1
|
Petersen PT, Bodilsen J, Jepsen MPG, Larsen L, Storgaard M, Hansen BR, Helweg-Larsen J, Wiese L, Lüttichau HR, Andersen CØ, Nielsen H, Brandt CT. Dexamethasone in adults with viral meningitis: an observational cohort study. Clin Microbiol Infect 2025; 31:87-92. [PMID: 39182578 DOI: 10.1016/j.cmi.2024.08.015] [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: 04/02/2024] [Revised: 08/07/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVES To investigate whether there is a dose-dependent association between empiric dexamethasone and outcome in viral meningitis. METHODS Observational cohort study of adults hospitalized for viral meningitis, both with and without a microbiologically confirmed diagnosis, in Denmark between 2015 and 2020. Dose-dependent associations between dexamethasone (one dose = 10 mg) and an unfavourable outcome (Glasgow Outcome Scale score 1-4) at 30 days after discharge were assessed using weighted logistic regression. Entropy balancing was used to compute weights. RESULTS Of 1025 included patients, 658 (64%) did not receive dexamethasone, 115 (11%) received 1-2 doses, 131 (13%) received 3-4 doses, and 121 (12%) received ≥5 doses. Among patients treated with dexamethasone, the median number of doses was higher for those without an identified pathogen than for those with a microbiologically confirmed viral aetiology (5 [interquartile range (IQR) 3-8] vs. 3 [IQR 2-5]; p < 0.001). Using no doses of dexamethasone as a reference, the weighted OR for an unfavourable outcome were 0.55 (95% CI, 0.29-1.07) for 1-2 doses, 1.13 (95% CI, 0.67-1.89) for 3-4 doses, and 1.43 (95% CI, 0.77-2.64) for ≥5 doses. In the subgroup of enteroviral meningitis, the weighted OR was 3.08 (95% CI, 1.36-6.94) for ≥5 doses, but decreased to 2.35 (95% CI, 0.65-8.40) when the reference group was restricted to patients treated with antibiotics for suspected bacterial meningitis. DISCUSSION This study showed no dose-dependent association between dexamethasone and an unfavourable outcome in patients with viral meningitis. In enteroviral meningitis, ≥5 doses were associated with an increased risk of an unfavourable outcome. However, sensitivity analysis indicated that the association was affected by unmeasured or residual confounding by severity.
Collapse
Affiliation(s)
- Pelle Trier Petersen
- Department of Pulmonary and Infectious Diseases, Nordsjællands Hospital, Hillerød, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jacob Bodilsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Lykke Larsen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| | - Merete Storgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Lothar Wiese
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | | | | | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
2
|
Beardsley J, McCoy A, Freeman M, Cramer N, Neville D, Owusu-Ansah S, Houtrow A, Sinha A. The complete acute and post-acute care course of children affected by acute flaccid myelitis in Western Pennsylvania: A case series. J Pediatr Rehabil Med 2023; 16:401-413. [PMID: 36776079 DOI: 10.3233/prm-210120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
Acute flaccid myelitis (AFM) is a "polio-like" neurologic disorder of the spinal cord gray matter characterized by asymmetric, flaccid limb weakness of rapid onset following prodromal viral illness. It has affected the pediatric population of the United States since 2014, but there is a paucity of literature describing the post-acute comprehensive rehabilitation management that maximizes functional outcomes for patients. This case series attempts to mitigate this by describing the complete acute and post-acute care course of six children diagnosed with AFM in Western Pennsylvania. It is critical that pediatric rehabilitation medicine providers be knowledgeable about the complex medical and rehabilitation management for patients with AFM.
Collapse
Affiliation(s)
| | - Andrew McCoy
- UPMC Physical Medicine and Rehabilitation, Pittsburgh, PA, USA
| | - Megan Freeman
- Pediatric Infectious Disease, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Natan Cramer
- Pediatric Emergency Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Desiree Neville
- Pediatric Emergency Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Silvia Owusu-Ansah
- Pediatric Emergency Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Amy Houtrow
- Pediatric Rehabilitation Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Amit Sinha
- Pediatric Rehabilitation Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Dinov D, Donowitz JR. Acute flaccid myelitis a review of the literature. Front Neurol 2022; 13:1034607. [PMID: 36605787 PMCID: PMC9807762 DOI: 10.3389/fneur.2022.1034607] [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: 09/01/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Acute flaccid myelitis (AFM) is a rare neurological disorder that first rose to national attention in 2014. This neurological disorder has a biennial presentation with every other even year being a peak year. Most patients present in childhood 5 days after a prodromal infection. Patients usually present with muscle weakness and hypo or areflexia in the summer or fall months. Clinical outcomes are variable however most patients do not improve. Currently there are no definitive prognostic factors or etiologies found. However, it is thought that enterovirus-D68 (EV-D68) could be a potential component in the pathobiology of AFM. Treatment options are limited with variable options and no consensus. Supportive therapy has been shown to be the most effective thus far. With our review of the literature, we highlight the recent growing evidence of a possible relationship between EV-D68 and AFM. Additionally, we identify the knowledge gaps in AFM with treatment and prognostic factors.
Collapse
Affiliation(s)
- Darina Dinov
- Department of Neurology, Children's Hospital of Richmond at Virginia Commonwealth University, Richmond, VA, United States,*Correspondence: Darina Dinov ✉
| | - Jeffrey R. Donowitz
- Department of Pediatrics, Children's Hospital of Richmond at Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
4
|
Lin JY, Weng KF, Chang CK, Gong YN, Huang GJ, Lee HL, Chen YC, Huang CC, Lu JY, Huang PN, Chiang HJ, Chen CM, Shih SR. Enterovirus A71 Induces Neurological Diseases and Dynamic Variants in Oral Infection of Human SCARB2-Transgenic Weaned Mice. J Virol 2021; 95:e0089721. [PMID: 34379497 PMCID: PMC8513470 DOI: 10.1128/jvi.00897-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/04/2021] [Indexed: 11/20/2022] Open
Abstract
Enterovirus A71 (EV-A71) and many members of the Picornaviridae family are neurotropic pathogens of global concern. These viruses are primarily transmitted through the fecal-oral route, and thus suitable animal models of oral infection are needed to investigate viral pathogenesis. An animal model of oral infection was developed using transgenic mice expressing human SCARB2 (hSCARB2 Tg), murine-adapted EV-A71/MP4 virus, and EV-A71/MP4 virus with an engineered nanoluciferase gene that allows imaging of viral replication and spread in infected mice. Next-generation sequencing of EV-A71 genomes in the tissues and organs of infected mice was also performed. Oral inoculation of EV-A71/MP4 or nanoluciferase-carrying MP4 virus stably induced neurological symptoms and death in infected 21-day-old weaned mice. In vivo bioluminescence imaging of infected mice and tissue immunostaining of viral antigens indicated that orally inoculated virus can spread to the central nervous system (CNS) and other tissues. Next-generating sequencing further identified diverse mutations in viral genomes that can potentially contribute to viral pathogenesis. This study presents an EV-A71 oral infection murine model that efficiently infects weaned mice and allows tracking of viral spread, features that can facilitate research into viral pathogenesis and neuroinvasion via the natural route of infection. IMPORTANCE Enterovirus A71 (EV-A71), a positive-strand RNA virus of the Picornaviridae, poses a persistent global public health problem. EV-A71 is primarily transmitted through the fecal-oral route, and thus suitable animal models of oral infection are needed to investigate viral pathogenesis. We present an animal model of EV-A71 infection that enables the natural route of oral infection in weaned and nonimmunocompromised 21-day-old hSCARB2 transgenic mice. Our results demonstrate that severe disease and death could be stably induced, and viral invasion of the CNS could be replicated in this model, similar to severe real-world EV-A71 infections. We also developed a nanoluciferase-containing EV-A71 virus that can be used with this animal model to track viral spread after oral infection in real time. Such a model offers several advantages over existing animal models and can facilitate future research into viral spread, tissue tropism, and viral pathogenesis, all pressing issues that remain unaddressed for EV-A71 infections.
Collapse
Affiliation(s)
- Jing-Yi Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei City, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| | - Kuo-Feng Weng
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chih-Kuang Chang
- Department of Laboratory Medicine, Taoyuan Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Yu-Nong Gong
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Guo-Jen Huang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Hui-Lan Lee
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Yen-Cheng Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Chien-Chih Huang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Jia-Ying Lu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Peng-Nien Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Division of Infectious Diseases, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Huan-Jung Chiang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Che-Min Chen
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Graduate Institute of Athletics and Coaching Science, National Taiwan Sport University, Taoyuan City, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| |
Collapse
|
5
|
Oliveira TESD, Leme RA, Agnol AMD, Gerez JR, Pelaquim IF, Miyabe FM, Alfieri AF, Alfieri AA, Headley SA. Seneca Valley virus induces immunodepressionin suckling piglets by selective apoptosis of B lymphocytes. Microb Pathog 2021; 158:105022. [PMID: 34129904 DOI: 10.1016/j.micpath.2021.105022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 02/09/2021] [Accepted: 03/08/2021] [Indexed: 11/27/2022]
Abstract
Seneca Valley virus (SVV) is the causative agent of an emerging infectious vesicular disease in swine that is clinically indistinguishable from other vesicular diseases of swine. This study utilized healthy suckling piglets (control) and SVV-naturally infected suckling piglets to determine the effects of SVV on lymphoid tissues and determined the SVV RNA load by quantitative RT-PCR (qRT-PCR). Furthermore, immunohistochemistry (IHC) analyses were performed to quantify the expression of T and B cell lymphocytes, natural killer cells, cleaved caspase 3, and ki-67. The main histopathologic finding in the infected group was severe lymphoid depletion. The highest average of SVV RNA load by qRT-PCR (Log10 genomic copies/g of tissue) occurred at the spleen (8.54 ± 0.8), followed by the tonsils (8.04 ± 1.42), and mesenteric lymph nodes (6.90 ± 1.42). The IHC analyses revealed that there was an increased in cellular apoptosis with concomitant reduction in the proliferation of B cells. The results from this study have demonstrated that SVV-infected piglets exhibited decreased lymphocyte density probably due to lymphoid apoptosis, affecting particularly B-cells lymphocytes.
Collapse
Affiliation(s)
- Thalita Evani Silva de Oliveira
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Raquel Arruda Leme
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Alais Maria Dall Agnol
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Juliana Rubira Gerez
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Isadora Fernanda Pelaquim
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Flavia Megumi Miyabe
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Alice Fernandes Alfieri
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Amauri Alcindo Alfieri
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil; Multi-User Animal Health Laboratory, Molecular Biology Unit, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil
| | - Selwyn Arlington Headley
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil; Multi-User Animal Health Laboratory, Tissue Processing Unit, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Paraná, Brazil.
| |
Collapse
|
6
|
Early-life EV-A71 infection augments allergen-induced airway inflammation in asthma through trained macrophage immunity. Cell Mol Immunol 2021; 18:472-483. [PMID: 33441966 PMCID: PMC8027667 DOI: 10.1038/s41423-020-00621-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/07/2020] [Indexed: 01/29/2023] Open
Abstract
Virus-induced asthma is prevalent among children, but its underlying mechanisms are unclear. Accumulated evidence indicates that early-life respiratory virus infection increases susceptibility to allergic asthma. Nonetheless, the relationship between systemic virus infections, such as enterovirus infection, and the ensuing effects on allergic asthma development is unknown. Early-life enterovirus infection was correlated with higher risks of allergic diseases in children. Adult mice exhibited exacerbated mite allergen-induced airway inflammation following recovery from EV-A71 infection in the neonatal period. Bone marrow-derived macrophages (BMDMs) from recovered EV-A71-infected mice showed sustained innate immune memory (trained immunity) that could drive naïve T helper cells toward Th2 and Th17 cell differentiation when in contact with mites. Adoptive transfer of EV-A71-trained BMDMs induced augmented allergic inflammation in naïve recipient mice, which was inhibited by 2-deoxy-D-glucose (2-DG) pretreatment, suggesting that trained macrophages following enterovirus infection are crucial in the progression of allergic asthma later in life.
Collapse
|
7
|
Jheng JR, Chen YS, Horng JT. Regulation of the proteostasis network during enterovirus infection: A feedforward mechanism for EV-A71 and EV-D68. Antiviral Res 2021; 188:105019. [PMID: 33484748 DOI: 10.1016/j.antiviral.2021.105019] [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: 10/13/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 10/25/2022]
Abstract
The proteostasis network guarantees successful protein synthesis, folding, transportation, and degradation. Mounting evidence has revealed that this network maintains proteome integrity and is linked to cellular physiology, pathology, and virus infection. Human enterovirus A71 (EV-A71) and EV-D68 are suspected causative agents of acute flaccid myelitis, a severe poliomyelitis-like neurologic syndrome with no known cure. In this context, further clarification of the molecular mechanisms underlying EV-A71 and EV-D68 infection is paramount. Here, we summarize the components of the proteostasis network that are intercepted by EV-A71 and EV-D68, as well as antivirals that target this network and may help develop improved antiviral drugs.
Collapse
Affiliation(s)
- Jia-Rong Jheng
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Yuan-Siao Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Jim-Tong Horng
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; Research Center for Industry of Human Ecology and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
8
|
Song C, Li Y, Zhou Y, Liang L, Turtle L, Wang F, Wu P, Qiu Q, Yang J, Wang K, Cui P, Cheng Y, Zhang T, Guo C, Zeng M, Long L, Peiris M, Zhou C, Cowling BJ, Yu H. Enterovirus genomic load and disease severity among children hospitalised with hand, foot and mouth disease. EBioMedicine 2020; 62:103078. [PMID: 33161231 PMCID: PMC7653080 DOI: 10.1016/j.ebiom.2020.103078] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/24/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Examining associations between viral genomic loads of enteroviruses and clinical severity is important for promoting and improving development of antiviral drugs related to hand, foot and mouth disease (HFMD). METHODS Throat swabs were collected from HFMD cases at acute phase of illness using a standardized technique in a prospective study. The viral genomic load was categorized into low, medium, and high groups using parameters of real-time reverse transcription-polymerase chain reaction. The clinical severities were assessed with four indicators, respectively. FINDINGS We analysed 1109 HFMD cases, including 538 children with CV-A6, 231 with CV-A16, 156 with EV-A71, 78 with CV-A10, 59 with CV-A4, and 47 with CV-A2. EV-A71 genomic load categories were associated with risks of diagnoses of CNS complications (p = 0.016), requiring systemic corticosteroids or IVIG (p = 0.011), intensive care unit admission (p = 0.002) and length of hospital stay over 5 days (p = 0.048). In the multivariate analyses, point estimates of adjusted odds ratio (OR) tended to increase with viral genomic loads for all four severe outcomes and ORs of highest viral genomic load were all significantly larger than one for EV-A71. INTERPRETATION HFMD clinical severities positively associate with viral genomic loads of EV-A71 in throat swabs. Specific antiviral drugs should be developed to reduce enterovirus load and to alleviate the clinical severities for HFMD cases. FUNDING National Science Fund for Distinguished Young Scholars.
Collapse
Affiliation(s)
- Chunlan Song
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou, China
| | - Yu Li
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yonghong Zhou
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Lu Liang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lance Turtle
- NIHR Health Protection Research Unit for Emerging and Zoonotic Infections, University of Liverpool, Liverpool, United Kingdom; Tropical & Infectious Disease Unit, Royal Liverpool University Hospital (member of Liverpool Health Partners), Liverpool, United Kingdom
| | - Fang Wang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou, China
| | - Peng Wu
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qi Qiu
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Jianli Yang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou, China
| | - Kai Wang
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Peng Cui
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Yibing Cheng
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou, China
| | - Tianchen Zhang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chun Guo
- School of Public Health, Huazhong University of Science and Technology, Wuhan, China
| | - Mengyao Zeng
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China; NHC key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Medical School, Fudan University, Shanghai, China
| | - Lu Long
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Malik Peiris
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chongchen Zhou
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou, China.
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hongjie Yu
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China.
| |
Collapse
|
9
|
|
10
|
Simon T, Cheuret E, Fiedler L, Mengelle C, Baudou E, Deiva K. Acute transverse myelitis following an opsoclonus-myoclonus syndrome: An unusual presentation. Eur J Paediatr Neurol 2018; 22:878-881. [PMID: 29773357 DOI: 10.1016/j.ejpn.2018.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 10/17/2022]
Abstract
Opso-myoclonus syndrome (OMS) is a very rare and severe condition. Ataxia, opsoclonus, myoclonus and/or behavioral and sleeping disturbances define that autoimmune disorder syndrome which is paraneoplastic or triggered by an infection. Here, we report a 3 year-old immunocompetent boy who developed an atypical OMS which was later complicated by an acute transverse myelitis. Screening for neuroblastoma was negative and extensive infectious screening revealed an active HHV-6 infection confirmed by blood and cerebrospinal fluid PCR. A parainfectious disease was suggested and immunosuppressive treatment was initiated. After 2 years of follow-up, the patient has a left leg paresia needing a splint and is otherwise normal. Transverse myelitis can be associated with parainfectious OMS and earlier immunosuppressive treatment in these cases may be useful especially in young and immunocompetent children.
Collapse
Affiliation(s)
- Thomas Simon
- Hôpital des enfants, Pediatric Neurology Department, 330 Avenue de grande Bretagne, 31000 Toulouse, France
| | - Emmanuel Cheuret
- Hôpital des enfants, Pediatric Neurology Department, 330 Avenue de grande Bretagne, 31000 Toulouse, France
| | - Léa Fiedler
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital Bicêtre, Pediatric Neurology Department, National Referral Center for Rare Inflammatory Brain and Spinal Diseases and Université Paris-Sud, UMR 1184-CEA-IDMIT, Center for Immunology of Viral Infections and Autoimmune Diseases, 94275, Le Kremlin Bicêtre, France
| | - Catherine Mengelle
- Hôpital Purpan, Virology Department, 330 Avenue de Grande Bretagne, 31000, Toulouse, France
| | - Eloïse Baudou
- Hôpital des enfants, Pediatric Neurology Department, 330 Avenue de grande Bretagne, 31000 Toulouse, France
| | - Kumaran Deiva
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital Bicêtre, Pediatric Neurology Department, National Referral Center for Rare Inflammatory Brain and Spinal Diseases and Université Paris-Sud, UMR 1184-CEA-IDMIT, Center for Immunology of Viral Infections and Autoimmune Diseases, 94275, Le Kremlin Bicêtre, France.
| |
Collapse
|
11
|
Flt3 ligand treatment reduces enterovirus A71 lethality in mice with enhanced B cell responses. Sci Rep 2018; 8:12184. [PMID: 30111869 PMCID: PMC6093920 DOI: 10.1038/s41598-018-30631-2] [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: 03/22/2018] [Accepted: 07/30/2018] [Indexed: 11/16/2022] Open
Abstract
Enterovirus A71 (EV-A71) infection can induce encephalitis, which causes death or long-term neurological sequelae, especially in young children. Using a murine infection model, we searched for anti-EV-A71 agents, because effective therapies are not available to control fatal infection. In EV-A71-infected mice, treatment with the hematopoietic growth factor, Fms-like tyrosine-kinase 3 ligand (Flt3 ligand) before infection reduced the lethality and tissue viral loads. Flt3 ligand failed to enhance the production of type I interferons. Instead, Flt3 ligand boosted the numbers of dendritic cells and, particularly lymphocytes in infected organs with an expansion of spleen B cells, and resulted in an increased titer of virus-specific antibody with neutralizing activity in the serum. The protective effect of Flt3 ligand was abolished in B cell-deficient mice. Our findings revealed that Flt3 ligand administration promotes resistance to EV-A71 infection with enhanced B cell response in a mechanism rarely reported before.
Collapse
|
12
|
Patabendige A, Michael BD, Craig AG, Solomon T. Brain microvascular endothelial-astrocyte cell responses following Japanese encephalitis virus infection in an in vitro human blood-brain barrier model. Mol Cell Neurosci 2018; 89:60-70. [PMID: 29635016 PMCID: PMC5984247 DOI: 10.1016/j.mcn.2018.04.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/16/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022] Open
Abstract
Japanese encephalitis virus (JEV) remains a leading cause of encephalitis, globally, which continues to grow in importance despite the availability of vaccines. Viral entry into the brain can occur via the blood-brain barrier (BBB), and inflammation at the BBB is a common final pathway in many brain infections. However, the role of the BBB during JEV infection and the contribution of the endothelial and astrocytic cell inflammation in facilitating virus entry into the brain are incompletely understood. We established a BBB model using human brain endothelial cells (HBECs) and human astrocytes. HBECs are polarised, and therefore the model was inoculated by JEV from the apical side to simulate the in vivo situation. The effects of JEV on the BBB permeability and release of inflammatory mediators from both apical and basolateral sides, representing the blood and the brain side respectively were investigated. JEV infected HBECs with limited active virus production, before crossing the BBB and infecting astrocytes. Control of JEV production by HBECs was associated with a significant increase in permeability, and with elevation of many host mediators, including cytokines, chemokines, cellular adhesion molecules, and matrix metalloproteases. When compared to the controls, significantly higher amounts of mediators were released from the apical side as opposed to the basolateral side. The increased release of mediators over time also correlated with increased BBB permeability. Treatment with dexamethasone led to a significant reduction in the release of interleukin 6 (IL6), C-C motif chemokine ligand 5 (CCL5) and C-X-C motif chemokine ligand 10 (CXCL10) from the apical side with a reduction in BBB disruption and no change in JEV production. The results are consistent with the hypothesis that JEV infection of the BBB triggers the production of a range of host mediators from both endothelial cells and astrocytes, which control JEV production but disrupt BBB integrity thus allowing virus entry into the brain. Dexamethasone treatment controlled the host response and limited BBB disruption in the model without increasing JEV production, supporting a re-investigation of its use therapeutically. Japanese encephalitis virus (JEV) infects human brain endothelial cells (HBECs). This triggers the production of a range of host mediators from both HBECs and astrocytes. JEV infection adversely affects blood-brain barrier (BBB) integrity. Dexamethasone treatment following JEV infection reduces the inflammation. Dexamethasone restores BBB integrity without increasing the levels of JEV particles.
Collapse
Affiliation(s)
- Adjanie Patabendige
- The Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; The School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, Australia; The Hunter Medical Research Institute, Newcastle, Australia.
| | - Benedict D Michael
- The Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; The Walton Centre NHS Foundation Trust, Liverpool, UK; Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, Harvard Medical School, USA
| | | | - Tom Solomon
- The Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; The Walton Centre NHS Foundation Trust, Liverpool, UK; National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
| |
Collapse
|
13
|
Wang LC, Yao HW, Chang CF, Wang SW, Wang SM, Chen SH. Suppression of interleukin-6 increases enterovirus A71 lethality in mice. J Biomed Sci 2017; 24:94. [PMID: 29233145 PMCID: PMC5726025 DOI: 10.1186/s12929-017-0401-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 12/06/2017] [Indexed: 12/27/2022] Open
Abstract
Background Enterovirus A71 (EV-A71) infection can induce fatal encephalitis in young children. Clinical reports show that interleukin-6 (IL-6) levels in the serum and cerebrospinal fluid of infected patients with brainstem encephalitis are significantly elevated. We used a murine model to address the significance of endogenous IL-6 in EV-A71 infection. Results EV-A71 infection transiently increased serum and brain IL-6 protein levels in mice. Most importantly, absence of IL-6 due to gene knockout or depletion of IL-6 using neutralizing monoclonal antibody enhanced the mortality and tissue viral load of infected mice. Absence of IL-6 increased the damage in the central nervous system and decreased the lymphocyte and virus-specific antibody responses of infected mice. Conclusions Endogenous IL-6 functions to clear virus and protect the host from EV-A71 infection. Our study raises caution over the use of anti-IL-6 antibody or pentoxifylline to reduce IL-6 for patient treatment. Electronic supplementary material The online version of this article (10.1186/s12929-017-0401-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Chiu Wang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China
| | - Hui-Wen Yao
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China
| | - Chuan-Fa Chang
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China.,Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China
| | - Shainn-Wei Wang
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China.,Institute of Molecular Medicine, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China
| | - Shih-Min Wang
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China.,Department of Pediatrics, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China
| | - Shun-Hua Chen
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China. .,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, 701, Republic of China.
| |
Collapse
|
14
|
Sahly A, Gauquelin L, Sébire G. Rapid Resolution of Enterovirus 71-Associated Opsoclonus Myoclonus Syndrome on Intravenous Immunoglobulin. Child Neurol Open 2017; 4:2329048X17733215. [PMID: 28975137 PMCID: PMC5613794 DOI: 10.1177/2329048x17733215] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 07/27/2017] [Accepted: 08/19/2017] [Indexed: 11/16/2022] Open
Abstract
Nonparaneoplastic opsoclonus–myoclonus ataxia syndrome is a rare neuroinflammatory condition featured by opsoclonus, myoclonus, ataxia, and cognitive behavioral disturbance. The authors report an observation of enterovirus 71-associated opsoclonus–myoclonus ataxia syndrome evolving toward full recovery on intravenous intravenous immunoglobulin (IG) treatment. Based on this case report, enterovirus 71 should be added to the list of infectious agents likely involved in opsoclonus–myoclonus ataxia syndrome, including the emerging subgroup of opsoclonus–myoclonus ataxia syndrome recovering without aggressive or prolonged immunosuppressive intervention. Further studies are mandatory to define the precise role, incidence, treatment, and outcome of enterovirus 71 and other infectious agents in benign forms of opsoclonus–myoclonus ataxia syndrome.
Collapse
Affiliation(s)
- Ahmed Sahly
- Division of Child Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Québec, Canada.,King Abdulaziz University, Jeddah, Saudi Arabia
| | - Laurence Gauquelin
- Division of Child Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Québec, Canada
| | - Guillaume Sébire
- Division of Child Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Québec, Canada
| |
Collapse
|
15
|
Zhong T, Zhang LY, Wang ZY, Wang Y, Song FM, Zhang YH, Yu JH. Rheum emodin inhibits enterovirus 71 viral replication and affects the host cell cycle environment. Acta Pharmacol Sin 2017; 38:392-401. [PMID: 27840410 PMCID: PMC5342659 DOI: 10.1038/aps.2016.110] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/02/2016] [Indexed: 12/21/2022] Open
Abstract
Human enterovirus 71 (EV71) is the primary causative agent of recent large-scale outbreaks of hand, foot, and mouth disease (HFMD) in Asia. Currently, there are no drugs available for the prevention and treatment of HFMD. In this study, we compared the anti-EV71 activities of three natural compounds, rheum emodin, artemisinin and astragaloside extracted from Chinese herbs Chinese rhubarb, Artemisia carvifolia and Astragalus, respectively, which have been traditionally used for the treatment and prevention of epidemic diseases. Human lung fibroblast cell line MRC5 was mock-infected or infected with EV71, and treated with drugs. The cytotoxicity of the drugs was detected with MTT assay. The cytopathic effects such as cell death and condensed nuclei were morphologically observed. The VP1-coding sequence required for EV71 genome replication was assayed with qRT-PCR. Viral protein expression was analyzed with Western blotting. Viral TCID50 was determined to evaluate EV71 virulence. Flow cytometry analysis of propidium iodide staining was performed to analyze the cell cycle distribution of MRC5 cells. Rheum emodin (29.6 μmol/L) effectively protected MRC5 cells from EV71-induced cytopathic effects, which resulted from the inhibiting viral replication: rheum emodin treatment decreased viral genomic levels by 5.34-fold, viral protein expression by less than 30-fold and EV71 virulence by 0.33107-fold. The fact that inhibition of rheum emodin on viral virulence was much stronger than its effects on genomic levels and viral protein expression suggested that rheum emodin inhibited viral maturation. Furthermore, rheum emodin treatment markedly diminished cell cycle arrest at S phase in MRC5 cells, which was induced by EV71 infection and favored the viral replication. In contrast, neither astragaloside (50 μmol/L) nor artemisinin (50 μmol/L) showed similar anti-EV71 activities. Among the three natural compounds tested, rheum emodin effectively suppressed EV71 viral replication, thus is a candidate anti-HFMD drug.
Collapse
|
16
|
Thymidine Kinase-Negative Herpes Simplex Virus 1 Can Efficiently Establish Persistent Infection in Neural Tissues of Nude Mice. J Virol 2017; 91:JVI.01979-16. [PMID: 27974554 DOI: 10.1128/jvi.01979-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/05/2016] [Indexed: 01/28/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) establishes latency in neural tissues of immunocompetent mice but persists in both peripheral and neural tissues of lymphocyte-deficient mice. Thymidine kinase (TK) is believed to be essential for HSV-1 to persist in neural tissues of immunocompromised mice, because infectious virus of a mutant with defects in both TK and UL24 is detected only in peripheral tissues, but not in neural tissues, of severe combined immunodeficiency mice (T. Valyi-Nagy, R. M. Gesser, B. Raengsakulrach, S. L. Deshmane, B. P. Randazzo, A. J. Dillner, and N. W. Fraser, Virology 199:484-490, 1994, https://doi.org/10.1006/viro.1994.1150). Here we find infiltration of CD4 and CD8 T cells in peripheral and neural tissues of mice infected with a TK-negative mutant. We therefore investigated the significance of viral TK and host T cells for HSV-1 to persist in neural tissues using three genetically engineered mutants with defects in only TK or in both TK and UL24 and two strains of nude mice. Surprisingly, all three mutants establish persistent infection in up to 100% of brain stems and 93% of trigeminal ganglia of adult nude mice at 28 days postinfection, as measured by the recovery of infectious virus. Thus, in mouse neural tissues, host T cells block persistent HSV-1 infection, and viral TK is dispensable for the virus to establish persistent infection. Furthermore, we found 30- to 200-fold more virus in neural tissues than in the eye and detected glycoprotein C, a true late viral antigen, in brainstem neurons of nude mice persistently infected with the TK-negative mutant, suggesting that adult mouse neurons can support the replication of TK-negative HSV-1. IMPORTANCE Acyclovir is used to treat herpes simplex virus 1 (HSV-1)-infected immunocompromised patients, but treatment is hindered by the emergence of drug-resistant viruses, mostly those with mutations in viral thymidine kinase (TK), which activates acyclovir. TK mutants are detected in brains of immunocompromised patients with persistent infection. However, answers to the questions as to whether TK-negative (TK-) HSV-1 can establish persistent infection in brains of immunocompromised hosts and whether neurons in vivo are permissive for TK- HSV-1 remain elusive. Using three genetically engineered HSV-1 TK- mutants and two strains of nude mice deficient in T cells, we found that all three HSV-1 TK- mutants can efficiently establish persistent infection in the brain stem and trigeminal ganglion and detected glycoprotein C, a true late viral antigen, in brainstem neurons. Our study provides evidence that TK- HSV-1 can persist in neural tissues and replicate in brain neurons of immunocompromised hosts.
Collapse
|
17
|
Mostafa HH, Vogel P, Srinivasan A, Russell CJ. Non-invasive Imaging of Sendai Virus Infection in Pharmacologically Immunocompromised Mice: NK and T Cells, but not Neutrophils, Promote Viral Clearance after Therapy with Cyclophosphamide and Dexamethasone. PLoS Pathog 2016; 12:e1005875. [PMID: 27589232 PMCID: PMC5010285 DOI: 10.1371/journal.ppat.1005875] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/17/2016] [Indexed: 11/19/2022] Open
Abstract
In immunocompromised patients, parainfluenza virus (PIV) infections have an increased potential to spread to the lower respiratory tract (LRT), resulting in increased morbidity and mortality. Understanding the immunologic defects that facilitate viral spread to the LRT will help in developing better management protocols. In this study, we immunosuppressed mice with dexamethasone and/or cyclophosphamide then monitored the spread of viral infection into the LRT by using a noninvasive bioluminescence imaging system and a reporter Sendai virus (murine PIV type 1). Our results show that immunosuppression led to delayed viral clearance and increased viral loads in the lungs. After cessation of cyclophosphamide treatment, viral clearance occurred before the generation of Sendai-specific antibody responses and coincided with rebounds in neutrophils, T lymphocytes, and natural killer (NK) cells. Neutrophil suppression using anti-Ly6G antibody had no effect on infection clearance, NK-cell suppression using anti-NK antibody delayed clearance, and T-cell suppression using anti-CD3 antibody resulted in no clearance (chronic infection). Therapeutic use of hematopoietic growth factors G-CSF and GM-CSF had no effect on clearance of infection. In contrast, treatment with Sendai virus-specific polysera or a monoclonal antibody limited viral spread into the lungs and accelerated clearance. Overall, noninvasive bioluminescence was shown to be a useful tool to study respiratory viral progression, revealing roles for NK and T cells, but not neutrophils, in Sendai virus clearance after treatment with dexamethasone and cyclophosphamide. Virus-specific antibodies appear to have therapeutic potential.
Collapse
Affiliation(s)
- Heba H. Mostafa
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Peter Vogel
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ashok Srinivasan
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Charles J. Russell
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Department of Microbiology, Immunology & Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Hand-foot-and-mouth disease (HFMD) is a common cause of viral rash in children with classic skin findings which are easily recognized by pediatricians. Recently, several atypical cutaneous manifestations of HFMD have been described. Awareness of these patterns may lead providers to appropriate diagnosis and management. This review also highlights the epidemiological patterns of more virulent strains and emerging research in disease prevention. RECENT FINDINGS Classic HFMD presents with tender lesions on the hands, feet, and oral mucosa. Atypical skin findings in HFMD may be seen in children with atopic dermatitis. These include 'eczema coxsackium', in which eczematous skin is superinfected with coxsackie virus, resembling herpes infection. Nail changes, such as shedding, may follow HFMD after a latency period. Enterovirus 71 is responsible for epidemic outbreaks of HFMD in Asia, with systemic manifestations and occasionally neurological sequelae. Research is underway to develop a vaccine which could curb epidemics, but for the present, supportive care and hygiene measures are the standard of care. SUMMARY Atypical manifestations of HFMD in children with atopic dermatitis may mimic herpetic superinfection. In a child presenting with nail changes, consider antecedent HFMD in the differential diagnosis. The mainstay of treatment for HMFD remains supportive care.
Collapse
|