Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.449
Filter
Add more filters

Publication year range
1.
Cell ; 169(5): 862-877.e17, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28502771

ABSTRACT

Herpes zoster (shingles) causes significant morbidity in immune compromised hosts and older adults. Whereas a vaccine is available for prevention of shingles, its efficacy declines with age. To help to understand the mechanisms driving vaccinal responses, we constructed a multiscale, multifactorial response network (MMRN) of immunity in healthy young and older adults immunized with the live attenuated shingles vaccine Zostavax. Vaccination induces robust antigen-specific antibody, plasmablasts, and CD4+ T cells yet limited CD8+ T cell and antiviral responses. The MMRN reveals striking associations between orthogonal datasets, such as transcriptomic and metabolomics signatures, cell populations, and cytokine levels, and identifies immune and metabolic correlates of vaccine immunity. Networks associated with inositol phosphate, glycerophospholipids, and sterol metabolism are tightly coupled with immunity. Critically, the sterol regulatory binding protein 1 and its targets are key integrators of antibody and T follicular cell responses. Our approach is broadly applicable to study human immunity and can help to identify predictors of efficacy as well as mechanisms controlling immunity to vaccination.


Subject(s)
Herpes Zoster Vaccine/immunology , Adaptive Immunity , Adult , Aged , Aging , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Gene Expression Profiling , Gene Regulatory Networks , Humans , Inositol Phosphates/immunology , Longitudinal Studies , Male , Metabolomics , Middle Aged , Sex Characteristics , Sterols/metabolism , Viral Load
2.
EMBO J ; 41(14): e109217, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35670106

ABSTRACT

Varicella-Zoster virus (VZV) causes chickenpox and shingles. Although the infection is associated with severe morbidity in some individuals, molecular mechanisms that determine innate immune responses remain poorly defined. We found that the cGAS/STING DNA sensing pathway was required for type I interferon (IFN) induction during VZV infection and that recognition of VZV by cGAS restricted its replication. Screening of a VZV ORF expression library identified the essential VZV tegument protein ORF9 as a cGAS antagonist. Ectopically or virally expressed ORF9 bound to endogenous cGAS leading to reduced type I IFN responses to transfected DNA. Confocal microscopy revealed co-localisation of cGAS and ORF9. ORF9 and cGAS also interacted directly in a cell-free system and phase-separated together with DNA. Furthermore, ORF9 inhibited cGAMP production by cGAS. Taken together, these results reveal the importance of the cGAS/STING DNA sensing pathway for VZV recognition and identify a VZV immune antagonist that partially but directly interferes with DNA sensing via cGAS.


Subject(s)
Herpesvirus 3, Human , Interferon Type I , Nucleotidyltransferases , Viral Proteins , DNA/metabolism , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/immunology , Humans , Immunity, Innate , Interferon Type I/immunology , Membrane Proteins/immunology , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/immunology , Viral Proteins/immunology
3.
J Virol ; 98(4): e0060323, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38517165

ABSTRACT

Herpesviruses replicate by cleaving concatemeric dsDNA into single genomic units that are packaged through an oligomeric portal present in preformed procapsids. In contrast to what is known about phage portal proteins, details concerning herpesvirus portal structure and function are not as well understood. A panel of 65 Varicella-Zoster virus (VZV) recombinant portal proteins with five amino acid in-frame insertions were generated by random transposon mutagenesis of the VZV portal gene, ORF54. Subsequently, 65 VZVLUC recombinant viruses (TNs) were generated via recombineering. Insertions were mapped to predicted portal domains (clip, wing, stem, wall, crown, and ß-hairpin tunnel-loop) and recombinant viruses were characterized for plaque morphology, replication kinetics, pORF54 expression, and classified based on replication in non-complementing (ARPE19) or complementing (ARPE54C50) cell lines. The N- and C-termini were tolerant to insertion mutagenesis, as were certain clip sub-domains. The majority of mutants mapping to the wing, wall, ß-hairpin tunnel loop, and stem domains were lethal. Elimination of the predicted ORF54 start codon revealed that the first 40 amino acids of the N-terminus were not required for viral replication. Stop codon insertions in the C-terminus showed that the last 100 amino acids were not required for viral replication. Lastly, a putative protease cleavage site was identified in the C-terminus of pORF54. Cleavage was likely orchestrated by a viral protease; however, processing was not required for DNA encapsidation and viral replication. The panel of recombinants should prove valuable in future studies to dissect mammalian portal structure and function.IMPORTANCEThough nucleoside analogs and a live-attenuated vaccine are currently available to treat some human herpesvirus family members, alternate methods of combating herpesvirus infection could include blocking viral replication at the DNA encapsidation stage. The approval of Letermovir provided proof of concept regarding the use of encapsidation inhibitors to treat herpesvirus infections in the clinic. We propose that small-molecule compounds could be employed to interrupt portal oligomerization, assembly into the capsid vertex, or affect portal function/dynamics. Targeting portal at any of these steps would result in disruption of viral DNA packaging and a decrease or absence of mature infectious herpesvirus particles. The oligomeric portals of herpesviruses are structurally conserved, and therefore, it may be possible to find a single compound capable of targeting portals from one or more of the herpesvirus subfamilies. Drug candidates from such a series would be effective against viruses resistant to the currently available antivirals.


Subject(s)
Herpesviridae Infections , Herpesvirus 3, Human , Animals , Humans , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/metabolism , Mutagenesis , Virus Replication , Herpesviridae Infections/genetics , DNA/metabolism , Amino Acids/genetics , Mammals/genetics
4.
J Virol ; : e0084824, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39051773

ABSTRACT

Varicella zoster virus (VZV) reactivates from ganglionic sensory neurons to produce herpes zoster (shingles) in a unilateral dermatomal distribution, typically in the thoracic region. Reactivation not only heightens the risk of stroke and other neurological complications but also increases susceptibility to co-infections with various viral and bacterial pathogens at sites distant from the original infection. The mechanism by which VZV results in complications remote from the initial foci remains unclear. Small extracellular vesicles (sEVs) are membranous signaling structures that can deliver proteins and nucleic acids to modify the function of distal cells and tissues during normal physiological conditions. Although viruses have been documented to exploit the sEV machinery to propagate infection, the role of non-infectious sEVs released from VZV-infected neurons in viral spread and disease has not been studied. Using multi-omic approaches, we characterized the content of sEVs released from VZV-infected human sensory neurons (VZV sEVs). One viral protein was detected (immediate-early 62), as well as numerous immunosuppressive and vascular disease-associated host proteins and miRNAs that were absent in sEVs from uninfected neurons. Notably, VZV sEVs are non-infectious yet transcriptionally altered primary human cells, suppressing the antiviral type 1 interferon response and promoting neuroinvasion of a secondary pathogen in vivo. These results challenge our understanding of VZV infection, proposing that the virus may contribute to distant pathologies through non-infectious sEVs beyond the primary infection site. Furthermore, this study provides a previously undescribed immune-evasion mechanism induced by VZV that highlights the significance of non-infectious sEVs in early VZV pathogenesis. IMPORTANCE: Varicella zoster virus (VZV) is a ubiquitous human virus that predominantly spreads by direct cell-cell contact and requires efficient and immediate host immune evasion strategies to spread. The mechanisms of immune evasion prior to virion entry have not been fully elucidated and represent a critical gap in our complete understanding of VZV pathogenesis. This study describes a previously unreported antiviral evasion strategy employed by VZV through the exploitation of the infected host cell's small extracellular vesicle (sEV) machinery. These findings suggest that non-infectious VZV sEVs could travel throughout the body, affecting cells remote from the site of infection and challenging the current understanding of VZV clinical disease, which has focused on local effects and direct infection. The significance of these sEVs in early VZV pathogenesis highlights the importance of further investigating their role in viral spread and secondary disease development to reduce systemic complications following VZV infections.

5.
Rev Med Virol ; 34(4): e2554, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38862398

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.


Subject(s)
Herpesvirus 3, Human , Humans , Herpesvirus 3, Human/immunology , Herpesvirus 3, Human/physiology , Herpesvirus 3, Human/pathogenicity , Herpes Zoster/virology , Herpes Zoster/immunology , Varicella Zoster Virus Infection/immunology , Varicella Zoster Virus Infection/virology , Nervous System Diseases/virology , Nervous System Diseases/immunology , Nervous System Diseases/etiology , Animals , Chickenpox/virology , Chickenpox/immunology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/virology
6.
Rev Med Virol ; 34(4): e2556, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38853706

ABSTRACT

Stroke is a common worldwide cause of death and disability, resulting from an obstruction or reduction in blood flow to the brain. Research has demonstrated that systemic infection such as herpes zoster (HZ) / ophthalmicus herpes zoster (HZO) can potentially trigger stroke. This study includes an updated systematic review and meta-analysis of the epidemiologic data on the connection between HZ/HZO infection and the risk of stroke. A meticulous search of different database yielded 905 studies. Furthermore, an additional 14 studies from a previous meta-analysis were incorporated. Eligible studies underwent rigorous screening, resulting in 18 papers. Statistical analyses, including random/fixed effects models and subgroup analyses, were conducted to assess pooled relative risk (RR) and heterogeneity. The meta-analysis consisted of 5,505,885 participants and found a statistically significant association between HZ infection and the risk of stroke (pooled RR = 1.22, 95% confidence interval [CI] 1.12-1.34). The HZO infection showed a significantly higher overall pooled RR of 1.71 (95% CI 1.06-2.75), indicating a strong connection with the risk of stroke. Subgroup analysis revealed that the odds ratio might play a significant role in causing heterogeneity. Time since infection emerged as a crucial factor, with heightened stroke risk in the initial year post-HZ/HZO exposure, followed by a decline after the first year. Asian/Non-Asian studies demonstrated varied results in HZ/HZO patients. Meta-analysis reveals a significant HZ/HZO-stroke link. Subgroups highlight varied risks and warrant extended Asian/non-Asian patient investigation.


Subject(s)
Herpes Zoster , Stroke , Humans , Stroke/epidemiology , Stroke/virology , Herpes Zoster/epidemiology , Herpes Zoster/virology , Herpes Zoster/complications , Risk Assessment , Risk Factors , Herpesvirus 3, Human
7.
Rev Med Virol ; 34(3): e2538, 2024 May.
Article in English | MEDLINE | ID: mdl-38658176

ABSTRACT

Serious adverse events following vaccination include medical complications that require hospitalisation. The live varicella vaccine that was approved by the Food and Drug Administration in the United States in 1995 has an excellent safety record. Since the vaccine is a live virus, adverse events are more common in immunocompromised children who are vaccinated inadvertently. This review includes only serious adverse events in children considered to be immunocompetent. The serious adverse event called varicella vaccine meningitis was first reported in a hospitalised immunocompetent child in 2008. When we carried out a literature search, we found 15 cases of immunocompetent children and adolescents with varicella vaccine meningitis; the median age was 11 years. Eight of the children had received two varicella vaccinations. Most of the children also had a concomitant herpes zoster rash, although three did not. The children lived in the United States, Greece, Germany, Switzerland, and Japan. During our literature search, we found five additional cases of serious neurological events in immunocompetent children; these included 4 cases of progressive herpes zoster and one case of acute retinitis. Pulses of enteral corticosteroids as well as a lack of herpes simplex virus antibody may be risk factors for reactivation in immunocompetent children. All 20 children with adverse events were treated with acyclovir and recovered; 19 were hospitalised and one child was managed as an outpatient. Even though the number of neurological adverse events remains exceedingly low following varicella vaccination, we recommend documentation of those caused by the vaccine virus.


Subject(s)
Chickenpox Vaccine , Meningitis, Viral , Adolescent , Child , Child, Preschool , Female , Humans , Male , Acyclovir/therapeutic use , Antiviral Agents/therapeutic use , Chickenpox/prevention & control , Chickenpox/virology , Chickenpox Vaccine/administration & dosage , Chickenpox Vaccine/adverse effects , Chickenpox Vaccine/immunology , Herpesvirus 3, Human/immunology , Meningitis, Viral/virology , Nervous System Diseases/virology , Nervous System Diseases/etiology , Vaccination/adverse effects , Virus Activation/drug effects
8.
J Infect Dis ; 229(1): 64-72, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-37410908

ABSTRACT

BACKGROUND: The incidence of herpes zoster (HZ) has increased in the United States concurrent with decrease in herpes simplex virus (HSV) prevalence. We hypothesized that lack of HSV-elicited cross-reactive immunity to varicella-zoster virus (VZV) results in an increased risk of HZ. Using specimens from the placebo arm of the Shingles Prevention Study, we investigated whether persons who develop HZ are less likely to have prior HSV infection than persons who do not develop HZ, and whether HZ is less severe in persons with HSV than in HSV seronegative persons. METHODS: We conducted a nested case-control (1:2) study comparing the seroprevalence of HSV-1 and HSV-2 in cases (persons with polymerase chain reaction-confirmed HZ) to age-, sex-, and health-matched controls (persons without HZ). RESULTS: Sera from 639 study participants (213 cases and 426 controls) yielded definitive HSV antibody results and were analyzed. Overall, HSV seropositivity rate was 75%. HSV seronegativity was significantly higher in HZ cases than controls (30.5% vs 22.3%; P = .024), with a 55% higher risk of HZ in HSV seronegative than HSV seropositive participants. HSV seropositivity was associated with more severe HZ (P = .021). CONCLUSIONS: Our study demonstrated that prior infection with HSV partly protects against HZ.


Subject(s)
Herpes Simplex , Herpes Zoster , Herpesvirus 1, Human , Humans , Herpes Simplex/complications , Herpes Simplex/epidemiology , Herpesvirus 3, Human , Seroepidemiologic Studies , Male , Female
9.
J Infect Dis ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38195164

ABSTRACT

The varicella-zoster virus (VZV) infects over 95% of the population. VZV reactivation causes herpes zoster (HZ), known as shingles, primarily affecting the elderly and immunocompromised individuals. However, HZ can also occur in otherwise healthy individuals. We analyzed the immune signature and risk profile in HZ patients using a genome-wide association study across different UK Biobank HZ cohorts. Additionally, we conducted one of the largest HZ HLA association studies to date, coupled with transcriptomic analysis of pathways underlying HZ susceptibility. Our findings highlight the significance of the MHC locus for HZ development, identifying five protective and four risk HLA alleles. This demonstrates that HZ susceptibility is largely governed by variations in the MHC. Furthermore, functional analyses revealed the upregulation of type I interferon and adaptive immune responses. These findings provide fresh molecular insights into the pathophysiology and the activation of innate and adaptive immune responses triggered by symptomatic VZV reactivation.

10.
Clin Infect Dis ; 78(4): 880-888, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38015658

ABSTRACT

BACKGROUND: Postherpetic neuralgia (PHN) is the most common chronic complication of herpes zoster (HZ) and results in severe refractory neuropathic pain. This study aimed at evaluating the efficacy of premedication with duloxetine in the prevention of PHN. METHODS: The PROCESS trial is a multicenter, randomized, open-label, blinded-endpoint trial used a 1:1 duloxetine:control ratio. Adults 50 years or older with HZ who presented with vesicles within 72 hours were recruited. The primary outcome was the incidence of PHN at 12 weeks. PHN was defined as any pain intensity score other than 0 mm on the visual analog scale (VAS) at week 12 after the onset of the rash. The secondary outcomes were the number of participants with VAS >0 and VAS ≥3. The modified intention-to-treat (mITT) principle and per-protocol (PP) principle were used for the primary outcome analysis. RESULTS: A total of 375 participants were randomly assigned to the duloxetine group and 375 were assigned to the control group. There was no significant difference in the incidence of PHN in the duloxetine group compared with the control group in the mITT analysis (86 [22.9%] of 375 vs 108 [28.8%] of 375; P = .067). PP analysis produced similar results. However, there were significant differences between the 2 groups in the number of participants with VAS >0 and VAS ≥3 (P < .05 for all comparisons). CONCLUSIONS: Although absolute prevention of PHN does not occur, this trial found that premedication with duloxetine can reduce pain associated with HZ, and therefore can have clinically relevant benefits. Clinical Trials Registration. Clinicaltrials.gov, NCT04313335. Registered on 18 March 2020.


Subject(s)
Herpes Zoster , Neuralgia, Postherpetic , Adult , Humans , Neuralgia, Postherpetic/drug therapy , Neuralgia, Postherpetic/prevention & control , Neuralgia, Postherpetic/epidemiology , Duloxetine Hydrochloride/therapeutic use , Herpes Zoster/complications , Herpes Zoster/drug therapy , Herpes Zoster/prevention & control , Herpesvirus 3, Human , Pain Measurement/adverse effects , Pain Measurement/methods
11.
Am J Transplant ; 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39009348

ABSTRACT

Recently, live-attenuated measles, rubella, varicella, and mumps vaccines have been administered to carefully selected post-liver transplant patients. Although attention has been focused on post-vaccination antibody titers and adverse events, the real-life clinical benefits remain unclear. A comprehensive analysis of breakthrough infections and natural boosters (asymptomatic cases with significant elevation in virus antibody titers) following immunization post-liver transplantation was conducted from 2002-2023, exploring the timing, frequency, correlation with domestic outbreaks, and degree of antibody elevation. During the median 10-year observation period among 68 post-liver transplant patients, breakthrough infections occurred only in chickenpox, with 7 mild cases (1 episode/64 person-years). A total of 59 natural booster episodes (1, 5, 20, and 33 for measles, rubella, chickenpox, and mumps, respectively) were observed, with incidence rates of 1 per 569, 110, 22, and 17 person-years, respectively. The timing of natural boosters closely correlated with domestic outbreaks (P < .05 in chickenpox and mumps), influenced by local vaccine coverage. The degree of antibody elevation was significantly higher in individuals with breakthrough infections than in those with natural boosters (P < .05). These findings suggest that immunization with live-attenuated vaccines for post-liver transplant patients has demonstrated clinical benefits. Furthermore, mass vaccination has a positive impact on post-transplant patient outcomes.

12.
Br J Haematol ; 204(5): 1617-1634, 2024 May.
Article in English | MEDLINE | ID: mdl-38532527

ABSTRACT

Vaccinations are fundamental tools in preventing infectious diseases, especially in immunocompromised patients like those affected by non-Hodgkin lymphomas (NHLs). The COVID-19 pandemic made clinicians increasingly aware of the importance of vaccinations in preventing potential life-threatening SARS-CoV-2-related complications in NHL patients. However, several studies have confirmed a significant reduction in vaccine-induced immune responses after anti-CD20 monoclonal antibody treatment, thus underscoring the need for refined immunization strategies in NHL patients. In this review, we summarize the existing data about COVID-19 and other vaccine's efficacy in patients with NHL and propose multidisciplinary team-based recommendations for the management of vaccines in this specific group of patients.


Subject(s)
COVID-19 , Lymphoma, Non-Hodgkin , SARS-CoV-2 , Vaccination , Humans , Lymphoma, Non-Hodgkin/therapy , Lymphoma, Non-Hodgkin/immunology , COVID-19/prevention & control , COVID-19/immunology , COVID-19/complications , SARS-CoV-2/immunology , Immunocompromised Host , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/therapeutic use
13.
J Clin Microbiol ; 62(6): e0010324, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38785446

ABSTRACT

The monkeypox virus (MPXV) outbreak, primarily endemic to Africa, has spread globally, with Brazil reporting the second-highest number of cases. The emergence of MPXV in non-endemic areas has raised concerns, particularly due to the co-circulation of other exanthematous viruses such as varicella-zoster virus (VZV) and molluscum contagiosum virus (MOCV). To perform an accurate differential diagnosis of MPXV during the ongoing outbreak in Minas Gerais, Brazil, a 5PLEX qPCR assay targeting orthopoxviruses (OPV), VZV, and MOCV was used to retrospectively analyze all clinical samples that tested negative for MPXV in the initial screening conducted at Funed. In summary, our study analyzed 1,175 clinical samples received from patients suspected of MPXV infection and found a positivity rate of 33.8% (397 samples) for MPXV using the non-variola qPCR assay. Testing the 778 MPXV-negative clinical samples using the 5PLEX qPCR assay revealed that 174 clinical samples (22.36%) tested positive for VZV. MOCV DNA was detected in 13 and other OPV in 3 clinical samples. The sequencing of randomly selected amplified clinical samples confirmed the initial molecular diagnosis. Analysis of patient profiles revealed a significant difference in the median age between groups testing positive for MPXV and VZV and a male predominance in MPXV cases. The geographic distribution of positive cases was concentrated in the most populous mesoregions of Minas Gerais state. This study highlights the challenges posed by emerging infectious diseases. It emphasizes the importance of epidemiological surveillance and accurate diagnosis in enabling timely responses for public health policies and appropriate medical care. IMPORTANCE: Brazil ranks second in the number of cases during the global monkeypox epidemic. The study, conducted in Minas Gerais, the second most populous state in Brazil with over 20 million inhabitants, utilized differential diagnostics, revealing a significant number of positive cases for other exanthematous viruses and emphasizing the need for accurate diagnoses. During the study, we were able to assess the co-circulation of other viruses alongside monkeypox, including varicella-zoster virus, molluscum contagiosum virus, and other orthopoxviruses. The significance of the research is underscored by the concentration of positive cases in populous areas, highlighting the challenges posed by emerging infectious diseases. This demographic context further amplifies the importance of the research in guiding public health policies and medical interventions, given the substantial population at risk. The study not only addresses a global concern but also holds critical implications for a state with such a large population and geographic expanse within Brazil. Overall, the study emphasizes the pivotal role of surveillance and precise diagnosis in guiding effective public health responses and ensuring appropriate medical interventions.


Subject(s)
Disease Outbreaks , Humans , Brazil/epidemiology , Retrospective Studies , Male , Female , Adult , Diagnosis, Differential , Child , Adolescent , Mpox (monkeypox)/diagnosis , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/virology , Young Adult , Child, Preschool , Middle Aged , Monkeypox virus/genetics , Monkeypox virus/isolation & purification , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/isolation & purification , Infant , Aged , Exanthema/virology , Exanthema/epidemiology , Real-Time Polymerase Chain Reaction
14.
Mamm Genome ; 35(2): 296-307, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38600211

ABSTRACT

Varicella-zoster virus (VZV), a common pathogen with humans as the sole host, causes primary infection and undergoes a latent period in sensory ganglia. The recurrence of VZV is often accompanied by severe neuralgia in skin tissue, which has a serious impact on the life of patients. During the acute infection of VZV, there are few related studies on the pathophysiological mechanism of skin tissue. In this study, transcriptome sequencing data from the acute response period within 2 days of VZV antigen stimulation of the skin were used to explore a model of the trajectory of skin tissue changes during VZV infection. It was found that early VZV antigen stimulation caused activation of mainly natural immune-related signaling pathways, while in the late phase activation of mainly active immune-related signaling pathways. JAK-STAT, NFκB, and TNFα signaling pathways are gradually activated with the progression of infection, while Hypoxia is progressively inhibited. In addition, we found that dendritic cell-mediated immune responses play a dominant role in the lesion damage caused by VZV antigen stimulation of the skin. This study provides a theoretical basis for the study of the molecular mechanisms of skin lesions during acute VZV infection.


Subject(s)
Herpesvirus 3, Human , Signal Transduction , Skin , Varicella Zoster Virus Infection , Herpesvirus 3, Human/genetics , Skin/pathology , Skin/virology , Skin/immunology , Animals , Varicella Zoster Virus Infection/virology , Varicella Zoster Virus Infection/immunology , Varicella Zoster Virus Infection/genetics , Varicella Zoster Virus Infection/pathology , Humans , Mice , Dendritic Cells/immunology , Herpes Zoster/virology , Herpes Zoster/pathology , Herpes Zoster/genetics , Herpes Zoster/immunology , Transcriptome , Disease Models, Animal , Antigens, Viral/immunology , Antigens, Viral/genetics , NF-kappa B/metabolism , NF-kappa B/genetics
15.
J Med Virol ; 96(5): e29667, 2024 May.
Article in English | MEDLINE | ID: mdl-38738524

ABSTRACT

The effectiveness of herpes zoster (HZ) vaccines in patients with diabetes over the age of 50 remains an active area of research. Utilizing a real-world database from the US community, this study spanning from 2006 to 2023, aimed to evaluate the impact of HZ vaccination on newly diagnosed diabetes patients who received an HZ vaccination within 1 year of diagnosis. Exclusion criteria were established to omit patients with immune deficiencies. The cohort consisted of 53 885 patients, with an average age of 63.5 years, including 43% females and 58% whites. After implementing 1:1 propensity score matching for age, sex, race, comorbidities, diabetes medication, and hemoglobin A1c to ensure comparability, the study population was further stratified into four groups: N1 comparing any HZ vaccination to non-HZ vaccination (53 882 matched pairs), N2 for Shingrix versus non-HZ vaccination (16 665 matched pairs), N3 for Zostavax versus non-HZ vaccination (12 058 matched pairs), and N4 for Shingrix versus Zostavax (11 721 matched pairs). Cox proportional hazards regression analysis revealed a hazard ratio (HR) for HZ incidence post any HZ vaccination of 0.92 (95% confidence interval [CI]: 0.83-1.01). Additional analyses yielded HRs of 1.12 (95% CI: 0.93-1.34) for Shingrix versus non-HZ vaccine, 1.02 (95% CI: 0.86-1.20) for Zostavax versus non-HZ vaccine, and 1.06 (95% CI: 0.87-1.29) for Shingrix versus Zostavax. Subgroup analyses across age, sex, and follow-up duration also showed no significant differences. These findings underscore the lack of a significant benefit from HZ vaccination in newly diagnosed diabetes patients aged over 50, highlighting the necessity for further prospective research.


Subject(s)
Herpes Zoster Vaccine , Herpes Zoster , Humans , Female , Male , Herpes Zoster Vaccine/immunology , Herpes Zoster Vaccine/administration & dosage , Middle Aged , Herpes Zoster/prevention & control , Herpes Zoster/epidemiology , Aged , Cohort Studies , Diabetes Mellitus , Vaccine Efficacy , Vaccination/statistics & numerical data , Aged, 80 and over , Proportional Hazards Models , United States/epidemiology , Herpesvirus 3, Human/immunology
16.
J Med Virol ; 96(5): e29629, 2024 May.
Article in English | MEDLINE | ID: mdl-38682607

ABSTRACT

To inform surveillance, prevention, and management strategies for the varicella zoster virus (VZV) during the COVID-19 pandemic, this study aimed to evaluate the risk of herpes zoster (HZ) occurrence/recurrence following COVID-19 infection and vaccination. A comprehensive search across seven databases was conducted up to January 31, 2024, to identify studies relevant to the occurrence of HZ following COVID-19 infection and vaccination. The meta-analysis included five studies on postinfection HZ and 13 studies on postvaccination HZ. Patients infected with COVID-19 had a 2.16-fold increased risk of HZ (95% confidence interval [CI]: 1.24-3.76) than uninfected individuals. However, there was no significant association between COVID-19 vaccination and the risk of HZ compared to controls, with a relative risk (RR) of 1.08 (95% CI: 0.84-1.39). Furthermore, a descriptive analysis of 74 postinfection and 153 postvaccination HZ studies found no significant differences on gender or age (<50 and ≥50 years) following COVID-19 infection. Notably, 44.0% of the HZ cases postinfection appeared within the first week, with 69.5% resolving within 10 days, predominantly presenting as skin lesions. In the postvaccination group, the majority (60.1%) developed HZ after the first dose and 66.7% occurred within 1 week. Moreover, 44.6% resolved within 10 days and 50.0% within a month, primarily exhibiting skin lesions and postherpetic neuralgia. The study found that COVID-19 infection increases the risk of HZ, but the COVID-19 vaccine does not. Further study is needed to explore the association between COVID-19 and HZ.


Subject(s)
COVID-19 Vaccines , COVID-19 , Herpes Zoster , Recurrence , Vaccination , Humans , Herpes Zoster/epidemiology , Herpes Zoster/prevention & control , COVID-19/prevention & control , COVID-19/epidemiology , Vaccination/statistics & numerical data , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , Herpesvirus 3, Human/immunology , Middle Aged , Female
17.
J Med Virol ; 96(6): e29690, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38804180

ABSTRACT

Autophagy is a degradational pathway with pivotal roles in cellular homeostasis and survival, including protection of neurons in the central nervous system (CNS). The significance of autophagy as antiviral defense mechanism is recognized and some viruses hijack and modulate this process to their advantage in certain cell types. Here, we present data demonstrating that the human neurotropic herpesvirus varicella zoster virus (VZV) induces autophagy in human SH-SY5Y neuronal cells, in which the pathway exerts antiviral activity. Productively VZV-infected SH-SY5Y cells showed increased LC3-I-LC3-II conversion as well as co-localization of the viral glycoprotein E and the autophagy receptor p62. The activation of autophagy was dependent on a functional viral genome. Interestingly, inducers of autophagy reduced viral transcription, whereas inhibition of autophagy increased viral transcript expression. Finally, the genotype of patients with severe ocular and brain VZV infection were analyzed to identify potential autophagy-associated inborn errors of immunity. Two patients expressing genetic variants in the autophagy genes ULK1 and MAP1LC3B2, respectively, were identified. Notably, cells of both patients showed reduced autophagy, alongside enhanced viral replication and death of VZV-infected cells. In conclusion, these results demonstrate a neuro-protective role for autophagy in the context of VZV infection and suggest that failure to mount an autophagy response is a potential predisposing factor for development of severe VZV disease.


Subject(s)
Autophagy , Herpesvirus 3, Human , Neurons , Humans , Herpesvirus 3, Human/physiology , Herpesvirus 3, Human/pathogenicity , Neurons/virology , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Virus Replication , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Varicella Zoster Virus Infection/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Cell Line , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Host-Pathogen Interactions
18.
J Neurovirol ; 30(1): 86-99, 2024 02.
Article in English | MEDLINE | ID: mdl-38453879

ABSTRACT

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.


Subject(s)
Amyloid beta-Peptides , Cytokines , Macaca mulatta , Animals , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/blood , Cytokines/cerebrospinal fluid , Cytokines/blood , Virus Activation , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/blood , Varicellovirus/genetics , Varicellovirus/immunology , Herpesvirus 3, Human/pathogenicity , Herpesvirus 3, Human/immunology , Herpesviridae Infections/cerebrospinal fluid , Herpesviridae Infections/virology , Herpesviridae Infections/blood , Herpesviridae Infections/immunology , Male , Herpes Zoster/cerebrospinal fluid , Herpes Zoster/virology , Herpes Zoster/blood , Herpes Zoster/immunology , Monkey Diseases/virology , Monkey Diseases/cerebrospinal fluid , Monkey Diseases/blood
19.
J Neurovirol ; 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38381256

ABSTRACT

Sleep problems was associated with increased risk for herpes zoster (HZ). This study examined subjects with insomnia or a combination of insomnia and depression and their risk of HZ. This retrospective cohort study included a total of 47,256 participants, with a control comprising 31,504 age- and sex-matched patients. Clinical data from 2000 to 2013 in the Taiwan National Health Insurance Research Database were used for analysis. Insomnia, depression, and HZ were defined according to the International Classification of Diseases, Ninth Revision, Clinical Modification. Subjects with insomnia had a significantly higher incidence of HZ (2.77 per 1000 person-years) than the controls (1.81 per 1000 person-years) as well as a higher risk of developing HZ (adjusted hazard ratio (AHR) = 1.62, 95% confidence interval (CI) = 1.35-1.93). Results shown subjects with insomnia durations of < 4 years, 4-6 years, and > 6 years had a significantly higher risk of HZ compared with the controls (AHR: 6.69, 95% CI 4.44-9.39; AHR: 4.42, 95% CI 3.07-6.36; AHR:1.38, 95% CI 1.14-1.87, respectively). We found a significantly higher risk of HZ in subjects with both insomnia and depression (AHR = 4.95; 95% CI = 3.99-7.02) than in those without related conditions. Patients with insomnia, and even more so those with comorbid depression, had a higher risk of developing HZ. This indicates a joint effect of insomnia and depression on HZ.

20.
Curr Top Microbiol Immunol ; 438: 247-272, 2023.
Article in English | MEDLINE | ID: mdl-34224015

ABSTRACT

The cerebral arteries are innervated by afferent fibers from the trigeminal ganglia. Varicella-zoster virus (VZV) frequently resides in the trigeminal ganglion. Reports of arterial ischemic stroke due to VZV cerebral vasculopathy in adults after herpes zoster have been described for decades. Reports of arterial ischemic stroke due to post-varicella cerebral arteriopathy in children have also been described for decades. One rationale for this review has been post-licensure studies that have shown an apparent protective effect from stroke in both adults who have received live zoster vaccine and children who have received live varicella vaccine. In this review, we define common features between stroke following varicella in children and stroke following herpes zoster in adults. The trigeminal ganglion and to a lesser extent the superior cervical ganglion are central to the stroke pathogenesis pathway because afferent fibers from these two ganglia provide the circuitry by which the virus can travel to the anterior and posterior circulations of the brain. Based on studies in pseudorabies virus (PRV) models, it is likely that VZV is carried to the cerebral arteries on a kinesin motor via gE, gI and the homolog of PRV US9. The gE product is an essential VZV protein.


Subject(s)
Chickenpox , Herpes Zoster , Ischemic Stroke , Stroke , Adult , Child , Humans , Herpesvirus 3, Human , Chickenpox/prevention & control , Trigeminal Ganglion/pathology , Stroke/pathology
SELECTION OF CITATIONS
SEARCH DETAIL