1
|
Wang J, Brückner N, Weissmann S, Günther T, Zhu S, Vogt C, Sun G, Guo R, Bruno R, Ritter B, Steinbrück L, Kaufer BB, Depledge DP, Grundhoff A, Viejo-Borbolla A. Repression of varicella zoster virus gene expression during quiescent infection in the absence of detectable histone deposition. PLoS Pathog 2025; 21:e1012367. [PMID: 39928684 DOI: 10.1371/journal.ppat.1012367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 02/19/2025] [Accepted: 01/20/2025] [Indexed: 02/12/2025] Open
Abstract
Varicella zoster virus (VZV) is a human-specific herpesvirus that establishes latency in peripheral neurons. The only transcripts detected in infected human trigeminal ganglia (TG) obtained shortly after death correspond to the VZV latency-associated transcript (VLT) and associated VLT-ORF63 splice variants. In vitro studies showed that VLT-ORF63 is translated into a protein (pVLT-ORF63) that induces VZV transcription. The mechanisms that lead to this restricted gene expression and the transition to lytic replication remain unknown, partly due to the difficulty of working with human neurons. In this study, we addressed whether the neuroblastoma-derived cell line SH-SY5Y could serve as a model to investigate the mechanisms that lead to repression of VZV gene expression followed by reactivation. VZV productively infected differentiated SH-SY5Y (dSH-SY5Y) whereas incubation with acyclovir (ACV) inhibited virus replication and induced a progressive repression of the virus. Upon removal of ACV there was production of viral particles in a subset of cells, while others contained non-replicating VZV genomes and VLT-containing transcripts for at least 20 days post-infection (dpi). Exogenous expression of VLT-ORF63 induced productive infection, suggesting that the non-replicating and repressed genomes remained functional. Interestingly, histone deposition was undetectable at VZV genomes in quiescently infected dSH-SY5Y cells, pointing to a potential novel mechanism leading to VZV repression in this neuronal setting.
Collapse
Affiliation(s)
- Jiayi Wang
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Nadine Brückner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | | | - Shuyong Zhu
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Excellence Cluster RESIST, Hannover Medical School, Hannover, Germany
| | - Carolin Vogt
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover, Germany
| | - Guorong Sun
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Rongrong Guo
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Renzo Bruno
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Birgit Ritter
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Lars Steinbrück
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Daniel P Depledge
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Excellence Cluster RESIST, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover, Germany
| | | | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Excellence Cluster RESIST, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Onisiforou A, Charalambous EG, Zanos P. Shattering the Amyloid Illusion: The Microbial Enigma of Alzheimer's Disease Pathogenesis-From Gut Microbiota and Viruses to Brain Biofilms. Microorganisms 2025; 13:90. [PMID: 39858858 PMCID: PMC11767882 DOI: 10.3390/microorganisms13010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
For decades, Alzheimer's Disease (AD) research has focused on the amyloid cascade hypothesis, which identifies amyloid-beta (Aβ) as the primary driver of the disease. However, the consistent failure of Aβ-targeted therapies to demonstrate efficacy, coupled with significant safety concerns, underscores the need to rethink our approach to AD treatment. Emerging evidence points to microbial infections as environmental factors in AD pathoetiology. Although a definitive causal link remains unestablished, the collective evidence is compelling. This review explores unconventional perspectives and emerging paradigms regarding microbial involvement in AD pathogenesis, emphasizing the gut-brain axis, brain biofilms, the oral microbiome, and viral infections. Transgenic mouse models show that gut microbiota dysregulation precedes brain Aβ accumulation, emphasizing gut-brain signaling pathways. Viral infections like Herpes Simplex Virus Type 1 (HSV-1) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) may lead to AD by modulating host processes like the immune system. Aβ peptide's antimicrobial function as a response to microbial infection might inadvertently promote AD. We discuss potential microbiome-based therapies as promising strategies for managing and potentially preventing AD progression. Fecal microbiota transplantation (FMT) restores gut microbial balance, reduces Aβ accumulation, and improves cognition in preclinical models. Probiotics and prebiotics reduce neuroinflammation and Aβ plaques, while antiviral therapies targeting HSV-1 and vaccines like the shingles vaccine show potential to mitigate AD pathology. Developing effective treatments requires standardized methods to identify and measure microbial infections in AD patients, enabling personalized therapies that address individual microbial contributions to AD pathogenesis. Further research is needed to clarify the interactions between microbes and Aβ, explore bacterial and viral interplay, and understand their broader effects on host processes to translate these insights into clinical interventions.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| | - Eleftheria G. Charalambous
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 1–2, Ellernholzstr., 17489 Greifswald, Germany
| | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| |
Collapse
|
3
|
Srikanth P, Arumugam I, Jeganathan SN, Ramesh R, Ranganathan LN, Vijayaraghavan S. Expanded spectrum of varicella disease and the need for vaccination in India. Hum Vaccin Immunother 2024; 20:2328955. [PMID: 38517089 PMCID: PMC10962579 DOI: 10.1080/21645515.2024.2328955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Varicella vaccine was first licensed in Japan and South Korea in 1989 for use in healthy children and was introduced in US in 1995. So far, 29 countries have adopted varicella vaccine in their universal immunization program (UIP). No Asian country, India included, has adopted the varicella vaccine as part of their UIP. The extra-cutaneous sites for VZV diseases are central nervous system and gastrointestinal tract, the expanded disease spectrum includes vasculopathy, myelitis, inflammatory bowel disease, perforated ulcers, and gastritis. The actual disease burden of varicella is not known as most of the infected individuals may not visit the physician. The amplifiable VZV DNA will not always be detectable in cerebrospinal fluid (CSF) samples in protracted illnesses such as vasculopathies, but demonstrable anti-VZV IgG in CSF has diagnostic value. The World Health Organization (WHO) position paper 2014 recommends two doses of varicella and zoster vaccines in targeted population. In India, varicella vaccine is not included in the UIP due to the cost and the belief that lifelong immunity occurs following primary infection. The expanded spectrum of VZV disease and the mounting body of evidence, however, suggest the need for both varicella and zoster vaccines in routine immunization schedule.
Collapse
Affiliation(s)
- Padma Srikanth
- Department of Microbiology, Sri Ramachandra Medical College and Research Institute (SRIHER, DU), Chennai, India
| | - Ilakkiya Arumugam
- Department of Microbiology, Sri Ramachandra Medical College and Research Institute (SRIHER, DU), Chennai, India
| | - Seetha N. Jeganathan
- Department of Microbiology, Sri Ramachandra Medical College and Research Institute (SRIHER, DU), Chennai, India
| | - Rithvik Ramesh
- Department of Neurology, Sri Ramachandra Medical College and Research Institute (SRIHER, DU), Chennai, India
| | | | - Shanthi Vijayaraghavan
- Department of Hepatology and Medical Gastroenterology, Sri Ramachandra Medical College and Research Institute (SRIHER, DU), Chennai, India
| |
Collapse
|
4
|
Zhong W, Lan C, Chen Y, Song K, Ma Z, Zeng J, Huang L, Zhang Y, Zhu Y, Xia H. Virus-Triggered Autoimmunity Was Associated With Hirschsprung's Disease Through Activation of Innate Immunity. J Immunol Res 2024; 2024:4838514. [PMID: 39493374 PMCID: PMC11531361 DOI: 10.1155/2024/4838514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 08/06/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
Background: Hirschsprung's disease (HSCR) is a congenital enteric nervous system (ENS) disorder. Genetics cannot explain most sporadic cases. To explore the relationship between pathogen infection, autoantibodies, innate immune, and HSCR. Methods: Pathogen microarray was conducted in the serum of the prospective neonatal abdominal distension (NAD) cohort, consisting of 56 children followed for at least 6 months until the final diagnosis of HSCR was determined or excluded. We conducted an autoantibody microarray in an HSCR cohort, which is comprised of diagnosed HSCR patients (HSCR) and healthy control subjects (HC). RNA-seq of colon tissues from aganglionic and ganglionic segments of HSCR patients was performed. Results: Experimental results show that the serum lgM and lgG of enterovirus 71 (EV71) were significantly higher in HSCR than in the gastrointestinal dysfunction (GI) group, with a prediagnose value reaching area under the curve (AUC) over 0.76. We discovered that a group of autoantibodies were significantly higher in HSCR including neuronal pentraxin 1 (NPTX1), amyloid, neuron lysate, and myelin-associated oligodendrocytic basic protein (MOBP) than that in the HC group. These four autoantibodies could distinguish HSCR from the HC group, with a combined AUC of over 0.90 using both serum IgG and IgM. Further analysis showed that wide activation of innate immune pathways, including toll-like receptor (TLR) signaling pathway, neutrophil-to-lymphocyte ratio (NLR) signaling pathway, red cell distribution width to lymphocyte ratio (RLR) signaling pathway, and cyclic adenosine monophosphate (cAMP) signaling pathway in aganglionic compared to ganglionic segments of HSCR. Conclusion: This study suggested that virus-triggered autoimmunity may contribute to HSCR through activation of innate immunity, which facilitates the diagnosis and prevention of HSCR.
Collapse
Affiliation(s)
- Weiyong Zhong
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Chaoting Lan
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yuqiong Chen
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Kai Song
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Zuyi Ma
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jixiao Zeng
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Lihua Huang
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yan Zhang
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yun Zhu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| |
Collapse
|
5
|
Wu J, Mao K, Zhang R, Fu Y. Extracellular vesicles in the pathogenesis of neurotropic viruses. Microb Pathog 2024; 195:106901. [PMID: 39218378 DOI: 10.1016/j.micpath.2024.106901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Neurotropic viruses, characterized by their capacity to invade the central nervous system, present a considerable challenge to public health and are responsible for a diverse range of neurological disorders. This group includes a diverse array of viruses, such as herpes simplex virus, varicella zoster virus, poliovirus, enterovirus and Japanese encephalitis virus, among others. Some of these viruses exhibit high neuroinvasiveness and neurovirulence, while others demonstrate weaker neuroinvasive and neurovirulent properties. The clinical manifestations of infections caused by neurotropic viruses can vary significantly, ranging from mild symptoms to severe life-threatening conditions. Extracellular vesicles (EVs) have garnered considerable attention due to their pivotal role in intracellular communication, which modulates the biological activity of target cells via the transport of biomolecules in both health and disease. Investigating EVs in the context of virus infection is crucial for elucidating their potential role contribution to viral pathogenesis. This is because EVs derived from virus-infected cells frequently transfer viral components to uninfected cells. Importantly, EVs released by virus-infected cells have the capacity to traverse the blood-brain barrier (BBB), thereby impacting neuronal activity and inducing neuroinflammation. In this review, we explore the roles of EVs during neurotropic virus infections in either enhancing or inhibiting viral pathogenesis. We will delve into our current comprehension of the molecular mechanisms that underpin these roles, the potential implications for the infected host, and the prospective diagnostic applications that could arise from this understanding.
Collapse
Affiliation(s)
- Junyi Wu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Kedan Mao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Rui Zhang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China.
| | - Yuxuan Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
6
|
Hu Y, Zhong M, Hu M, Zhang L. Varicella‑zoster virus‑associated meningitis followed peripheral facial palsy: A case report. Exp Ther Med 2024; 28:380. [PMID: 39113905 PMCID: PMC11304175 DOI: 10.3892/etm.2024.12669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
Although central nervous system infection following varicella zoster virus infection is relatively common, subsequent peripheral nervous system infection is comparatively rare. The present case documents a case of meningitis after varicella-zoster virus (VZV) infection, which was then followed by peripheral facial palsy. Specifically, a 54-year-old female patient was first admitted to Shengli Oilfield Central Hospital (Dongying, China) with headache and fever. Physical examination revealed herpes that formed along the intercostal nerve in the left forebreast, armpit and back. Subsequently, neurological examination found cervical resistance in more than three fingers (neck resistance of less than two transverse fingers is not evidence of meningeal irritation; the neck resistance of this patient was approximately three transverse fingers, so the patient was presumed to be positive for meningeal irritation, highly suggestive of meningitis) and Kernig sign was positive. There were no significant abnormalities according to brain MRI and lumbar puncture pressure was 330 mmH2O. In addition, the leukocyte count was 734x106/l, 50% monocyte count, 50% multinucleated cells, chloride levels of 109.1 mmol/l, protein levels of 235 mg/dl and glucose levels of 4.18 mmol/l in the cerebrospinal fluid. DNA and RNA metagenomic detection of pathogenic microorganisms in the cerebrospinal fluid revealed the presence of VZV. The patient was therefore treated with acyclovir, ceftriaxone, mannitol and methylprednisolone, but then developed right peripheral facial palsy at 10 days after treatment. This complication was not found in the literature, and the occurrence of facial neuritis was unexpected. The active period of VZV virus was 21 days, and the patient had herpes 5 days before admission. The active period of the virus was considered to have subsided and the patient was in the recovery period. Moreover, the results of lumbar puncture showed that the white blood cells, the proportion of neutrophils and the protein in cerebrospinal fluid were all decreasing, which also indicated that the patient had entered the recovery period. The patient was discharged 18 days after admission. In conclusion, observations from the present case suggested that the clinical manifestations of VZV infection can be complex and varied, requiring the clinician to have an accurate understanding of its disease progression and treatment.
Collapse
Affiliation(s)
- Yaozhi Hu
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying, Shandong 257000, P.R. China
| | - Mengfei Zhong
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying, Shandong 257000, P.R. China
| | - Mengliang Hu
- Department of Neurology, Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Ligong Zhang
- Department of Neurology, Shengli Oilfield Central Hospital, Dongying, Shandong 257000, P.R. China
| |
Collapse
|
7
|
Wang H, Chen G, Gong Q, Wu J, Chen P. Primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome: a Mendelian randomization study. Front Immunol 2024; 15:1403429. [PMID: 39253091 PMCID: PMC11381235 DOI: 10.3389/fimmu.2024.1403429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Background Currently, evidence regarding the causal relationship between primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome is limited and inconsistent. Therefore, this study employs Mendelian randomization (MR) methodology to investigate the causal relationship between the two. Methods This study selected 110 single-nucleotide polymorphisms (SNPs) of primary immunodeficiency-related genes as instrumental variables (IVs). Genetic associations of primary immunodeficiency-related genes were derived from recent genome-wide association studies (GWAS) data on human plasma protein levels and circulating immune cells. Data on genes associated with varicella-zoster virus reactivation syndrome were obtained from the GWAS Catalog and FINNGEN database, primarily analyzed using inverse variance weighting (IVW) and sensitivity analysis. Results Through MR analysis, we identified 9 primary immunodeficiency-related genes causally associated with herpes zoster and its subsequent neuralgia; determined causal associations of 20 primary immunodeficiency-related genes with three vascular lesions (stroke, cerebral aneurysm, giant cell arteritis); revealed causal associations of 10 primary immunodeficiency-related genes with two ocular diseases (retinopathy, keratitis); additionally, three primary immunodeficiency-related genes each were associated with encephalitis, cranial nerve palsy, and gastrointestinal infections. Conclusions This study discovers a certain association between primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome, yet further investigations are warranted to explore the specific mechanisms underlying these connections.
Collapse
Affiliation(s)
- Hao Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Guanglei Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Qian Gong
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jing Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Peng Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
8
|
Hakami MA, Khan FR, Abdulaziz O, Alshaghdali K, Hazazi A, Aleissi AF, Abalkhail A, Alotaibi BS, Alhazmi AYM, Kukreti N, Binshaya AS. Varicella-zoster virus-related neurological complications: From infection to immunomodulatory therapies. Rev Med Virol 2024; 34:e2554. [PMID: 38862398 DOI: 10.1002/rmv.2554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 06/13/2024]
Abstract
The Varicella-zoster virus (VZV), classified as a neurotropic member of the Herpesviridae family, exhibits a characteristic pathogenicity, predominantly inducing varicella, commonly known as chickenpox, during the initial infectious phase, and triggering the reactivation of herpes zoster, more commonly recognized as shingles, following its emergence from a latent state. The pathogenesis of VZV-associated neuroinflammation involves a complex interplay between viral replication within sensory ganglia and immune-mediated responses that contribute to tissue damage and dysfunction. Upon primary infection, VZV gains access to sensory ganglia, establishing latent infection within neurons. During reactivation, the virus can spread along sensory nerves, triggering a cascade of inflammatory mediators, chemokines, and immune cell infiltration in the affected neural tissues. The role of both adaptive and innate immune reactions, including the contributions of T and B cells, macrophages, and dendritic cells, in orchestrating the immune-mediated damage in the central nervous system is elucidated. Furthermore, the aberrant activation of the natural defence mechanism, characterised by the dysregulated production of immunomodulatory proteins and chemokines, has been implicated in the pathogenesis of VZV-induced neurological disorders, such as encephalitis, myelitis, and vasculopathy. The intricate balance between protective and detrimental immune responses in the context of VZV infection emphasises the necessity for an exhaustive comprehension of the immunopathogenic mechanisms propelling neuroinflammatory processes. Despite the availability of vaccines and antiviral therapies, VZV-related neurological complications remain a significant concern, particularly in immunocompromised individuals and the elderly. Elucidating these mechanisms might facilitate the emergence of innovative immunomodulatory strategies and targeted therapies aimed at mitigating VZV-induced neuroinflammatory damage and improving clinical outcomes. This comprehensive understanding enhances our grasp of viral pathogenesis and holds promise for pioneering therapeutic strategies designed to mitigate the neurological ramifications of VZV infections.
Collapse
Affiliation(s)
- Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Farhan R Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Osama Abdulaziz
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Makkah Province, Saudi Arabia
| | - Khalid Alshaghdali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Awad F Aleissi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Public Health and Health Informatics, Qassim University, Buraydah, Qassim, Saudi Arabia
| | - Bader S Alotaibi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | | | - Neelima Kukreti
- Graphic Era Hill University, Clement Town, Dehradun, India
- Graphic Era (Deemed to be University), Clement Town, Dehradun, India
| | - Abdulkarim S Binshaya
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
9
|
Singal A, Schwartz RA, Bhate C. Herpes zoster infection in pregnancy: features and consequences. Arch Dermatol Res 2024; 316:107. [PMID: 38489022 DOI: 10.1007/s00403-024-02842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/17/2024]
Abstract
Herpes (varicella) zoster (HZ) infection occurs in 4 people per 1000 in the general US population (irrespective of prior varicella infection and vaccination status) each year and has been the subject of scientific inquiry for decades. The consequences of infection are myriad and may depend on the dermatome of involvement as well as host factors such as age, comorbidities, prior treatment or immunization, and immunologic status. Pregnancy is associated with an altered immune and hormonal status in the mother. While maternal HZ infection during pregnancy is not uncommon, the implications for both mother and child are not well established, although multiple studies of perinatal maternal HZ infection suggest no intrauterine transmission to the fetus. We review the current literature on herpes zoster infection in pregnancy, including epidemiology, diagnosis, potential immunologic sequelae, and strategies for prevention and treatment.
Collapse
Affiliation(s)
- Amit Singal
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Robert A Schwartz
- Dermatology and Pathology, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ, 07103, USA.
| | - Chinmoy Bhate
- Dermatology and Pathology, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ, 07103, USA
| |
Collapse
|
10
|
Li Y, Chen Q, Wang L, Chen X, Wang B, Zhong W. The mechanisms of nerve injury caused by viral infection in the occurrence of gastrointestinal motility disorder-related diseases. Virol J 2023; 20:251. [PMID: 37915051 PMCID: PMC10621196 DOI: 10.1186/s12985-023-02185-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/14/2023] [Indexed: 11/03/2023] Open
Abstract
Gastrointestinal motility refers to the peristalsis and contractility of gastrointestinal muscles, including the force and frequency of gastrointestinal muscle contraction. Gastrointestinal motility maintains the normal digestive function of the human body and is a critical component of the physiological function of the digestive tract. At present, gastrointestinal motility disorder-related diseases are gradually affecting human production and life. In recent years, it has been consistently reported that the enteric nervous system has a coordinating and controlling role in gastrointestinal motility. Motility disorders are closely related to functional or anatomical changes in the gastrointestinal nervous system. At the same time, some viral infections, such as herpes simplex virus and varicella-zoster virus infections, can cause damage to the gastrointestinal nervous system. Therefore, this paper describes the mechanisms of viral infection in the gastrointestinal nervous system and the associated clinical manifestations. Studies have indicated that the means by which viruses can cause the infection of the enteric nervous system are various, including retrograde transport, hematogenous transmission and centrifugal transmission from the central nervous system. When viruses infect the enteric nervous system, they can cause clinical symptoms, such as abdominal pain, abdominal distension, early satiation, belching, diarrhea, and constipation, by recruiting macrophages, lymphocytes and neutrophils and regulating intestinal microbes. The findings of several case‒control studies suggest that viruses are the cause of some gastrointestinal motility disorders. It is concluded that one of the causes of gastrointestinal motility disorders is viral infection of the enteric nervous system. In such disorders, the relationships between viruses and nerves remain to be studied more deeply. Further studies are necessary to evaluate whether prophylactic antiviral therapy is feasible in gastrointestinal motility disorders.
Collapse
Affiliation(s)
- Yaqian Li
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiuyu Chen
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Gastroenterology, Tianjin First Central Hospital, Tianjin, 300110, China
| | - Liwei Wang
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
11
|
Jin X, Wang W, Zhao X, Jiang W, Shao Q, Chen Z, Huang C. The battle between the innate immune cGAS-STING signaling pathway and human herpesvirus infection. Front Immunol 2023; 14:1235590. [PMID: 37600809 PMCID: PMC10433641 DOI: 10.3389/fimmu.2023.1235590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
The incidence of human herpesvirus (HHVs) is gradually increasing and has affected a wide range of population. HHVs can result in serious consequences such as tumors, neonatal malformations, sexually transmitted diseases, as well as pose an immense threat to the human health. The cGAS-STING pathway is one of the innate immune pattern-recognition receptors discovered recently. This article discusses the role of the cGAS-STING pathway in human diseases, especially in human herpesvirus infections, as well as highlights how these viruses act on this pathway to evade the host immunity. Moreover, the author provides a comprehensive overview of modulators of the cGAS-STING pathway. By focusing on the small molecule compounds based on the cGAS-STING pathway, novel targets and concepts have been proposed for the development of antiviral drugs and vaccines, while also providing a reference for the investigation of disease models related to the cGAS-STING pathway. HHV is a double-stranded DNA virus that can trigger the activation of intracellular DNA sensor cGAS, after which the host cells initiate a cascade of reactions that culminate in the secretion of type I interferon to restrict the viral replication. Meanwhile, the viral protein can interact with various molecules in the cGAS-STING pathway. Viruses can evade immune surveillance and maintain their replication by inhibiting the enzyme activity of cGAS and reducing the phosphorylation levels of STING, TBK1 and IRF3 and suppressing the interferon gene activation. Activators and inhibitors of the cGAS-STING pathway have yielded numerous promising research findings in vitro and in vivo pertaining to cGAS/STING-related disease models. However, there remains a dearth of small molecule modulators that have been successfully translated into clinical applications, which serves as a hurdle to be overcome in the future.
Collapse
Affiliation(s)
- Ximing Jin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjia Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinwei Zhao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhua Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Shao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
13
|
The Potential Role of Microorganisms on Enteric Nervous System Development and Disease. Biomolecules 2023; 13:biom13030447. [PMID: 36979382 PMCID: PMC10046024 DOI: 10.3390/biom13030447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/14/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The enteric nervous system (ENS), the inherent nervous system of the gastrointestinal (GI) tract is a vast nervous system that controls key GI functions, including motility. It functions at a critical interface between the gut luminal contents, including the diverse population of microorganisms deemed the microbiota, as well as the autonomic and central nervous systems. Critical development of this axis of interaction, a key determinant of human health and disease, appears to occur most significantly during early life and childhood, from the pre-natal through to the post-natal period. These factors that enable the ENS to function as a master regulator also make it vulnerable to damage and, in turn, a number of GI motility disorders. Increasing attention is now being paid to the potential of disruption of the microbiota and pathogenic microorganisms in the potential aetiopathogeneis of GI motility disorders in children. This article explores the evidence regarding the relationship between the development and integrity of the ENS and the potential for such factors, notably dysbiosis and pathogenic bacteria, viruses and parasites, to impact upon them in early life.
Collapse
|
14
|
Yuan Y, Zhang Y, Wang J, Liu H, Zhang H, Yan Y. Immune changes and their relationship with prognosis in patients with varicella-zoster virus encephalitis/meningitis. Am J Transl Res 2023; 15:1421-1429. [PMID: 36915759 PMCID: PMC10006762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/05/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVES This study aimed to investigate the immune changes in patients with varicella-zoster virus (VZV) encephalitis/meningitis and explored their relationships with prognosis. METHODS A total of 129 patients with herpes zoster (HZ), 32 patients with VZV encephalitis/meningitis and 31 non-HZ and non-VZV people as healthy controls were included into the present study. The numbers of peripheral T lymphocytes and the serum levels of complements 3 (C3), complements 4 (C4) and immunoglobulin A (IgA), immunoglobulin G (IgG), and immunoglobulin M (IgM) were detected and compared among groups. In 32 patients with VZV encephalitis/meningitis, the immune related variables were compared between the favorable and the unfavorable prognosis group and their relationships with prognosis were further evaluated. RESULTS There were marked differences in the peripheral CD3+, CD4+ and CD8+ cells and CD4+/CD8+ ratio in the three groups (P<0.05). As compared with HZ and control groups, the peripheral CD3+ and CD4+ cells were reduced dramatically in patients with VZV encephalitis/meningitis (P<0.05). In 32 patients with VZV encephalitis/meningitis, the absolute CD3+ and CD4+ cells in patients with favorable prognosis were significantly higher than in patients with unfavorable prognosis (P<0.05), and they were positively related to the prognosis of these patients (r=0.3852, P=0.0295; r=0.3719, P=0.0361). CONCLUSION These immune changes were compromised in VZV encephalitis/meningitis. The peripheral CD3+ and CD4+ levels may be employed to predict prognosis.
Collapse
Affiliation(s)
- Yanrong Yuan
- Department of Neurology, The Third People's Hospital of Hangzhou Hangzhou, Zhejiang, China
| | - Yan Zhang
- Department of Neurology, The Third People's Hospital of Hangzhou Hangzhou, Zhejiang, China
| | - Jun Wang
- Department of Neurology, The Third People's Hospital of Hangzhou Hangzhou, Zhejiang, China
| | - Huili Liu
- Department of Neurology, The Third People's Hospital of Hangzhou Hangzhou, Zhejiang, China
| | - Hua Zhang
- Department of Anesthesiology and Pain Management, The Third People's Hospital of Hangzhou Hangzhou, Zhejiang, China
| | - Yongxing Yan
- Department of Neurology, The Third People's Hospital of Hangzhou Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Gauthier Y, Lepreux S, Cario‐Andre M, Rambert J, Dakdaki A, Lafon M, Abouqal R, Benzekri L. Varicella-zoster virus in actively spreading segmental vitiligo skin: Pathological, immunochemical, and ultrastructural findings (a first and preliminary study). Pigment Cell Melanoma Res 2023; 36:78-85. [PMID: 36112095 PMCID: PMC10092484 DOI: 10.1111/pcmr.13064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 07/07/2022] [Accepted: 08/15/2022] [Indexed: 12/31/2022]
Abstract
Segmental vitiligo (SV) is a unilateral subtype of vitiligo which is clinically characterized by a cutaneous depigmentation and histologically by a melanocyte loss from the epidermis and hair follicle reservoirs. To date, its pathogenesis remains a mystery. In many cases, this skin depigmentation shares several clinical features and dysfunctions with herpes zoster (HZ). So, for the first time, we examined whether any nucleus and cell fusion associated with a positive immunolabelling of varicella-zoster virus (VZV) and VZV mature virions could be found in SV skin samples as in herpes zoster (HZ). A total of 40 SV samples were used for histological and immunochemical studies. Control samples were obtained from three HZ, and 10 generalized vitiligo lesions. For ultrastructural study, three recent SV and one HZ as controls were recruited. Here, we report that nuclear fusion in epidermal cells were statistically associated with recent SV (p < .001), whereas syncytia formation was associated with long-lasting SV (p = .001). A positive detection of VZV antigen was statistically associated in the epidermis with recent SV and in the dermis with long-lasting SV (p = .001). Finally, the discovery of mature virions in 3/3 recent SV samples provides additional arguments for our viral hypothesis.
Collapse
Affiliation(s)
- Yvon Gauthier
- Vitiligo and Melasma Research Association (V.M.R.A.)BordeauxFrance
| | | | - Muriel Cario‐Andre
- Bordeaux University, INSERM, BRIC, U1312BordeauxFrance
- National Reference Center for Rare skin DiseasesBordeaux University HospitalBordeauxFrance
- AquidermBordeauxFrance
| | - Jérome Rambert
- National Reference Center for Rare skin DiseasesBordeaux University HospitalBordeauxFrance
| | - Adrien Dakdaki
- Tumor Bank and Tumor Biology LaboratoryCHU BordeauxPessacFrance
| | - Marie‐Edith Lafon
- Department of Virology Bordeaux University HospitalBordeaux University, CNRS, UMR5234BordeauxFrance
| | - Redouane Abouqal
- Laboratory of Biostatistics, Clinical Research and EpidemiologyMohammed V University in Rabat, Acute Medical Unit, Ibn Sina Teaching HospitalRabatMorocco
| | - Laila Benzekri
- Dermatology Department, Ibn Sina Teaching HospitalMohammed V University in Rabat, Pigmentary Disorders Outpatient ClinicRabatMorocco
| |
Collapse
|
16
|
Bougnères P, Le Fur S, Valleron AJ. Early varicella infection is associated with a delayed onset of childhood type 1 diabetes. DIABETES & METABOLISM 2022; 48:101394. [PMID: 36170944 DOI: 10.1016/j.diabet.2022.101394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Affiliation(s)
- Pierre Bougnères
- Institut National de la Santé et de la Recherche Médicale (U1195), Le Kremlin-Bicêtre, France; Paris-Saclay University, France.
| | - Sophie Le Fur
- Institut National de la Santé et de la Recherche Médicale (U1195), Le Kremlin-Bicêtre, France
| | - Alain-Jacques Valleron
- Institut National de la Santé et de la Recherche Médicale (U1195), Le Kremlin-Bicêtre, France
| |
Collapse
|
17
|
Sakakibara R, Sawai S, Ogata T. Varicella-zoster virus infection and autonomic dysfunction. Auton Neurosci 2022; 242:103018. [PMID: 35863181 DOI: 10.1016/j.autneu.2022.103018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/18/2022] [Accepted: 07/05/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND AND PURPOSE Autonomic dysfunction has been occasionally described in varicella-zoster virus (VZV) infection, while few systematic reviews are available. We systematically review autonomic dysfunction due to VZV infection. METHODS This study followed the PRISMA guideline, and three databases were researched and included cross-sectional studies in full-length publications in the English language using appropriate search keywords. RESULTS A total of 102 articles were identified initially; finally 45 studies were used for review, comprising pupillomotor dysfunction in 4, sudomotor dysfunction in 2, cardiovascular dysfunction in 2, gastrointestinal dysfunction in 14, and urogenital dysfunction in 23. They can be summarized as (1) VZV infection rarely produces orthostatic hypotension, which involves diffuse sympathetic dysfunction by polyneuropathy. (2) In contrast, VZV infection produces dysfunction of the bladder and the bowel, which involves segmental parasympathetic or sympathetic dysfunction by dorsal root ganglionopathy. CONCLUSIONS Awareness of VZV-related autonomic dysfunction is important, because such patients may first visit a gastroenterology or urology clinic. Close collaboration among neurologists, dermatologists, gastroenterologists, and urologists is important to start early antiviral agents and maximize bowel and bladder care in such patients.
Collapse
Affiliation(s)
- Ryuji Sakakibara
- Department of Neurology, Sakura Medical Center, Toho University, Sakura, Japan.
| | - Setsu Sawai
- Department of Neurology, Sakura Medical Center, Toho University, Sakura, Japan
| | - Tsuyoshi Ogata
- Department of Neurology, Sakura Medical Center, Toho University, Sakura, Japan
| |
Collapse
|
18
|
Immunogenicity of Varicella Zoster Virus DNA Vaccines Encoding Glycoprotein E and Immediate Early Protein 63 in Mice. Viruses 2022; 14:v14061214. [PMID: 35746685 PMCID: PMC9230688 DOI: 10.3390/v14061214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/05/2023] Open
Abstract
Herpes zoster (HZ) is caused by the reactivation of latent varicella-zoster virus (VZV) from the sensory ganglia due to aging or immunosuppression. Glycoprotein E (gE) is a widely used vaccine antigen for specific humoral and cellular immune responses. Immediate early protein 63 (IE63) is expressed during latency, suggesting that it is a potential antigen against HZ reactivation. In this study, HZ DNA vaccines encoding gE, IE63, IE63-2A-gE (where 2A is a self-cleaving sequence), or IE63-linker-gE were developed and investigated for immunogenicity in mice. The results showed that each HZ DNA vaccine induced VZV-specific antibody production. The neutralizing antibody titer elicited by IE63-2A-gE was comparable to that elicited by gE or live attenuated HZ vaccine (LAV). IE63-2A-gE-induced gE or IE63-specific INF-γ+ T cell frequencies in splenocytes were comparable to those of LAV. Furthermore, IE63-2A-gE, gE, or IE63 led to a significant increase in IFN-γ (IE63 stimulation) and IL-2 (gE stimulation) secretion compared to LAV, showing a Th1-biased immune response. Moreover, IE63-2A-gE and gE induced cytotoxic activity of CD8+ T cells compared to that of LAV. This study elucidates that the IE63-2A-gE DNA vaccine can induce both humoral and cell-mediated immune responses, which provides a candidate for the development of an HZ vaccine.
Collapse
|
19
|
Gastric herpes simplex virus type 1 infection is associated with functional gastrointestinal disorders in the presence and absence of comorbid fibromyalgia: a pilot case-control study. Infection 2022; 50:1303-1311. [PMID: 35445970 PMCID: PMC9522778 DOI: 10.1007/s15010-022-01823-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/31/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE Animal studies have linked gastric herpesvirus infections to symptoms associated with functional gastrointestinal disorders (FGIDs). Herpesviruses have also been hypothesized to contribute to fibromyalgia (FM), a chronic pain syndrome frequently comorbid with FGIDs. The purpose of this study was to compare the prevalence of gastric herpesvirus infection in patients with FGIDs, with and without comorbid FM, to that of controls. METHODS For this pilot case-control study, we enrolled 30 patients who met both the Rome IV diagnostic criteria for one or more FGIDs and the American College of Rheumatology 2010 criteria for FM, 15 patients with one or more FGIDs without comorbid FM, and 15 control patients. Following endoscopic examination, gastric biopsies were analyzed for herpesvirus DNA and protein, Helicobacter pylori infection, and histological evidence of gastritis. Importantly, the viral nonstructural protein ICP8 was used as a marker to differentiate cell-associated actively replicating virus from latent infection and/or free virus passing through the GI tract. RESULTS Gastric herpes simplex virus type 1 (HSV-1) infection, as indicated by ICP8 presence, was significantly associated with FGIDs in the presence (OR 70.00, 95% CI 7.42-660.50; P < .001) and absence (OR 38.50, 95% CI 3.75-395.40; P < .001) of comorbid FM. Neither histological gastritis nor H. pylori infection were found to be associated with FGIDs or FM. CONCLUSIONS HSV-1 infection was identified in gastric mucosal biopsies from patients with diverse FGIDs, with and without comorbid FM. Larger, multi-center studies investigating the prevalence of this association are warranted.
Collapse
|
20
|
Adducchio S, Reyes I, Chikkannaiah M, Rasch M, Kumar G. Bilateral Ptosis, Zosteriform Rash and Flaccid Bladder in a 10-Year-old boy. Child Neurol Open 2022; 9:2329048X221079429. [PMID: 35174255 PMCID: PMC8841934 DOI: 10.1177/2329048x221079429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 11/17/2022] Open
Abstract
We present a case report of a 10-year-old completely immunized boy presenting with a 2-week history of bilateral eyelid drooping, fatigue followed by bladder and bowel paralysis. This was followed by the appearance of a vesicular painful and itchy rash which directed further diagnosis and treatment as it was consistent with a varicella reactivation rash. This case is a very important addition to the current body of literature on varicella-related neurological complications. It outlines that varicella reactivation can present in completely vaccinated, immunocompetent young children as a neurological syndrome affecting the autonomic nervous system primarily and the rash can occur a few weeks later after presentation of the neurological symptoms.
Collapse
Affiliation(s)
| | - Irma Reyes
- Dayton Children’s Hospital, Dayton, Ohio, USA
- Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Mahesh Chikkannaiah
- Dayton Children’s Hospital, Dayton, Ohio, USA
- Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Matthew Rasch
- Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Gogi Kumar
- Dayton Children’s Hospital, Dayton, Ohio, USA
- Wright State University Boonshoft School of Medicine, Dayton, Ohio
| |
Collapse
|
21
|
Arostegui D, Castro K, Schwarz S, Vaidy K, Rabinowitz S, Wallach T. Persistent SARS-CoV-2 Nucleocapsid Protein Presence in the Intestinal Epithelium of a Pediatric Patient 3 Months After Acute Infection. JPGN REPORTS 2022; 3:e152. [PMID: 37168753 PMCID: PMC10158423 DOI: 10.1097/pg9.0000000000000152] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/01/2021] [Indexed: 05/13/2023]
Abstract
In addition to the severe impact of acute respiratory disease during the SARS-CoV-2 pandemic, the issue of "Long COVID" illness has impacted large numbers of patients following the initial infection. Wide ranges of Long Covid incidence have been reported, ranging from 30 to 87%. Long COVID has a variety of clinical manifestations, including gastrointestinal symptoms. Here, we report a case of persistent abdominal pain, 3 months following a SARS-CoV-2 diagnosis, associated with chronic colonic inflammation and the presence of mucosal SARS-CoV-2 virions.
Collapse
Affiliation(s)
- Dalia Arostegui
- From the SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Kenny Castro
- From the SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Steven Schwarz
- From the SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Katherine Vaidy
- From the SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Simon Rabinowitz
- From the SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Thomas Wallach
- From the SUNY Downstate Health Sciences University, Brooklyn, NY
| |
Collapse
|
22
|
Goldstein RS, Kinchington PR. Varicella Zoster Virus Neuronal Latency and Reactivation Modeled in Vitro. Curr Top Microbiol Immunol 2021; 438:103-134. [PMID: 34904194 DOI: 10.1007/82_2021_244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Latency and reactivation in neurons are critical aspects of VZV pathogenesis that have historically been difficult to investigate. Viral genomes are retained in many human ganglia after the primary infection, varicella; and about one-third of the naturally infected VZV seropositive population reactivates latent virus, which most often clinically manifests as herpes zoster (HZ or Shingles). HZ is frequently complicated by acute and chronic debilitating pain for which there remains a need for more effective treatment options. Understanding of the latent state is likely to be essential in the design of strategies to reduce reactivation. Experimentally addressing VZV latency has been difficult because of the strict human species specificity of VZV and the fact that until recently, experimental reactivation had not been achieved. We do not yet know the neuron subtypes that harbor latent genomes, whether all can potentially reactivate, what the drivers of VZV reactivation are, and how immunity interplays with the latent state to control reactivation. However, recent advances have enabled a picture of VZV latency to start to emerge. The first is the ability to detect the latent viral genome and its expression in human ganglionic tissues with extraordinary sensitivity. The second, the subject of this chapter, is the development of in vitro human neuron systems permitting the modeling of latent states that can be experimentally reactivated. This review will summarize recent advances of in vitro models of neuronal VZV latency and reactivation, the limitations of the current systems, and discuss outstanding questions and future directions regarding these processes using these and yet to be developed models. Results obtained from the in vitro models to date will also be discussed in light of the recent data gleaned from studies of VZV latency and gene expression learned from human cadaver ganglia, especially the discovery of VZV latency transcripts that seem to parallel the long-studied latency-associated transcripts of other neurotropic alphaherpesviruses.
Collapse
Affiliation(s)
| | - Paul R Kinchington
- Department of Ophthalmology, and Department of Molecular Microbiology and Genetics, University of Pittsburgh, EEI 1020, 203 Lothrop Street, Pittsburgh, PA, 156213, USA.
| |
Collapse
|
23
|
Prikhodchenko NG. Varicella-pox virus infection: features of the course, clinical manifestations, complications, and possibilities for prevention. TERAPEVT ARKH 2021; 93:1401-1406. [DOI: 10.26442/00403660.2021.11.201192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 11/22/2022]
Abstract
Varicella zoster virus (VZV) is a pathogenic human herpes virus that causes chickenpox as a primary infection, after which it persists for a long time and latently in the peripheral ganglia. Decades later, the virus can reactivate spontaneously, or after exposure to a number of triggering factors, causing herpes zoster (shingles). The reasons for the long-term persistence of VZV are gradually being revealed, but some issues remain unknown at the moment. Chickenpox and its complications are especially difficult in immunocompromised patients, but they are often found in people without risk factors. The most frequent and important complication of VZV reactivation is postherpetic neuralgia; encephalitis, segmental motor weakness and myelopathy, cranial neuropathies, and gastrointestinal complications often develop. The only scientifically proven effective and affordable way of mass prevention at the moment is vaccination. Chickenpox vaccines are safe and effective in preventing morbidity and mortality associated with the disease.
Collapse
|
24
|
Gershon AA, Gershon MD. Widespread use of varicella vaccine does not reduce immunity to zoster of others. J Infect Dis 2021; 225:361-363. [PMID: 34609507 PMCID: PMC8807174 DOI: 10.1093/infdis/jiab501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Anne A Gershon
- Departments of Pediatrics and Pathology and Cell Biology, Columbia University, Vagelos College of P&S, New York, NY
| | - Michael D Gershon
- Departments of Pediatrics and Pathology and Cell Biology, Columbia University, Vagelos College of P&S, New York, NY
| |
Collapse
|
25
|
Naik RD, Vaezi MF, Gershon AA, Higginbotham T, Chen J, Flores E, Holzman M, Patel DP, Gershon MD. Association of Achalasia With Active Varicella Zoster Virus Infection of the Esophagus. Gastroenterology 2021; 161:719-721.e2. [PMID: 33932481 PMCID: PMC8601651 DOI: 10.1053/j.gastro.2021.04.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/17/2021] [Accepted: 04/26/2021] [Indexed: 12/02/2022]
Affiliation(s)
- RD Naik
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt Medical Center, Nashville, TN, USA
| | - MF Vaezi
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt Medical Center, Nashville, TN, USA
| | - AA Gershon
- Department of Pediatrics; Columbia University Vagelos College of Physicians and Surgeons; New York, NY, USA
| | - T Higginbotham
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt Medical Center, Nashville, TN, USA
| | - J Chen
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons; New York, NY, USA
| | - E Flores
- Department of Pediatrics; Columbia University Vagelos College of Physicians and Surgeons; New York, NY, USA
| | - M Holzman
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt Medical Center, Nashville, TN, USA
| | - DP Patel
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt Medical Center, Nashville, TN, USA
| | - MD Gershon
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons; New York, NY, USA
| |
Collapse
|
26
|
Abstract
The enteric nervous system (ENS) is the largest division of the peripheral nervous system and closely resembles components and functions of the central nervous system. Although the central role of the ENS in congenital enteric neuropathic disorders, including Hirschsprung disease and inflammatory and functional bowel diseases, is well acknowledged, its role in systemic diseases is less understood. Evidence of a disordered ENS has accumulated in neurodegenerative diseases ranging from amyotrophic lateral sclerosis, Alzheimer disease and multiple sclerosis to Parkinson disease as well as neurodevelopmental disorders such as autism. The ENS is a key modulator of gut barrier function and a regulator of enteric homeostasis. A 'leaky gut' represents the gateway for bacterial and toxin translocation that might initiate downstream processes. Data indicate that changes in the gut microbiome acting in concert with the individual genetic background can modify the ENS, central nervous system and the immune system, impair barrier function, and contribute to various disorders such as irritable bowel syndrome, inflammatory bowel disease or neurodegeneration. Here, we summarize the current knowledge on the role of the ENS in gastrointestinal and systemic diseases, highlighting its interaction with various key players involved in shaping the phenotypes. Finally, current flaws and pitfalls related to ENS research in addition to future perspectives are also addressed.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Varicella zoster virus (VZV) causes varicella, establishes latency, then reactivates to produce herpes zoster. VZV reactivation can also cause central nervous system (CNS) disease with or without rash. Herein, we review these CNS diseases, pathogenesis, diagnosis, and treatment. RECENT FINDINGS The most common CNS manifestation of VZV infection is vasculopathy that presents as headache, cognitive decline, and/or focal neurological deficits. VZV vasculopathy has also been associated with cerebral amyloid angiopathy and moyamoya syndrome. Rarely, VZV will produce a meningitis, encephalitis, cerebellitis, and myelopathy. Pathogenic mechanisms include direct VZV infection of affected tissue, persistent inflammation, and/or virus-induced hypercoagulability. Diagnosis is confirmed by the temporal association of rash to disease onset, intrathecal synthesis of anti-VZV antibodies, and/or the presence of VZV DNA in CSF. Most cases respond to intravenous acyclovir with corticosteroids. SUMMARY VZV produces a wide spectrum of CNS disorders that may be missed as some cases do not have an associated rash or a CSF pleocytosis. Clinicians must be vigilant in including VZV in their differential diagnosis of CNS infections as VZV is a ubiquitous pathogen; importantly, VZV CNS infections are treatable with intravenous acyclovir therapy and corticosteroids.
Collapse
|
28
|
Clinicopathologic Features of Varicella Zoster Virus Infection of the Upper Gastrointestinal Tract. Am J Surg Pathol 2021; 45:209-214. [PMID: 32826528 DOI: 10.1097/pas.0000000000001576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Reactivation of latent varicella zoster virus (VZV) may be limited to a dermatome or involve multiple organs, including the gastrointestinal tract. Although gastrointestinal manifestations of disseminated zoster have been likened to those of herpes simplex virus (HSV), histologic features of VZV-related injury to the tubular gut are not well-documented. We performed this study to describe the clinicopathologic features of VZV-related gastrointestinal injury. We identified 6 such patients with VZV infection. All involved the upper gastrointestinal tract, affecting the esophagus (n=3), stomach (n=2), or both (n=1). All patients were immunocompromised adults with hematologic malignancies (n=5) or a heart transplant (n=1); 3 with hematologic malignancies had received stem cell transplants. Five patients had cutaneous and gastrointestinal zoster; 1 had gastrointestinal disease alone. When compared with 14 HSV-related esophagitis controls, there were several notable differences. VZV caused hemorrhagic ulcers with nodularity or erythema, whereas HSV produced round, shallow ulcers on a background of nearly normal mucosa (P=0.01). VZV-related ulcers featured fibrin-rich, pauci-inflammatory exudates compared with the macrophage-rich exudates of HSV (P=0.003). The cytopathic changes of VZV were present at all levels of the squamous epithelium, especially in a peripapillary distribution. In contrast, HSV inclusions were located in the superficial layers (P=0.003) and detached keratinocytes. Unlike HSV, VZV involved the stomach, producing hemorrhage accompanied by striking apoptosis in the deep glands. We conclude that VZV produces unique patterns of gastrointestinal injury that facilitate its diagnosis. Recognition of gastrointestinal VZV infection is important because it heralds potentially life-threatening disseminated disease.
Collapse
|
29
|
Windster JD, Ouwendijk WJD, Sloots CEJ, Verjans GMGM, Verdijk RM. Ileocolic Intussusception as the Presenting Symptom of Primary Enteric Varicella-Zoster Virus Infection in a 7-Month-Old Infant. J Infect Dis 2021; 222:305-308. [PMID: 32232390 PMCID: PMC7323496 DOI: 10.1093/infdis/jiaa148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/27/2020] [Indexed: 12/29/2022] Open
Abstract
Ileocolic intussusception is the invagination of ileum into the colon. In a subset of patients, the disease is caused by mesenteric lymphadenopathy in response to (viral) infection. We present a case of an ileocolic intussusception necessitating surgery in a 7-month-old immunocompetent infant with concurrent primary wild-type varicella-zoster virus (VZV) infection, in whom chickenpox rash developed 2 days after surgery. Detailed in situ analyses of resected intestine for specific cell type markers and VZV RNA demonstrated VZV-infected lymphocytes and neurons in the gut wall and in ganglion cells of the myenteric plexus.
Collapse
Affiliation(s)
- Jonathan D Windster
- Department of Pediatric Surgery, Erasmus University Medical Center Rotterdam, Sophia's Children's Hospital, Rotterdam, the Netherlands
| | - Werner J D Ouwendijk
- Department of Viroscience, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Cornelius E J Sloots
- Department of Pediatric Surgery, Erasmus University Medical Center Rotterdam, Sophia's Children's Hospital, Rotterdam, the Netherlands
| | - Georges M G M Verjans
- Department of Viroscience, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Robert M Verdijk
- Department of Pathology, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
30
|
Corticosteroids Contribute to Serious Adverse Events Following Live Attenuated Varicella Vaccination and Live Attenuated Zoster Vaccination. Vaccines (Basel) 2021; 9:vaccines9010023. [PMID: 33418856 PMCID: PMC7825138 DOI: 10.3390/vaccines9010023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/28/2020] [Accepted: 01/01/2021] [Indexed: 12/28/2022] Open
Abstract
Corticosteroids, when given in high dosages, have long been recognized as a risk factor for severe infection with wild-type varicella-zoster virus in both children and adults. The goal of this review is to assess the degree to which both low-dosage and high-dosage corticosteroids contribute to serious adverse events (SAEs) following live varicella vaccination and live zoster vaccination. To this end, we examined multiple published reports of SAEs following varicella vaccination (VarivaxTM) and zoster vaccination (ZostavaxTM). We observed that five of eight viral SAEs following varicella vaccination, including two deaths, occurred in children receiving corticosteroids, while one of three fatal viral SAEs following live zoster vaccination occurred in an adult being treated with low-dosage prednisone. The latter death after live zoster vaccination occurred in a 70 year-old man with rheumatoid arthritis, being treated with prednisone 10 mg daily. Thus, corticosteroids contributed to more severe infectious complications in subjects immunized with each of the two live virus vaccines. Further, when we surveyed the rheumatology literature as well as individual case reports, we documented examples where daily dosages of 7.5–20 mg prednisone were associated with increased rates of severe wild-type varicella-zoster virus infections in children and adults.
Collapse
|
31
|
Ouwendijk WJD, Depledge DP, Rajbhandari L, Lenac Rovis T, Jonjic S, Breuer J, Venkatesan A, Verjans GMGM, Sadaoka T. Varicella-zoster virus VLT-ORF63 fusion transcript induces broad viral gene expression during reactivation from neuronal latency. Nat Commun 2020; 11:6324. [PMID: 33303747 PMCID: PMC7730162 DOI: 10.1038/s41467-020-20031-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Varicella-zoster virus (VZV) establishes lifelong neuronal latency in most humans world-wide, reactivating in one-third to cause herpes zoster and occasionally chronic pain. How VZV establishes, maintains and reactivates from latency is largely unknown. VZV transcription during latency is restricted to the latency-associated transcript (VLT) and RNA 63 (encoding ORF63) in naturally VZV-infected human trigeminal ganglia (TG). While significantly more abundant, VLT levels positively correlated with RNA 63 suggesting co-regulated transcription during latency. Here, we identify VLT-ORF63 fusion transcripts and confirm VLT-ORF63, but not RNA 63, expression in human TG neurons. During in vitro latency, VLT is transcribed, whereas VLT-ORF63 expression is induced by reactivation stimuli. One isoform of VLT-ORF63, encoding a fusion protein combining VLT and ORF63 proteins, induces broad viral gene transcription. Collectively, our findings show that VZV expresses a unique set of VLT-ORF63 transcripts, potentially involved in the transition from latency to lytic VZV infection.
Collapse
Affiliation(s)
- Werner J D Ouwendijk
- Department of Viroscience, Erasmus Medical Centre, 3015 CN, Rotterdam, The Netherlands
| | - Daniel P Depledge
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Labchan Rajbhandari
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Meyer 6-113, Baltimore, MD, 21287, USA
| | - Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, 51000, Croatia
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, 51000, Croatia
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK
| | - Arun Venkatesan
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Meyer 6-113, Baltimore, MD, 21287, USA
| | - Georges M G M Verjans
- Department of Viroscience, Erasmus Medical Centre, 3015 CN, Rotterdam, The Netherlands
| | - Tomohiko Sadaoka
- Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
32
|
Kofahi RM, Kofahi HM, Sabaheen S, Qawasmeh MA, Momani A, Yassin A, Alhayk K, El-Salem K. Prevalence of seropositivity of selected herpesviruses in patients with multiple sclerosis in the North of Jordan. BMC Neurol 2020; 20:397. [PMID: 33121451 PMCID: PMC7596955 DOI: 10.1186/s12883-020-01977-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
Background Multiple sclerosis (MS) is a neurological disease that is caused by an autoimmune response that results in the neuron’s demyelination in the central nervous system. The exact etiology of MS is not clear; however, several environmental and genetic factors are believed to participate in its initiation and development, including exposure to viruses. This study aims to investigate the association between the seropositivity and antibody titer of selected herpesviruses and MS in Jordanian MS patients. Method In this study, 55 MS patients and 40 age- and gender-matching apparently healthy volunteers were recruited from two main hospitals in the north of Jordan. MS patients were grouped into three types of MS based on the clinical presentation of the disease. Blood samples were collected from the participants and the IgG antibodies for human herpesvirus 6 (HHV-6), Epstein-Barr virus (EBV) nuclear antigen (EBNA), EBV viral capsid antigen (VCA) and varicella-zoster virus (VZV) were assayed by ELISA. The prevalence of seropositivity and the antibody level for each of the antibodies were compared between MS patients and controls and between the three types of MS. Results There was no significant difference in the prevalence of seropositivity and in the levels of antibodies for HHV-6, EBNA and VCA between MS patients and controls and between the three types of MS. In contrast, the number of seropositive patients and the level of IgG antibodies for VZV were significantly higher in MS patients compared to the control. Conclusion This study showed that patients with MS in the north of Jordan were more likely to be seropositive for VZV than the general population. Based on this finding, we recommend further studies to evaluate the seropositivity to VZV to be carried out in other parts of Jordan and the greater middle east to find out if there is a correlation between MS and previous infection with VZV. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-020-01977-w.
Collapse
Affiliation(s)
- Raid M Kofahi
- Department of Neurosciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Hassan M Kofahi
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Suhib Sabaheen
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Majdi Al Qawasmeh
- Department of Neurosciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Aiman Momani
- Department of Neurosciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Ahmed Yassin
- Department of Neurosciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Kefah Alhayk
- Department of Neurosciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Khalid El-Salem
- Department of Neurosciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| |
Collapse
|
33
|
Schreiner P, Mueller NJ, Fehr J, Maillard MH, Brand S, Michetti P, Schoepfer A, Restellini S, Vulliemoz M, Vavricka SR, Juillerat P, Rogler G, Biedermann L. Varicella zoster virus in inflammatory bowel disease patients: what every gastroenterologist should know. J Crohns Colitis 2020; 15:jjaa132. [PMID: 32592587 DOI: 10.1093/ecco-jcc/jjaa132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Indexed: 12/17/2022]
Abstract
Primary Varicella Zoster virus (VZV) infection results in varicella (chickenpox) while its reactivation results in herpes zoster (HZ; shingles). Patients with Inflammatory Bowel Disease (IBD) are susceptible to complications of primary VZV infection and have an increased risk of HZ. Concerns of VZV and HZ infection in the IBD population has been highlighted by the emergence of JAK-inhibitors and their safety profile in this patient population such as tofacitinib for the treatment of ulcerative colitis (UC). The current pipeline of emerging therapies include novel molecules targeting multiple pathways including JAK/signal transducer and cytokine signalling pathways such as JAK/STAT. Hence VZV and HZ will be increasingly relevant for gastroenterologists treating IBD patients in light of these emerging therapies.
Collapse
Affiliation(s)
- Philipp Schreiner
- Department of Gastroenterology & Hepatology, University Hospital Zurich
| | - Nicolas J Mueller
- Department of Infectious Diseases & Hospital Epidemiology, University Hospital Zurich, Switzerland
| | - Jan Fehr
- Department of Infectious Diseases & Hospital Epidemiology, University Hospital Zurich, Switzerland
- Department of Public & Global Health, University of Zurich, Zurich, Switzerland
| | - Michel H Maillard
- Crohn and Colitis Center, Gastroentérologie Beaulieu SA, Lausanne, Switzerland
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Stephan Brand
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kantonsspital Sankt Gallen, St. Gallen, Switzerland
| | - Pierre Michetti
- Crohn and Colitis Center, Gastroentérologie Beaulieu SA, Lausanne, Switzerland
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Alain Schoepfer
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Sophie Restellini
- Department of Gastroenterology and Hepatology, Geneva University Hospitals and University of Geneva, Switzerland
| | - Marianne Vulliemoz
- Crohn and Colitis Center, Gastroentérologie Beaulieu SA, Lausanne, Switzerland
| | - Stephan R Vavricka
- Department of Gastroenterology & Hepatology, University Hospital Zurich
- Center of Gastroenterology and Hepatology, CH, Zurich, Switzerland
| | - Pascal Juillerat
- Gastroenterology, Clinic for Visceral Surgery and Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology & Hepatology, University Hospital Zurich
| | | |
Collapse
|
34
|
Bello-Morales R, Ripa I, López-Guerrero JA. Extracellular Vesicles in Viral Spread and Antiviral Response. Viruses 2020; 12:E623. [PMID: 32521696 PMCID: PMC7354624 DOI: 10.3390/v12060623] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Viral spread by both enveloped and non-enveloped viruses may be mediated by extracellular vesicles (EVs), including microvesicles (MVs) and exosomes. These secreted vesicles have been demonstrated to be an efficient mechanism that viruses can use to enter host cells, enhance spread or evade the host immune response. However, the complex interplay between viruses and EVs gives rise to antagonistic biological tasks-to benefit the viruses, enhancing infection and interfering with the immune system or to benefit the host, by mediating anti-viral responses. Exosomes from cells infected with herpes simplex type 1 (HSV-1) may transport viral and host transcripts, proteins and innate immune components. This virus may also use MVs to expand its tropism and evade the host immune response. This review aims to describe the current knowledge about EVs and their participation in viral infection, with a specific focus on the role of exosomes and MVs in herpesvirus infections, particularly that of HSV-1.
Collapse
Affiliation(s)
- Raquel Bello-Morales
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; (I.R.); (J.A.L.-G.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049 Madrid, Spain
| | - Inés Ripa
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; (I.R.); (J.A.L.-G.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049 Madrid, Spain
| | - José Antonio López-Guerrero
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; (I.R.); (J.A.L.-G.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
35
|
Bubak AN, Beseler C, Como CN, Tyring SK, Haley C, Mescher T, Hassell JE, Cohrs RJ, Potter H, Nagel MA. Acute zoster plasma contains elevated amyloid, correlating with Aβ42 and amylin levels, and is amyloidogenic. J Neurovirol 2020; 26:422-428. [PMID: 32385803 DOI: 10.1007/s13365-020-00830-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 12/19/2022]
Abstract
Herpes zoster is associated with an increased dementia and neovascular macular degeneration risk and a decline in glycemic control in diabetes mellitus. Because amyloid is present and pathogenic in these diseases, we quantified amyloid, Aβ40, Aβ42, and amylin in 14 zoster and 10 control plasmas. Compared with controls, zoster plasma had significantly elevated amyloid that correlated with Aβ42 and amylin levels and increased amyloid aggregation with addition of exogenous Aβ42 or amylin. These results suggest that zoster plasma contains factor(s) that promotes aggregation of amyloidogenic peptides, potentially contributing to the toxic amyloid burden and explaining accelerated disease progression following zoster.
Collapse
Affiliation(s)
- Andrew N Bubak
- Department of Neurology, University of Colorado School of Medicine, 12700 E. 19th Avenue, Mail Stop B182, Aurora, CO, 80045, USA
| | - Cheryl Beseler
- Department of Psychology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Christina N Como
- Department of Neurology, University of Colorado School of Medicine, 12700 E. 19th Avenue, Mail Stop B182, Aurora, CO, 80045, USA
| | - Stephen K Tyring
- Department of Dermatology, Center for Clinical Studies, University of Texas Health Science Center, Houston, TX, 77598, USA
| | - Christopher Haley
- Department of Dermatology, Center for Clinical Studies, University of Texas Health Science Center, Houston, TX, 77598, USA
| | - Teresa Mescher
- Department of Neurology, University of Colorado School of Medicine, 12700 E. 19th Avenue, Mail Stop B182, Aurora, CO, 80045, USA
| | - James E Hassell
- Department of Neurology, University of Colorado School of Medicine, 12700 E. 19th Avenue, Mail Stop B182, Aurora, CO, 80045, USA
| | - Randall J Cohrs
- Department of Neurology, University of Colorado School of Medicine, 12700 E. 19th Avenue, Mail Stop B182, Aurora, CO, 80045, USA.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Huntington Potter
- Department of Neurology, University of Colorado School of Medicine, 12700 E. 19th Avenue, Mail Stop B182, Aurora, CO, 80045, USA
| | - Maria A Nagel
- Department of Neurology, University of Colorado School of Medicine, 12700 E. 19th Avenue, Mail Stop B182, Aurora, CO, 80045, USA. .,Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
36
|
Bubak AN, Como CN, Coughlan CM, Johnson NR, Hassell JE, Mescher T, Niemeyer CS, Mahalingam R, Cohrs RJ, Boyd TD, Potter H, Russ HA, Nagel MA. Varicella-Zoster Virus Infection of Primary Human Spinal Astrocytes Produces Intracellular Amylin, Amyloid-β, and an Amyloidogenic Extracellular Environment. J Infect Dis 2020; 221:1088-1097. [PMID: 31665341 PMCID: PMC7075411 DOI: 10.1093/infdis/jiz560] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Herpes zoster is linked to amyloid-associated diseases, including dementia, macular degeneration, and diabetes mellitus, in epidemiological studies. Thus, we examined whether varicella-zoster virus (VZV)-infected cells produce amyloid. METHODS Production of intracellular amyloidogenic proteins (amylin, amyloid precursor protein [APP], and amyloid-β [Aβ]) and amyloid, as well as extracellular amylin, Aβ, and amyloid, was compared between mock- and VZV-infected quiescent primary human spinal astrocytes (qHA-sps). The ability of supernatant from infected cells to induce amylin or Aβ42 aggregation was quantitated. Finally, the amyloidogenic activity of viral peptides was examined. RESULTS VZV-infected qHA-sps, but not mock-infected qHA-sps, contained intracellular amylin, APP, and/or Aβ, and amyloid. No differences in extracellular amylin, Aβ40, or Aβ42 were detected, yet only supernatant from VZV-infected cells induced amylin aggregation and, to a lesser extent, Aβ42 aggregation into amyloid fibrils. VZV glycoprotein B (gB) peptides assembled into fibrils and catalyzed amylin and Aβ42 aggregation. CONCLUSIONS VZV-infected qHA-sps produced intracellular amyloid and their extracellular environment promoted aggregation of cellular peptides into amyloid fibrils that may be due, in part, to VZV gB peptides. These findings suggest that together with host and other environmental factors, VZV infection may increase the toxic amyloid burden and contribute to amyloid-associated disease progression.
Collapse
Affiliation(s)
- Andrew N Bubak
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Christina N Como
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Christina M Coughlan
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Alzheimer’s Disease Center, University of Colorado School of Medicine, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Research, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Noah R Johnson
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Alzheimer’s Disease Center, University of Colorado School of Medicine, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Research, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - James E Hassell
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Teresa Mescher
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Christy S Niemeyer
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ravi Mahalingam
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Randall J Cohrs
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Timothy D Boyd
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Alzheimer’s Disease Center, University of Colorado School of Medicine, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Research, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Huntington Potter
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Alzheimer’s Disease Center, University of Colorado School of Medicine, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Research, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Holger A Russ
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Maria A Nagel
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
37
|
Ahn J, Barber GN. STING signaling and host defense against microbial infection. Exp Mol Med 2019; 51:1-10. [PMID: 31827069 PMCID: PMC6906460 DOI: 10.1038/s12276-019-0333-0] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
The first line of host defense against infectious agents involves activation of innate immune signaling pathways that recognize specific pathogen-associated molecular patterns (PAMPs). Key triggers of innate immune signaling are now known to include microbial-specific nucleic acid, which is rapidly detected in the cytosol of the cell. For example, RIG-I-like receptors (RLRs) have evolved to detect viral RNA species and to activate the production of host defense molecules and cytokines that stimulate adaptive immune responses. In addition, host defense countermeasures, including the production of type I interferons (IFNs), can also be triggered by microbial DNA from bacteria, viruses and perhaps parasites and are regulated by the cytosolic sensor, stimulator of interferon genes (STING). STING-dependent signaling is initiated by cyclic dinucleotides (CDNs) generated by intracellular bacteria following infection. CDNs can also be synthesized by a cellular synthase, cGAS, following interaction with invasive cytosolic self-DNA or microbial DNA species. The importance of STING signaling in host defense is evident since numerous pathogens have developed strategies to prevent STING function. Here, we review the relevance of STING-controlled innate immune signaling in host defense against pathogen invasion, including microbial endeavors to subvert this critical process.
Collapse
Affiliation(s)
- Jeonghyun Ahn
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Glen N Barber
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
38
|
Laemmle L, Goldstein RS, Kinchington PR. Modeling Varicella Zoster Virus Persistence and Reactivation - Closer to Resolving a Perplexing Persistent State. Front Microbiol 2019; 10:1634. [PMID: 31396173 PMCID: PMC6667558 DOI: 10.3389/fmicb.2019.01634] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/02/2019] [Indexed: 12/20/2022] Open
Abstract
The latent state of the human herpesvirus varicella zoster virus (VZV) has remained enigmatic and controversial. While it is well substantiated that VZV persistence is established in neurons after the primary infection (varicella or chickenpox), we know little of the types of neurons harboring latent virus genomes, if all can potentially reactivate, what exactly drives the reactivation process, and the role of immunity in the control of latency. Viral gene expression during latency has been particularly difficult to resolve, although very recent advances indicate that it is more restrictive than was once thought. We do not yet understand how genes expressed in latency function in the maintenance and reactivation processes. Model systems of latency are needed to pursue these questions. This has been especially challenging for VZV because the development of in vivo models of VZV infection has proven difficult. Given that up to one third of the population will clinically reactivate VZV to develop herpes zoster (shingles) and suffer from its common long term problematic sequelae, there is still a need for both in vivo and in vitro model systems. This review will summarize the evolution of models of VZV persistence and address insights that have arisen from the establishment of new in vitro human neuron culture systems that not only harbor a latent state, but permit experimental reactivation and renewed virus production. These models will be discussed in light of the recent data gleaned from the study of VZV latency in human cadaver ganglia.
Collapse
Affiliation(s)
- Lillian Laemmle
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Paul R Kinchington
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
39
|
Current In Vivo Models of Varicella-Zoster Virus Neurotropism. Viruses 2019; 11:v11060502. [PMID: 31159224 PMCID: PMC6631480 DOI: 10.3390/v11060502] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/24/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
Varicella-zoster virus (VZV), an exclusively human herpesvirus, causes chickenpox and establishes a latent infection in ganglia, reactivating decades later to produce zoster and associated neurological complications. An understanding of VZV neurotropism in humans has long been hampered by the lack of an adequate animal model. For example, experimental inoculation of VZV in small animals including guinea pigs and cotton rats results in the infection of ganglia but not a rash. The severe combined immune deficient human (SCID-hu) model allows the study of VZV neurotropism for human neural sub-populations. Simian varicella virus (SVV) infection of rhesus macaques (RM) closely resembles both human primary VZV infection and reactivation, with analyses at early times after infection providing valuable information about the extent of viral replication and the host immune responses. Indeed, a critical role for CD4 T-cell immunity during acute SVV infection as well as reactivation has emerged based on studies using RM. Herein we discuss the results of efforts from different groups to establish an animal model of VZV neurotropism.
Collapse
|
40
|
Clinical Features of Varicella-Zoster Virus Infection. Viruses 2018; 10:v10110609. [PMID: 30400213 PMCID: PMC6266119 DOI: 10.3390/v10110609] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/30/2022] Open
Abstract
Varicella-zoster virus (VZV) is a pathogenic human herpes virus that causes varicella (chickenpox) as a primary infection, following which it becomes latent in peripheral ganglia. Decades later, the virus may reactivate either spontaneously or after a number of triggering factors to cause herpes zoster (shingles). Varicella and its complications are more severe in the immunosuppressed. The most frequent and important complication of VZV reactivation is postherpetic neuralgia, the cause of which is unknown and for which treatment is usually ineffective. Reactivation of VZV may also cause a wide variety of neurological syndromes, the most significant of which is a vasculitis, which is treated with corticosteroids and the antiviral drug acyclovir. Other VZV reactivation complications include an encephalitis, segmental motor weakness and myelopathy, cranial neuropathies, Guillain–Barré syndrome, enteric features, and zoster sine herpete, in which the viral reactivation occurs in the absence of the characteristic dermatomally distributed vesicular rash of herpes zoster. There has also been a recent association of VZV with giant cell arteritis and this interesting finding needs further corroboration. Vaccination is now available for the prevention of both varicella in children and herpes zoster in older individuals.
Collapse
|