| | - R.J. Cohrs
- Department of Neurology, University of Colorado Denver School of Medicine, 12700 East 19 Avenue, Box B182, Denver, CO 80045, USA
| | - D. Gilden
- Department of Neurology, University of Colorado Denver School of Medicine, 12700 East 19 Avenue, Box B182, Denver, CO 80045, USA
- Department of Microbiology, University of Colorado Denver School of Medicine, 12700 East 19 Avenue, Box B182, Denver, CO 80045, USA
| |
Collapse
|
15
|
Molecular characterization of varicella zoster virus in latently infected human ganglia: physical state and abundance of VZV DNA, Quantitation of viral transcripts and detection of VZV-specific proteins. Curr Top Microbiol Immunol 2010; 342:229-41. [PMID: 20186615 DOI: 10.1007/82_2009_2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Varicella zoster virus (VZV) establishes latency in neurons of human peripheral ganglia where the virus genome is most likely maintained as a circular episome bound to histones. There is considerable variability among individuals in the number of latent VZV DNA copies. The VZV DNA burden does not appear to exceed that of herpes simplex type 1 (HSV-1). Expression of VZV genes during latency is highly restricted and is regulated epigenetically. Of the VZV open reading frames (ORFs) that have been analyzed for transcription during latency using cDNA sequencing, only ORFs 21, 29, 62, 63, and 66 have been detected. VZV ORF 63 is the most frequently and abundantly transcribed VZV gene detected in human ganglia during latency, suggesting a critical role for this gene in maintaining the latent state and perhaps the early stages of virus reactivation. The inconsistent detection and low abundance of other VZV transcripts suggest that these genes play secondary roles in latency or possibly reflect a subpopulation of neurons undergoing VZV reactivation. New technologies, such as GeXPS multiplex PCR, have the sensitivity to detect multiple low abundance transcripts and thus provide a means to elucidate the entire VZV transcriptome during latency.
Collapse
|
16
|
Gilden D, Nagel MA, Mahalingam R, Mueller NH, Brazeau EA, Pugazhenthi S, Cohrs RJ. Clinical and molecular aspects of varicella zoster virus infection. FUTURE NEUROLOGY 2009; 4:103-117. [PMID: 19946620 PMCID: PMC2782836 DOI: 10.2217/14796708.4.1.103] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A declining cell-mediated immunity to varicella zoster virus (VZV) with advancing age or immunosuppression results in virus reactivation from latently infected human ganglia anywhere along the neuraxis. Virus reactivation produces zoster, often followed by chronic pain (postherpetic neuralgia or PHN) as well as vasculopathy, myelopathy, retinal necrosis and cerebellitis. VZV reactivation also produces pain without rash (zoster sine herpete). Vaccination after age 60 reduces the incidence of shingles by 51%, PHN by 66% and the burden of illness by 61%. However, even if every healthy adult over age 60 years is vaccinated, there would still be about 500,000 zoster cases annually in the United States alone, about 200,000 of whom will experience PHN. Analyses of viral nucleic acid and gene expression in latently infected human ganglia and in an animal model of varicella latency in primates are serving to determine the mechanism(s) of VZV reactivation with the aim of preventing reactivation and the clinical sequelae.
Collapse
Affiliation(s)
- Don Gilden
- Author for correspondence: Department of Neurology, University of Colorado Denver School of Medicine, 4200 E. 9 Avenue, Mail Stop B182, Denver, CO 80262, USA. Tel: 1-303-315-8281; Fax: 1-303-315-8281;
| | | | | | | | | | | | | |
Collapse
|
17
|
Varicella-zoster virus immediate-early 63 protein interacts with human antisilencing function 1 protein and alters its ability to bind histones h3.1 and h3.3. J Virol 2008; 83:200-9. [PMID: 18971269 DOI: 10.1128/jvi.00645-08] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Varicella-zoster virus (VZV) immediate-early 63 protein (IE63) is abundantly expressed during both acute infection in vitro and latent infection in human ganglia. Using the yeast two-hybrid system, we found that VZV IE63 interacts with human antisilencing function 1 protein (ASF1). ASF1 is a nucleosome assembly factor which is a member of the H3/H4 family of histone chaperones. IE63 coimmunoprecipitated and colocalized with ASF1 in transfected cells expressing IE63 and in VZV-infected cells. IE63 also colocalized with ASF1 in both lytic and latently VZV-infected enteric neurons. ASF1 exists in two isoforms, ASF1a and ASF1b, in mammalian cells. IE63 preferentially bound to ASF1a, and the amino-terminal 30 amino acids of ASF1a were critical for its interaction with IE63. VZV IE63 amino acids 171 to 208 and putative phosphorylation sites of IE63, both of which are critical for virus replication and latency in rodents, were important for the interaction of IE63 with ASF1. Finally, we found that IE63 increased the binding of ASF1 to histone H3.1 and H3.3, which suggests that IE63 may help to regulate levels of histones in virus-infected cells. Since ASF1 mediates eviction and deposition of histones during transcription, the interaction of VZV IE63 with ASF1 may help to regulate transcription of viral or cellular genes during lytic and/or latent infection.
Collapse
|
18
|
Mueller NH, Gilden DH, Cohrs RJ, Mahalingam R, Nagel MA. Varicella zoster virus infection: clinical features, molecular pathogenesis of disease, and latency. Neurol Clin 2008; 26:675-97, viii. [PMID: 18657721 PMCID: PMC2754837 DOI: 10.1016/j.ncl.2008.03.011] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Varicella zoster virus (VZV) is an exclusively human neurotropic alphaherpesvirus. Primary infection causes varicella (chickenpox), after which virus becomes latent in cranial nerve ganglia, dorsal root ganglia, and autonomic ganglia along the entire neuraxis. Years later, in association with a decline in cell-mediated immunity in elderly and immunocompromised individuals, VZV reactivates and causes a wide range of neurologic disease. This article discusses the clinical manifestations, treatment, and prevention of VZV infection and reactivation; pathogenesis of VZV infection; and current research focusing on VZV latency, reactivation, and animal models.
Collapse
Affiliation(s)
- Niklaus H Mueller
- Department of Neurology, University of Colorado School of Medicine, 4200 East 9th Avenue, Mail Stop B182, Denver, CO 80262, USA
| | | | | | | | | |
Collapse
|
19
|
Steiner I, Kennedy PGE, Pachner AR. The neurotropic herpes viruses: herpes simplex and varicella-zoster. Lancet Neurol 2007; 6:1015-28. [PMID: 17945155 DOI: 10.1016/s1474-4422(07)70267-3] [Citation(s) in RCA: 325] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Herpes simplex viruses types 1 and 2 (HSV1 and HSV2) and varicella-zoster virus (VZV) establish latent infection in dorsal root ganglia for the entire life of the host. From this reservoir they can reactivate to cause human morbidity and mortality. Although the viruses vary in the clinical disorders they cause and in their molecular structure, they share several features that affect the course of infection of the human nervous system. HSV1 is the causative agent of encephalitis, corneal blindness, and several disorders of the peripheral nervous system; HSV2 is responsible for meningoencephalitis in neonates and meningitis in adults. Reactivation of VZV, the pathogen of varicella (chickenpox), is associated with herpes zoster (shingles) and central nervous system complications such as myelitis and focal vasculopathies. We review the biological, medical, and neurological aspects of acute, latent, and reactivated infections with the neurotropic herpes viruses.
Collapse
Affiliation(s)
- Israel Steiner
- Neurological Sciences Unit, Hadassah University Hospital, Mount Scopus, Jerusalem, Israel.
| | | | | |
Collapse
|
20
|
Mahalingam R, Traina-Dorge V, Wellish M, Lorino R, Sanford R, Ribka EP, Alleman SJ, Brazeau E, Gilden DH. Simian varicella virus reactivation in cynomolgus monkeys. Virology 2007; 368:50-9. [PMID: 17651776 DOI: 10.1016/j.virol.2007.06.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 06/02/2007] [Accepted: 06/11/2007] [Indexed: 10/23/2022]
Abstract
SVV infection of primates closely resembles VZV infection of humans. Like VZV, SVV becomes latent in ganglionic neurons. We used this model to study the effect of immunosuppression on varicella reactivation. Cynomolgus monkeys latently infected with SVV were irradiated and treated with tacrolimus and prednisone. Of four latently infected monkeys that were immunosuppressed and subjected to the stress of transportation and isolation, one developed zoster, and three others developed features of subclinical reactivation. Another non-immunosuppressed latently infected monkey that was subjected to the same stress of travel and isolation showed features of subclinical reactivation. Virus reactivation was confirmed not only by the occurrence of zoster in one monkey, but also by the presence of late SVV RNA in ganglia, and the detection of SVV DNA in non-ganglionic tissue, and SVV antigens in skin, ganglia and lung.
Collapse
Affiliation(s)
- Ravi Mahalingam
- Department of Neurology , University of Colorado Health Sciences Center, Denver, CO 80262, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Grinfeld E, Kennedy PGE. The pattern of viral persistence in monkeys intra-tracheally infected with Simian varicella virus. Virus Genes 2007; 35:289-92. [PMID: 17253123 DOI: 10.1007/s11262-007-0077-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Accepted: 01/02/2007] [Indexed: 10/23/2022]
Abstract
In situ PCR (ISPCR) and in situ hybridisation (ISH) was performed on 32 tissues from 10 monkeys, intra-tracheally (IT) infected with simian varicella virus (SVV) and 5 tissues from 3 uninfected control animals. The results showed persistence of SVV DNA up to 2 years post-infection (pi) and the localisation of SVV to be confined to neurons except at time points 9 and 10 months pi where SVV positive satellite cells were also detected. There was no evidence for transcription of SVV ORFs 63 and 21 in the ganglia of the one IT infected and 2 naturally infected monkeys investigated using RNA ISH.
Collapse
Affiliation(s)
- Esther Grinfeld
- Department of Neurology, Institute of Neurological Sciences, University of Glasgow, Southern General Hospital, 1345 Govan Road, Glasgow G51 4TF, Scotland, UK
| | | |
Collapse
|
22
|
Hamza MA, Higgins DM, Ruyechan WT. Two alphaherpesvirus latency-associated gene products influence calcitonin gene-related peptide levels in rat trigeminal neurons. Neurobiol Dis 2006; 25:553-60. [PMID: 17184994 PMCID: PMC1853241 DOI: 10.1016/j.nbd.2006.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 09/22/2006] [Accepted: 10/29/2006] [Indexed: 11/27/2022] Open
Abstract
Herpes simplex virus type-1 (HSV-1) initially infects mucoepithelial tissues of the eye and the orofacial region. Subsequently, the virus is retrogradely transported through the axons of the trigeminal sensory neurons. HSV-1 establishes a life-long latent infection in these neurons, during which the transcription of the viral genome is silent, except for the sequences encoding the latency-associated transcript (LAT). To determine if HSV-1 latency might affect calcitonin gene-related peptide (CGRP) expression in trigeminal sensory neurons, we transfected primary neuronal cultures of trigeminal ganglia from rat embryos with plasmids expressing LAT. In the presence of Bone Morphogenetic Protein-7 (BMP7), CGRP was expressed in 49% of sensory neurons. However, this percentage was reduced to 19% in neurons transfected with LAT expressing plasmids. We also found that transfection of the IE63 gene of varicella-zoster virus (VZV) reduced the percentage of trigeminal neurons containing CGRP. However, the observed effect of IE63 in contrast to that of LAT was completely reversed by treatment of cultures with MgCl2, which indicates that the effect of IE63 was due to increased release of CGRP from trigeminal neurons. We provide here the first evidence that HSV-1 LAT decreases the level of CGRP in trigeminal neurons. These effects may be important for reducing the neuroinflammatory response, thus protecting host neuronal cells during HSV-1 latency in trigeminal neurons. In contrast, increased release of CGRP in the presence of IE63 protein may contribute to the neuralgias associated with VZV infection.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Protein 7
- Bone Morphogenetic Proteins/pharmacology
- Calcitonin Gene-Related Peptide/metabolism
- Cell Survival/physiology
- Cells, Cultured
- DNA, Viral/genetics
- DNA, Viral/pharmacology
- Ganglia, Spinal/cytology
- Ganglia, Spinal/physiology
- Ganglia, Spinal/virology
- Gene Deletion
- Gene Expression Regulation, Viral/drug effects
- Gene Expression Regulation, Viral/physiology
- Herpes Simplex/physiopathology
- Herpes Simplex/virology
- Herpesvirus 1, Human/genetics
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Magnesium Chloride/pharmacology
- MicroRNAs
- Neurons, Afferent/cytology
- Neurons, Afferent/physiology
- Neurons, Afferent/virology
- Rats
- Rats, Sprague-Dawley
- Transfection
- Transforming Growth Factor beta/pharmacology
- Trigeminal Ganglion/cytology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
- Viral Proteins/genetics
- Virus Latency/physiology
Collapse
Affiliation(s)
- Mohamed A Hamza
- Department of Pharmacology and Toxicology, SUNY, Buffalo, NY 14214, USA
| | | | | |
Collapse
|
23
|
Kolappaswamy K, Mahalingam R, Traina-Dorge V, Shipley ST, Gilden DH, Kleinschmidt-Demasters BK, McLeod CG, Hungerford LL, DeTolla LJ. Disseminated simian varicella virus infection in an irradiated rhesus macaque (Macaca mulatta). J Virol 2006; 81:411-5. [PMID: 17079326 PMCID: PMC1797240 DOI: 10.1128/jvi.01825-06] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We describe correlative clinicopathological/virological findings from a simian varicella virus (SVV)-seronegative monkey that developed disseminated varicella 105 days after gamma-irradiation. Twelve other monkeys in the colony were also irradiated, none of which developed varicella. Before irradiation, sera from the monkey that developed disseminated infection and one asymptomatic monkey were available. Analysis indicated that subclinical reactivation of latent SVV from an asymptomatic irradiated monkey likely led to disseminated varicella in the seronegative irradiated monkey. These findings parallel those from humans with disseminated varicella infection and support the usefulness of SVV infection as a model for human varicella-zoster virus infection, particularly virus reactivation after gamma-irradiation.
Collapse
Affiliation(s)
- Krishnan Kolappaswamy
- Program of Comparative Medicine, Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Cohrs RJ, Laguardia JJ, Gilden D. Distribution of latent herpes simplex virus type-1 and varicella zoster virus DNA in human trigeminal Ganglia. Virus Genes 2006; 31:223-7. [PMID: 16025248 DOI: 10.1007/s11262-005-1799-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Accepted: 04/20/2005] [Indexed: 10/25/2022]
Abstract
Trigeminal ganglia removed at autopsy from immunocompetent individuals without cutaneous signs of herpesvirus infection were fixed, cut into 5-microm sections, and screened at 100-microm intervals (20 adjacent sections) by PCR for latent herpes simplex type 1(HSV-1) and varicella zoster virus (VZV) DNA. Sections that contained >5 neurons with nuclei stained by hematoxylin/eosin revealed HSV-1 DNA in most samples and VZV DNA in approximately 50% of samples. HSV-1 and VZV DNA were distributed throughout each latently infected ganglion.
Collapse
Affiliation(s)
- Randall J Cohrs
- Departments of Neurology, University of Colorado Health Sciences Center, 4200 E. 9th Avenue, Mail Stop B182, Denver, CO 80262, USA.
| | | | | |
Collapse
|
25
|
Abstract
Herpesviruses are among the most successful human pathogens. In healthy individuals, primary infection is most often inapparent. After primary infection, the virus becomes latent in ganglia or blood mononuclear cells. Three major subfamilies of herpesviruses have been identified based on similar growth characteristics, genomic structure, and tissue predilection. Each herpesvirus has evolved its own unique ecological niche within the host that allows the maintenance of latency over the life of the individual (e.g. the adaptation to specific cell types in establishing latent infection and the mechanisms, including expression of different sets of genes, by which the virus remains latent). Neurotropic alphaherpesviruses become latent in dorsal root ganglia and reactivate to produce epidermal ulceration, either localized (herpes simplex types 1 and 2) or spread over several dermatomes (varicalla-zoster virus). Human cytomegalovirus, the prototype betaherpesvirus, establishes latency in bone marrow-derived myeloid progenitor cells. Reactivation of latent virus is especially serious in transplant recipients and AIDS patients. Lymphotropic gammaherpesviruses (Epstein-Barr virus) reside latent in resting B cells and reactivate to produce various neurologic complications. This review highlights the alphaherpesvirus, specifically herpes simplex virus type 1 and varicella-zoster virus, and describes the characteristics of latent infection.
Collapse
Affiliation(s)
- R J Cohrs
- Department of Neurology, University of Colorado, Health Sciences Center, Denver 80262, USA.
| | | |
Collapse
|
26
|
Wang K, Lau TY, Morales M, Mont EK, Straus SE. Laser-capture microdissection: refining estimates of the quantity and distribution of latent herpes simplex virus 1 and varicella-zoster virus DNA in human trigeminal Ganglia at the single-cell level. J Virol 2006; 79:14079-87. [PMID: 16254342 PMCID: PMC1280223 DOI: 10.1128/jvi.79.22.14079-14087.2005] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There remains uncertainty and some controversy about the percentages and types of cells in human sensory nerve ganglia that harbor latent herpes simplex virus 1 (HSV-1) and varicella-zoster virus (VZV) DNA. We developed and validated laser-capture microdissection and real-time PCR (LCM/PCR) assays for the presence and copy numbers of HSV-1 gG and VZV gene 62 sequences in single cells recovered from sections of human trigeminal ganglia (TG) obtained at autopsy. Among 970 individual sensory neurons from five subjects, 2.0 to 10.5% were positive for HSV-1 DNA, with a median of 11.3 copies/positive cell, compared with 0.2 to 1.5% of neurons found to be positive by in situ hybridization (ISH) for HSV-1 latency-associated transcripts (LAT), the classical surrogate marker for HSV latency. This indicates a more pervasive latent HSV-1 infection of human TG neurons than originally thought. Combined ISH/LCM/PCR assays revealed that the majority of the latently infected neurons do not accumulate LAT to detectable levels. We detected VZV DNA in 1.0 to 6.9% of individual neurons from 10 subjects. Of the total 1,722 neurons tested, 4.1% were VZV DNA positive, with a median of 6.9 viral genomes/positive cell. After removal by LCM of all visible neurons on a slide, all surrounding nonneuronal cells were harvested and assayed: 21 copies of HSV-1 DNA were detected in approximately 5,200 nonneuronal cells, while nine VZV genomes were detected in approximately 14,200 nonneuronal cells. These data indicate that both HSV-1 and VZV DNAs persist in human TG primarily, if not exclusively, in a moderate percentage of neuronal cells.
Collapse
MESH Headings
- Base Sequence
- DNA Primers
- DNA, Viral/genetics
- DNA, Viral/isolation & purification
- DNA, Viral/ultrastructure
- Gene Expression Regulation, Viral
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/isolation & purification
- Herpesvirus 3, Human/genetics
- Herpesvirus 3, Human/isolation & purification
- Humans
- Lasers
- Microdissection/methods
- Polymerase Chain Reaction
- RNA, Viral/genetics
- RNA, Viral/isolation & purification
- Trigeminal Ganglion/virology
Collapse
Affiliation(s)
- Kening Wang
- Medical Virology Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
27
|
Vossen MTM, Gent MR, Peters KMC, Wertheim-van Dillen PME, Dolman KM, van Breda A, van Lier RAW, Kuijpers TW. Persistent detection of varicella-zoster virus DNA in a previously healthy child after severe chickenpox. J Clin Microbiol 2005; 43:5614-21. [PMID: 16272494 PMCID: PMC1287842 DOI: 10.1128/jcm.43.11.5614-5621.2005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In immunocompetent children with primary varicella-zoster virus (VZV) infection, peak viral loads are detected in peripheral blood near the onset of the vesicular rash. VZV DNA concentrations normally diminish and become undetectable within 3 weeks after the appearance of the exanthem. Here, we present a previously healthy, human immunodeficiency virus-negative, 4-year-old boy admitted with severe varicella. High viral loads (>340,000 copies/ml) were found in his blood, and the viral loads remained high for at least 1.5 years. Clinical recovery preceded complete clearance of the virus. General and VZV-specific immune reactivity were intact. NK cells and CD8(+) T cells were activated during acute infection, and VZV-specific CD4(+) T cells were detected at high frequencies. VZV DNA was initially detected in B cells, NK cells, and both CD4(+) and CD8(+) T cells. In contrast, during the persistent phase of VZV DNA detection, the viral DNA was primarily located in CD8(+) T cells. For the first time, we describe the persistent detection of VZV DNA in a previously healthy child.
Collapse
Affiliation(s)
- Mireille T M Vossen
- Academic Medical Center, Emma Children's Hospital, Room G8-205, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Zerboni L, Ku CC, Jones CD, Zehnder JL, Arvin AM. Varicella-zoster virus infection of human dorsal root ganglia in vivo. Proc Natl Acad Sci U S A 2005; 102:6490-5. [PMID: 15851670 PMCID: PMC1088374 DOI: 10.1073/pnas.0501045102] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Varicella-zoster virus (VZV) causes varicella and establishes latency in sensory ganglia. VZV reactivation results in herpes zoster. We developed a model using human dorsal root ganglion (DRG) xenografts in severe combined immunodeficient (SCID) mice to investigate VZV infection of differentiated neurons and satellite cells in vivo. DRG engrafted under the kidney capsule and contained neurons and satellite cells within a typical DRG architecture. VZV clinical isolates infected the neurons within DRG. At 14 days postinfection, VZ virions were detected by electron microscopy in neuronal cell nuclei and cytoplasm but not in satellite cells. The VZV genome copy number was 7.1 x 10(7) to 8.0 x 10(8) copies per 10(5) cells, and infectious virus was recovered. This initial phase of viral replication was followed within 4-8 weeks by a transition to VZV latency, characterized by the absence of infectious virus release, the cessation of virion assembly, and a reduction in VZV genome copies to 3.7 x 10(5) to 4.7 x 10(6) per 10(5) cells. VZV persistence in DRG was achieved without any requirement for VZV-specific adaptive immunity and was associated with continued transcription of the ORF63 regulatory gene. The live attenuated varicella vaccine virus exhibited the same pattern of short-term replication, persistence of viral DNA, and prominent ORF63 transcription as the clinical isolates. VZV-infected T cells transferred virus from the circulation into DRG, suggesting that VZV lymphotropism facilitates its neurotropism. DRG xenografts may be useful for investigating neuropathogenic mechanisms of other human viruses.
Collapse
MESH Headings
- Animals
- Chickenpox/pathology
- DNA Primers
- Ganglia, Spinal/pathology
- Ganglia, Spinal/transplantation
- Ganglia, Spinal/virology
- Genome, Viral
- Herpesvirus 3, Human/genetics
- Herpesvirus 3, Human/physiology
- Humans
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Immunohistochemistry
- In Situ Hybridization
- Mice
- Mice, SCID
- Microscopy, Electron, Transmission
- Neurons/ultrastructure
- Neurons/virology
- Reverse Transcriptase Polymerase Chain Reaction
- Satellite Cells, Perineuronal/ultrastructure
- Satellite Cells, Perineuronal/virology
- T-Lymphocytes/virology
- Transplantation, Heterologous
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
- Virion/physiology
- Virion/ultrastructure
- Virus Replication/physiology
Collapse
Affiliation(s)
- Leigh Zerboni
- Department of Pediatrics,Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | | | | | |
Collapse
|
29
|
Grinfeld E, Sadzot-Delvaux C, Kennedy PGE. Varicella-Zoster virus proteins encoded by open reading frames 14 and 67 are both dispensable for the establishment of latency in a rat model. Virology 2004; 323:85-90. [PMID: 15165821 DOI: 10.1016/j.virol.2004.02.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2004] [Revised: 02/17/2004] [Accepted: 02/25/2004] [Indexed: 10/26/2022]
Abstract
A rat model of Varicella-Zoster virus (VZV) provides a system in which to investigate the molecular determinants of viral latency in dorsal root ganglia (DRG). In this study, we determined whether the VZV glycoproteins gC and gI, corresponding to VZV open reading frames (ORFs) 14 and 67, respectively, were required for the establishment of latency in this model. A VZV gI deletion mutant (DeltagI) derived from a recombinant Oka (rOka) cosmid and a gC null mutant obtained from a clinical isolate were inoculated into the footpads of 6-week-old rats, and the presence of viral DNA and eight different VZV RNA transcripts corresponding to the three classes of genes was investigated by in situ RT-PCR amplification and in situ hybridization (ISH) in the DRG at 1 week, 1 month, and 18-24 months after infection. VZV DNA and restricted RNA expression was established with both deletion mutants as well as the parental rOka virus. Both VZV DNA and RNA were detected in neurons and non-neuronal cells. The pattern of viral RNA expression detected with both gC and gI mutants was restricted with transcripts for VZV genes 62 and 63 most frequently expressed 18-24 months after infection. Transcripts for VZV genes 18, 28, and 29 were also detected at these time points but at a slightly lower frequency. Transcripts for the late gene 40 were never detected. We conclude that VZV ORFs 14 and 67 are dispensable for the establishment of a latent infection in this model.
Collapse
Affiliation(s)
- Esther Grinfeld
- Department of Neurology, Division of Clinical Neurosciences, Institute of Neurological Sciences, Southern General Hospital, Glasgow, Scotland, UK
| | | | | |
Collapse
|
30
|
Abstract
In normal infants and children, zoster can occur at any time after varicella or varicella vaccination. It is usually diagnosed clinically: a unilateral vesicular eruption following a dermatome or dermatomes. The incidence of zoster increases with age, although children who have had varicella during the first year of life (or in utero) are at increased risk of developing zoster. The incidence of zoster is less after varicella vaccination than after natural infection. Zoster in children is frequently mild, postzoster neuralgia rarely if ever occurs, and antiviral therapy is usually not needed. In a previously normal child with zoster, if the history and physical examination are normal, a laboratory search for occult immunodeficiency or malignancy is not needed. We present five cases of zoster in healthy children and review zoster in the pediatric age group.
Collapse
Affiliation(s)
- Henry M Feder
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | | |
Collapse
|
31
|
Taylor SL, Kinchington PR, Brooks A, Moffat JF. Roscovitine, a cyclin-dependent kinase inhibitor, prevents replication of varicella-zoster virus. J Virol 2004; 78:2853-62. [PMID: 14990704 PMCID: PMC353735 DOI: 10.1128/jvi.78.6.2853-2862.2004] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Understanding the interactions between varicella-zoster virus (VZV) and host cells can be addressed by using small molecule inhibitors of cellular enzymes. Roscovitine (Rosco) is a purine derivative that inhibits cyclin-dependent kinase 1 (cdk1), cdk2, cdk5, cdk7, and cdk9, which are key regulators of the cell cycle and transcription. Herpesviruses are known to interact with cell cycle proteins; thus, the antiviral effects of Rosco on VZV growth were evaluated. In a plaque reduction assay, 25 micro M Rosco prevented VZV replication, and the antiviral effect was reversible for at least up to 24 h posttreatment. Rosco also reduced expression of the major transactivator, IE62, over 48 h. Confocal microscopy studies indicated that Rosco caused the immediate-early proteins ORF4 and IE62 to abnormally localize in infected cells and prevented cell-cell spread of VZV over 48 h. Rosco was found to inhibit VZV DNA synthesis as measured by real-time PCR, and this technique was used to estimate the 50% effective concentration (EC(50)) of 14 micro M. This value was close to the EC(50) estimate of 12 micro M determined from plaque reduction assays. At 25 micro M, Rosco was not cytotoxic over 48 h in a neutral red uptake assay, and proliferation was slowed as the cells accumulated in a G(2)-like state. These results demonstrate the importance of cdk's in VZV replication and suggest that cdk inhibitors could serve as useful VZV antivirals.
Collapse
Affiliation(s)
- Shannon L Taylor
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | | | | | | |
Collapse
|
32
|
Mehta SK, Cohrs RJ, Forghani B, Zerbe G, Gilden DH, Pierson DL. Stress-induced subclinical reactivation of varicella zoster virus in astronauts. J Med Virol 2004; 72:174-9. [PMID: 14635028 DOI: 10.1002/jmv.10555] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Varicella zoster virus (VZV) becomes latent in human ganglia after primary infection. VZV reactivation occurs primarily in elderly individuals, organ transplant recipients, and patients with cancer and AIDS, correlating with a specific decline in cell-mediated immunity to the virus. VZV can also reactivate after surgical stress. The unexpected occurrence of thoracic zoster 2 days before space flight in a 47-year-old healthy astronaut from a pool of 81 physically fit astronauts prompted our search for VZV reactivation during times of stress to determine whether VZV can also reactivate after non-surgical stress. We examined total DNA extracted from 312 saliva samples of eight astronauts before, during, and after space flight for VZV DNA by polymerase chain reaction: 112 samples were obtained 234-265 days before flight, 84 samples on days 2 through 13 of space flight, and 116 samples on days 1 through 15 after flight. Before space flight, only one of the 112 saliva samples from a single astronaut was positive for VZV DNA. In contrast, during and after space flight, 61 of 200 (30%) saliva samples were positive in all eight astronauts. No VZV DNA was detected in any of 88 saliva samples from 10 healthy control subjects. These results indicate that VZV can reactivate subclinically in healthy individuals after non-surgical stress.
Collapse
Affiliation(s)
- Satish K Mehta
- Enterprise Advisory Services Inc., Lyndon B. Johnson Space Center, Houston, Texas
| | | | | | | | | | | |
Collapse
|
33
|
Gilden DH, Cohrs RJ, Mahalingam R. Clinical and molecular pathogenesis of varicella virus infection. Viral Immunol 2004; 16:243-58. [PMID: 14583142 DOI: 10.1089/088282403322396073] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Varicella zoster virus (VZV) is a neurotropic human herpesvirus that infects nearly all humans and causes chickenpox (varicella). After chickenpox, VZV becomes latent in cranial nerve, dorsal root, and autonomic nervous system ganglia along the entire neuraxis. Virus reactivation produces shingles (zoster), characterized by pain and rash usually restricted to 1-3 dermatomes. Zoster is often complicated by postherpetic neuralgia (PHN), pain that persists for months to years after rash resolves. Virus may also spread to the spinal cord and blood vessels of the brain, producing a unifocal or multifocal vasculopathy, particularly in immunocompromised individuals. The increased incidence of zoster in elderly and immunocompromised individuals appears to be due to a VZV-specific host immunodeficiency. PHN may reflect a chronic VZV ganglionitis, and VZV vasculopathy is due to productive virus infection in cerebral arteries. Strategies that might boost host cell-mediated immunity to VZV are discussed, as well as the physical state of viral nucleic acid during latency and the possible mechanisms by which herpesvirus latency is maintained and virus is reactivated. A current summary of varicella latency and pathogenesis produced by simian varicella virus (SVV), the counterpart of human VZV, points to the usefulness of a primate model of natural infection to study varicella latency, as well as the experimental model of intratracheal inoculation to study the effectiveness of antiviral agents in driving persistent varicella virus into a latent state.
Collapse
Affiliation(s)
- Donald H Gilden
- Department of Neurology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | | | |
Collapse
|
34
|
Theil D, Paripovic I, Derfuss T, Herberger S, Strupp M, Arbusow V, Brandt T. Dually infected (HSV-1/VZV) single neurons in human trigeminal ganglia. Ann Neurol 2003; 54:678-82. [PMID: 14595659 DOI: 10.1002/ana.10746] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Human trigeminal ganglia were tested by double fluorescence in situ hybridization for the presence and distribution of herpes simplex virus type 1 (HSV-1) and varicella-zoster virus (VZV) latency. Latency transcripts of both viruses were detected in common areas within the ganglia. Also, a few single neurons were shown to harbor HSV-1 and VZV together.
Collapse
Affiliation(s)
- Diethilde Theil
- Department of Neurology, Klinikum Grosshadern, Ludwig-Maximilians University, Marchioninistrasse 23, 81377 Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
35
|
Schmidt M, Kress M, Heinemann S, Fickenscher H. Varicella-zoster virus isolates, but not the vaccine strain OKA, induce sensitivity to alpha-1 and beta-1 adrenergic stimulation of sensory neurones in culture. J Med Virol 2003; 70 Suppl 1:S82-9. [PMID: 12627494 DOI: 10.1002/jmv.10327] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The reactivation of varicella-zoster virus (VZV) from its persistent state in sensory neurones causes shingles and induces severe, long-lasting pain and hyperalgesia that often lead to postherpetic neuralgia. To investigate the VZV-induced neuropathic changes, we established conditions for the active infection of sensory neurones from rat dorsal root ganglia in vitro. After 2 days of culture, up to 50% of the cells expressed viral antigens of the immediate-early and late replication phase. The intracellular calcium ion concentration was monitored in individual cells by microfluorimetry. Whereas the calcium response to capsaicin was preserved, the VZV-infected neurones gained an unusual sensitivity to noradrenaline stimulation in contrast to non-infected cells. The adrenergic agonists phenylephrine and isoproterenol had a similar efficacy demonstrating that both alpha(1)- and beta(1)-adrenoreceptors were involved. The sensitivity to adrenergic stimulation was observed after infection with different wildtype isolates, but not with the attenuated vaccine strain OKA. The lack of noradrenaline sensitivity of vaccine-infected neurones demands a structural comparison of wildtype and vaccine viruses with and without phenotype. A partial sequence evaluation (26 kb) of the European OKA vaccine strain surprisingly revealed a series of nucleotide exchanges in comparison to presumably identical OKA strains from other sources, although VZV is generally considered genetically stable. In summary, we report that the infection with wildtype VZV isolates, but not with the vaccine strain, induces noradrenaline sensitivity in sensory neurones, which correlates with clinical and experimental observations of adrenergic effects involved in VZV-induced neuralgia.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Chickenpox Vaccine
- Herpes Zoster/etiology
- Herpes Zoster/physiopathology
- Herpes Zoster/virology
- Herpesvirus 3, Human/genetics
- Herpesvirus 3, Human/isolation & purification
- Herpesvirus 3, Human/pathogenicity
- In Vitro Techniques
- Isoproterenol/pharmacology
- Neurons, Afferent/drug effects
- Neurons, Afferent/physiology
- Neurons, Afferent/virology
- Norepinephrine/pharmacology
- Phenylephrine/pharmacology
- Rats
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-1/physiology
- Receptors, Adrenergic, beta-1/drug effects
- Receptors, Adrenergic, beta-1/physiology
- Virus Cultivation
Collapse
Affiliation(s)
- Michaela Schmidt
- Abteilung Virologie, Hygiene-Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
36
|
Levin MJ, Cai GY, Manchak MD, Pizer LI. Varicella-zoster virus DNA in cells isolated from human trigeminal ganglia. J Virol 2003; 77:6979-87. [PMID: 12768016 PMCID: PMC156183 DOI: 10.1128/jvi.77.12.6979-6987.2003] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To determine the type of cell(s) that contain latent varicella-zoster virus (VZV) DNA, we prepared pure populations of neurons and satellite cells from trigeminal ganglia of 18 humans who had previously had a VZV infection. VZV DNA was present in 34 of 2,226 neurons (1.5%) and in none of 20,700 satellite cells. There was an average of 4.7 (range of 2 to 9) copies of VZV DNA per latently infected neuron. Latent VZV DNA was primarily present in large neurons, whereas the size distribution of herpes simplex virus DNA was markedly different.
Collapse
Affiliation(s)
- Myron J Levin
- Section of Pediatric Infectious Diseases, School of Medicine, University of Colorado, Denver, Colorado 80262, USA.
| | | | | | | |
Collapse
|
37
|
Arvin AM, Sharp M, Moir M, Kinchington PR, Sadeghi-Zadeh M, Ruyechan WT, Hay J. Memory cytotoxic T cell responses to viral tegument and regulatory proteins encoded by open reading frames 4, 10, 29, and 62 of varicella-zoster virus. Viral Immunol 2003; 15:507-16. [PMID: 12479399 DOI: 10.1089/088282402760312377] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cytotoxic T cell recognition of tegument and regulatory proteins encoded by open reading frames (ORFs) 4, 10, 29, and 62 of varicella-zoster virus (VZV) was evaluated using limiting dilution conditions to estimate the precursor frequencies of memory T cells specific for these proteins in immune subjects. Responder cell frequencies for ORFs 4, 10, and 62 gene products, which are virion tegument components and function as immediate early viral transactivating proteins, were equivalent. CTLp recognition of VZV proteins made in latently infected cells, which include ORF4 and ORF62 proteins, was not maintained preferentially when compared to ORF10 protein, which has not been shown to be expressed during latency. T cell recognition of ORF29 protein, the major DNA binding protein, which is expressed during replication but not incorporated into the virion tegument, was less common than responses to ORFs 4, 10, and 62 gene products. Older individuals had diminished numbers of memory CTLp that lysed autologous targets expressing IE62 protein; these responses were increased after immunization with live attenuated varicella vaccine to the range observed in younger adults. Adaptive immunity to VZV is characterized by a broad repertoire of memory CTL responses to proteins that comprise the virion tegument and regulate viral gene expression in infected cells.
Collapse
Affiliation(s)
- Ann M Arvin
- Department of Pediatrics Stanford University School of Medicine, Stanford, California 94305, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Mitchell BM, Bloom DC, Cohrs RJ, Gilden DH, Kennedy PGE. Herpes simplex virus-1 and varicella-zoster virus latency in ganglia. J Neurovirol 2003; 9:194-204. [PMID: 12707850 DOI: 10.1080/13550280390194000] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2002] [Revised: 11/11/2002] [Accepted: 11/13/2002] [Indexed: 01/07/2023]
Abstract
Two human alpha-herpesviruses, herpes simplex virus (HSV)-1 and varicella zoster virus (VZV), account for the most frequent and serious neurologic disease caused by any of the eight human herpesviruses. Both HSV-1 and VZV become latent in ganglia. In this review, the authors describe features of latency for these viruses, such as distribution, prevalence, abundance, and configuration of viral DNA in latently infected human ganglia, as well as transcription, translation, and cell type infected. Studies of viral latency in animal models are also discussed. For each virus, remaining questions and future studies to understand the mechanism of latency are discussed with respect to prevention of serious cutaneous, ocular, and neurologic disease produced by virus reactivation.
Collapse
Affiliation(s)
- Bradley M Mitchell
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Varicella-zoster virus (VZV) is a human herpesvirus which causes varicella (chickenpox) as a primary infection, and, following a variable period during which it remains in latent form in trigeminal and dorsal root ganglia, reactivates in later life to cause herpes zoster (shingles). VZV is a significant cause of neurological disease including post-herpetic neuralgia which may be persistent and highly resistant to treatment, and small and large vessel encephalitis. VZV infections are more frequent with advancing age and in immunocompromised individuals. An understanding of the mechanisms of latency is crucial in developing effective therapies for VZV infections of the nervous system. Such studies have been hampered by the difficulties in working with the virus and also the lack of a good animal model of VZV latency. It is known that the ganglionic VZV burden during latency is low. Two of the key questions that have been addressed are the cellular site of latent VZV and the identity of the viral genes which are transcribed during latency. There is now a consensus that latent VZV resides predominantly in ganglionic neurons with less frequent infection of non-neuronal satellite cells. There is considerable evidence to show that at least five viral genes are transcribed during latency. Unlike herpes simplex virus-1 latency, viral protein expression has been demonstrated during VZV latency. A precise knowledge of which viral genes are expressed is crucial in devising novel antiviral therapy using expressed genes as therapeutic targets. Whether gene expression at both the transcriptional and translational levels is more extensive than currently reported will require much more work and probably new molecular technology.
Collapse
Affiliation(s)
- Peter G E Kennedy
- Glasgow University Department of Neurology, Institute of Neurological Sciences, Southern General Hospital, Glasgow G51 4TF, Scotland, UK.
| |
Collapse
|
40
|
Cohrs RJ, Wischer J, Essman C, Gilden DH. Characterization of varicella-zoster virus gene 21 and 29 proteins in infected cells. J Virol 2002; 76:7228-38. [PMID: 12072522 PMCID: PMC136324 DOI: 10.1128/jvi.76.14.7228-7238.2002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Varicella-zoster virus (VZV) transcription is limited in latently infected human ganglia. Note that much of the transcriptional capacity of the virus genome has not been analyzed in detail; to date, only VZV genes mapping to open reading frames (ORFs) 4, 21, 29, 62, and 63 have been detected. ORF 62 encodes the major immediate-early virus transcription transactivator IE62, ORF 29 encodes the major virus DNA binding protein, and ORF 21 encodes a protein associated with the developing virus nucleocapsid. We analyzed the cellular location of proteins encoded by ORF 21 (21p) and ORF 29 (29p), their phosphorylation state during productive infection, and their ability form a protein-protein complex. The locations of both 21p and 29p within infected cells mimic those of their herpes simplex virus type 1 (HSV-1) homologues (UL37 and ICP8); however, unlike these homologues, 21p is not phosphorylated and neither 21p nor 29p exhibits a protein-protein interaction. Transient transfection assays to determine the effect of 21p and 29p on transcription from VZV gene 20, 21, 28, and 29 promoters revealed no significant activation of transcription by 21p or 29p from any of the VZV gene promoters tested, and 21p did not significantly modulate the ability of IE62 to activate gene transcription. A modest increase in IE62-induced activation of gene 28 and 29 promoters was seen in the presence of 29p; however, IE62-induced activation of gene 28 and 29 promoters was reduced in the presence of 21p. A Saccharomyces cerevisiae two-hybrid analysis of 21p indicated that the protein can activate transcription when tethered within a responsive promoter. Together, the data reveal that while VZV gene 21 and HSV-1 UL37 share homology at the nucleic acid level, these proteins differ functionally.
Collapse
Affiliation(s)
- Randall J Cohrs
- Department of Neurology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | | | | | |
Collapse
|
41
|
White TM, Mahalingam R, Traina-Dorge V, Gilden DH. Simian varicella virus DNA is present and transcribed months after experimental infection of adult African green monkeys. J Neurovirol 2002; 8:191-203. [PMID: 12053274 DOI: 10.1080/13550280290049705] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
To study the pathogenesis of simian varicella virus (SVV) infection in its natural primate host, we inoculated adult SVV-seronegative African green monkeys intratracheally with 10(3)-10(4) PFU of SVV, sacrificed them 11 days, 2, 5, 10, and 12 months postinfection (p.i.), and examined lung, liver, and ganglia for SVV DNA and RNA. PCR analysis revealed SVV DNA in ganglia and viscera at 11 days and 2, 5, and 10 months p.i. Similarly, SVV transcripts corresponding to immediate early (IE), putative early (E), and late (L) SVV open-reading frames (ORFs) were found in liver, lung, and ganglia of most monkeys at multiple intervals for the 12-month study period. SVV-specific antigens were detected in ganglia and liver during acute varicella, but not in ganglia 12 months p.i. Analysis of control tissue (ganglia, lung, and liver) from uninfected SVV-seronegative adult African green monkeys did not reveal SVV DNA, SVV RNA, SVV-specific antigen, or varicella-specific pathological changes. Overall, intratracheal inoculation of SVV in African green monkeys resulted in the presence of viral DNA and transcription of multiple viral genes in many tissues for months after experimental infection.
Collapse
Affiliation(s)
- Tiffany M White
- Department of Neurology, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | | | |
Collapse
|
42
|
Cai GY, Pizer LI, Levin MJ. Fractionation of neurons and satellite cells from human sensory ganglia in order to study herpesvirus latency. J Virol Methods 2002; 104:21-32. [PMID: 12020789 DOI: 10.1016/s0166-0934(02)00032-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
A method is described for fractionating human trigeminal ganglia into highly purified populations of neurons and satellite cells in order to study alpha-herpesvirus latency. The method was validated by microscopy of the separated populations and by the observation that only the neuronal population, not the satellite cells, contained herpes simplex virus (HSV) DNA. The frequency of detecting HSV in neurons from ganglia was 3% (43 of 1440 neurons). HSV DNA was not detected in approximately 17,500 satellite cells. The HSV DNA genome copy number in single cells ranged from 2 to 50. These data on the frequency and cellular location of latent HSV indicate that our mechanical fractionation of cell types results in low levels of cross-contamination and provides samples from which cells infected latently can be studied at the single cell level.
Collapse
Affiliation(s)
- Guang-Yun Cai
- Department of Pediatrics, Section of Pediatric Infectious Diseases, School of Medicine, University of Colorado, Box C-227, 4200 East Ninth Avenue, Denver, CO 80262, USA
| | | | | |
Collapse
|
43
|
Kleinschmidt-DeMasters BK, DeBiasi RL, Tyler KL. Polymerase chain reaction as a diagnostic adjunct in herpesvirus infections of the nervous system. Brain Pathol 2001; 11:452-64. [PMID: 11556691 PMCID: PMC8098446 DOI: 10.1111/j.1750-3639.2001.tb00414.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Polymerase chain reaction (PCR) is a powerful technique that allows detection of minute quantities of DNA or RNA in cerebrospinal fluid (CSF), vesicle and endoneurial fluids, blood, fresh-frozen, and even formalin-fixed tissues. Various infectious agents can be detected with high specificity and sensitivity, including bacteria, parasites, rickettsia and viruses. PCR analysis of CSF has revolutionized the diagnosis of nervous system viral infections, particularly those caused by human herpesviruses (HHV), and has now replaced brain biopsy as the gold standard for diagnosis of herpes simplex virus (HSV) encephalitis. PCR analysis of both CSF and nervous system tissues has also broadened our understanding of the spectrum of disease caused by HSV-1 and -2, cytomegalovirus (CMV), Epstein-Barr virus (EBV), varicella zoster virus (VZV), and HHV-6. Nonetheless, positive tissue PCR results must be interpreted cautiously, especially in cases that lack corroborating clinical and neuropathologic evidence of infection. Moreover, positive PCR results from tissues do not distinguish latent from productive (lytic) viral infections. In several neurological diseases, negative PCR results have provided strong evidence against a role for herpesviruses as the causative agents. This review focuses on the use of PCR tests to diagnose HSV and VZV infections of the nervous system.
Collapse
Affiliation(s)
- B K Kleinschmidt-DeMasters
- Department of Pathology, University of Colorado Health Sciences Center and The Denver Veterans Administration Hospital, 80262, USA.
| | | | | |
Collapse
|
44
|
Kleinschmidt-DeMasters BK, Gilden DH. Varicella-Zoster virus infections of the nervous system: clinical and pathologic correlates. Arch Pathol Lab Med 2001; 125:770-80. [PMID: 11371229 DOI: 10.5858/2001-125-0770-vzviot] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Diseases that present with protean manifestations are the diseases most likely to pose diagnostic challenges for both clinicians and pathologists. Among the most diverse disorders caused by a single known toxic, metabolic, neoplastic, or infectious agent are the central and peripheral nervous system complications of varicella-zoster virus (VZV). METHODS The pathologic correlates of the neurologic complications of VZV infection, as well as current methods for detecting viral infections, are discussed and presented in pictorial format for the practicing pathologist. RESULTS Varicella-zoster virus causes chickenpox (varicella), usually in childhood; most children manifest only mild neurologic sequelae. After chickenpox resolves, the virus becomes latent in neurons of cranial and spinal ganglia of nearly all individuals. In elderly and immunocompromised individuals, the virus may reactivate to produce shingles (zoster). After zoster resolves, many elderly patients experience postherpetic neuralgia. Uncommonly, VZV can spread to large cerebral arteries to cause a spectrum of large-vessel vascular damage, ranging from vasculopathy to vasculitis, with stroke. In immunocompromised individuals, especially those with cancer or acquired immunodeficiency syndrome, deeper tissue penetration of the virus may occur (as compared with immunocompetent individuals), with resultant myelitis, small-vessel vasculopathy, ventriculitis, and meningoencephalitis. Detection of the virus in neurons, oligodendrocytes, meningeal cells, ependymal cells, or the blood vessel wall often requires a combination of morphologic, immunohistochemical, in situ hybridization, and polymerase chain reaction (PCR) methods. The PCR analysis of cerebrospinal fluid remains the mainstay for diagnosing the neurologic complications of VZV during life. CONCLUSIONS Varicella-zoster virus infects a wide variety of cell types in the central and peripheral nervous system, explaining the diversity of clinical disorders associated with the virus.
Collapse
|
45
|
Kress M, Fickenscher H. Infection by human varicella‐zoster virus confers norepinephrine sensitivity to sensory neurons from rat dorsal root ganglia. FASEB J 2001. [DOI: 10.1096/fsb2fj000440com] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michaela Kress
- Institut für Physiologie und Experimentelle Pathophysiologie, and Institut für Klinische und Molekulare Virologie, Friedrich‐Alexander‐Universitaat Erlangen‐Nürnberg D‐91054 Erlangen Germany
| | - Helmut Fickenscher
- Institut für Physiologie und Experimentelle Pathophysiologie, and Institut für Klinische und Molekulare Virologie, Friedrich‐Alexander‐Universitaat Erlangen‐Nürnberg D‐91054 Erlangen Germany
| |
Collapse
|
46
|
Kress M, Fickenscher H. Infection by human varicella-zoster virus confers norepinephrine sensitivity to sensory neurons from rat dorsal root ganglia. FASEB J 2001; 15:1037-43. [PMID: 11292665 DOI: 10.1096/fj.00-0440com] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Varicella-zoster virus (VZV) is a widespread human herpes virus causing chicken pox on primary infection and persisting in sensory neurons. Reactivation causes shingles, which are characterized by severe pain and often lead to postherpetic neuralgia. To elucidate the mechanisms of VZV-associated hyperalgesia, we elaborated an in vitro model for the VZV infection of sensory neurons from rat dorsal root ganglia. Between 35 and 50% of the neurons showed strong expression of the immediate-early virus antigens IE62 and IE63 and the late glycoprotein gE. When the intracellular calcium concentration was monitored microfluorometrically for individual cells after infection, the sensitivity to GABA or capsaicin was similar in controls and in VZV-infected neurons. However, the baseline calcium concentration was increased. Neurons became de novo sensitive to adrenergic stimulation after VZV infection. Norepinephrine-responsive neurons were more frequent and calcium responses to norepinephrine were significantly higher after infection with wild-type isolates than with the attenuated vaccine strain OKA. The adrenergic agonists phenylephrine and isoproterenol had similar efficacy. We suggest that the infection with wild-type VZV isolates confers norepinephrine sensitivity to sensory neurons by using alpha(1)- and/or beta(1)-adrenergic receptors providing a model for the pathophysiology of the severe pain associated with the acute reactivation of VZV.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Coculture Techniques
- Female
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/pathology
- Ganglia, Spinal/virology
- Herpes Zoster/pathology
- Herpesvirus 3, Human
- Humans
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Neurons, Afferent/virology
- Norepinephrine/pharmacology
- Rats
- Rats, Wistar
- Receptors, Adrenergic/metabolism
Collapse
Affiliation(s)
- M Kress
- Institut für Physiologie und Experimentelle Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| | | |
Collapse
|
47
|
Kennedy PG, Grinfeld E, Bell JE. Varicella-zoster virus gene expression in latently infected and explanted human ganglia. J Virol 2000; 74:11893-8. [PMID: 11090189 PMCID: PMC112472 DOI: 10.1128/jvi.74.24.11893-11898.2000] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A consistent feature of varicella-zoster virus (VZV) latency is the restricted pattern of viral gene expression in human ganglionic tissues. To understand further the significance of this gene restriction, we used in situ hybridization (ISH) to detect the frequency of RNA expression for nine VZV genes in trigeminal ganglia (TG) from 35 human subjects, including 18 who were human immunodeficiency virus (HIV) positive. RNA for VZV gene 21 was detected in 7 of 11 normal and 6 of 10 HIV-positive subjects, RNA for gene 29 was detected in 5 of 14 normal and 11 of 11 HIV-positive subjects, RNA for gene 62 was detected in 4 of 10 normal and 6 of 9 HIV-positive subjects, and RNA for gene 63 was detected in 8 of 17 normal and 12 of 15 HIV-positive subjects. RNA for VZV gene 4 was detected in 2 of 13 normal and 4 of 9 HIV-positive subjects, and RNA for gene 18 was detected in 4 of 15 normal and 5 of 15 HIV-positive subjects. By contrast, RNAs for VZV genes 28, 40, and 61 were rarely or never detected. In addition, immunocytochemical analysis detected the presence of VZV gene 63-encoded protein in five normal and four HIV-positive subjects. VZV RNA was also analyzed in explanted fresh human TG and dorsal root ganglia from five normal human subjects over a period of up to 11 days in culture. We found a very different pattern of gene expression in these explants, with transcripts for VZV genes 18, 28, 29, 40, and 63 all frequently detected, presumably as a result of viral reactivation. Taken together, these data provide further support for the notion of significant and restricted viral gene expression in VZV latency.
Collapse
Affiliation(s)
- P G Kennedy
- Glasgow University Department of Neurology, Southern General Hospital, Glasgow, United Kingdom.
| | | | | |
Collapse
|
48
|
Cohrs RJ, Randall J, Smith J, Gilden DH, Dabrowski C, van Der Keyl H, Tal-Singer R. Analysis of individual human trigeminal ganglia for latent herpes simplex virus type 1 and varicella-zoster virus nucleic acids using real-time PCR. J Virol 2000; 74:11464-71. [PMID: 11090142 PMCID: PMC112425 DOI: 10.1128/jvi.74.24.11464-11471.2000] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) and varicella-zoster virus (VZV) establish latent infections in the peripheral nervous system following primary infection. During latency both virus genomes exhibit limited transcription, with the HSV-1 LATs and at least four VZV transcripts consistently detected in latently infected human ganglia. In this study we used real-time PCR quantitation to determine the viral DNA copy number in individual trigeminal ganglia (TG) from 17 subjects. The number of HSV-1 genomes was not significantly different between the left and right TG from the same individual and varied per subject from 42.9 to 677.9 copies per 100 ng of DNA. The number of VZV genomes was also not significantly different between left and right TG from the same individual and varied per subject from 37.0 to 3,560.5 copies per 100 ng of DNA. HSV-1 LAT transcripts were consistently detected in ganglia containing latent HSV-1 and varied in relative expression by >500-fold. Of the three VZV transcripts analyzed, only transcripts mapping to gene 63 were consistently detected in latently infected ganglia and varied in relative expression by >2,000-fold. Thus, it appears that, similar to LAT transcription in HSV-1 latently infected ganglia, VZV gene 63 transcription is a hallmark of VZV latency.
Collapse
Affiliation(s)
- R J Cohrs
- Departments of Neurology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Gilden DH, Kleinschmidt-DeMasters BK, LaGuardia JJ, Mahalingam R, Cohrs RJ. Neurologic complications of the reactivation of varicella-zoster virus. N Engl J Med 2000; 342:635-45. [PMID: 10699164 DOI: 10.1056/nejm200003023420906] [Citation(s) in RCA: 513] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- D H Gilden
- Department of Neurology, University of Colorado Health Sciences Center, Denver 80262, USA.
| | | | | | | | | |
Collapse
|