Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.027
Filtrer
1.
Mol Biol Evol ; 41(7)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38879872

RÉSUMÉ

Antiviral therapy is constantly challenged by the emergence of resistant pathogens. At the same time, experimental approaches to understand and predict resistance are limited by long periods required for evolutionary processes. Here, we present a herpes simplex virus 1 mutant with impaired proofreading capacity and consequently elevated mutation rates. Comparing this hypermutator to parental wild type virus, we study the evolution of antiviral drug resistance in vitro. We model resistance development and elucidate underlying genetic changes against three antiviral substances. Our analyzes reveal no principle difference in the evolutionary behavior of both viruses, adaptive processes are overall similar, however significantly accelerated for the hypermutator. We conclude that hypermutator viruses are useful for modeling adaptation to antiviral therapy. They offer the benefit of expedited adaptation without introducing apparent bias and can therefore serve as an accelerator to predict natural evolution.


Sujet(s)
Antiviraux , Résistance virale aux médicaments , Évolution moléculaire , Herpèsvirus humain de type 1 , Résistance virale aux médicaments/génétique , Antiviraux/pharmacologie , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/effets des médicaments et des substances chimiques , Mutation , Taux de mutation , Évolution biologique , Humains
2.
Vopr Virusol ; 69(2): 187-192, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38843024

RÉSUMÉ

INTRODUCTION: Herpes simplex virus type 1 (HSV-1) is one of the most common human viral infections and has a double-stranded DNA genome belonging to the Herpesviridae family. Smoking is one of the leading causes of disease and premature death worldwide, responsible for the death of up to six million people annually. The purpose of the current study was to determine the seroprevalence of HSV-1 infection among smokers. Methods. The search strategy was conducted in the period from December 2022 to January 2023. The study included a random sample of 94 (88 males, and 6 females) healthy participants, aged between ≤ 20 to ≥ 60 years, with 50 participants as the control group. The HSV serological testing consisted of detecting antibodies to HSV-1 IgG with the help of ELISA. RESULTS: Most participants were university students, consisting of 45.7% males and 5.3% females, followed by employed smokers, consisting of 0.2% males and 1.1% females. The number of females was much lower than that of males reaching 6.4 and 93.6% respectively, due to customs and traditions. The seroprevalence was 24.47, 22.3 and 2.1% in males and females respectively. The seroprevalence rate was 13.8% in hookah and cigarette smokers, 9% in cigarette smokers and 1.1% in hookah smokers exclusively. The highest rate was observed in the age groups of 21-30 and 31-40 years with 12.80% and 7.40% respectively. CONCLUSIONS: The study revealed that the seroprevalence of HSV-1 IgG was 24.47%, and was higher among hookah and cigarette smokers compared to those who exclusively smoked cigarettes or hookah.


Sujet(s)
Anticorps antiviraux , Herpès , Herpèsvirus humain de type 1 , Fumeurs , Humains , Mâle , Herpèsvirus humain de type 1/immunologie , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/isolement et purification , Femelle , Études séroépidémiologiques , Adulte , Adulte d'âge moyen , Herpès/épidémiologie , Herpès/virologie , Herpès/sang , Anticorps antiviraux/sang , Immunoglobuline G/sang , Jeune adulte , Fumer/épidémiologie , Sujet âgé , Adolescent
3.
PLoS Pathog ; 20(6): e1012307, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38857310

RÉSUMÉ

Multiple functions are associated with HSV-1 latency associated transcript (LAT), including establishment of latency, virus reactivation, and antiapoptotic activity. LAT encodes two sncRNAs that are not miRNAs and previously it was shown that they have antiapoptotic activity in vitro. To determine if we can separate the antiapoptotic function of LAT from its latency-reactivation function, we deleted sncRNA1 and sncRNA2 sequences in HSV-1 strain McKrae, creating ΔsncRNA1&2 recombinant virus. Deletion of the sncRNA1&2 in ΔsncRNA1&2 virus was confirmed by complete sequencing of ΔsncRNA1&2 virus and its parental virus. Replication of ΔsncRNA1&2 virus in tissue culture or in the eyes of WT infected mice was similar to that of HSV-1 strain McKrae (LAT-plus) and dLAT2903 (LAT-minus) viruses. The levels of gB DNA in trigeminal ganglia (TG) of mice latently infected with ΔsncRNA1&2 virus was intermediate to that of dLAT2903 and McKrae infected mice, while levels of LAT in TG of latently infected ΔsncRNA1&2 mice was significantly higher than in McKrae infected mice. Similarly, the levels of LAT expression in Neuro-2A cells infected with ΔsncRNA1&2 virus was significantly higher than in McKrae infected cells. Reactivation in TG of ΔsncRNA1&2 infected mice was similar to that of McKrae and time of reactivation in both groups were significantly faster than dLAT2903 infected mice. However, levels of apoptosis in Neuro-2A cells infected with ΔsncRNA1&2 virus was similar to that of dLAT2903 and significantly higher than that of McKrae infected cells. Our results suggest that the antiapoptotic function of LAT resides within the two sncRNAs, which works independently of its latency-reactivation function and it has suppressive effect on LAT expression in vivo and in vitro.


Sujet(s)
Apoptose , Herpèsvirus humain de type 1 , Neurones , Activation virale , Latence virale , Animaux , Souris , Herpèsvirus humain de type 1/physiologie , Herpèsvirus humain de type 1/génétique , Activation virale/physiologie , Neurones/virologie , Neurones/métabolisme , Latence virale/physiologie , ARN viral/génétique , ARN viral/métabolisme , Petit ARN non traduit/génétique , Petit ARN non traduit/métabolisme , Cellules cultivées , Femelle , microARN
4.
Nucleic Acids Res ; 52(12): 7292-7304, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38806233

RÉSUMÉ

Herpes simplex virus 1 (HSV-1), a double-stranded DNA virus, replicates using seven essential proteins encoded by its genome. Among these, the UL30 DNA polymerase, complexed with the UL42 processivity factor, orchestrates leading and lagging strand replication of the 152 kb viral genome. UL30 polymerase is a prime target for antiviral therapy, and resistance to current drugs can arise in immunocompromised individuals. Using electron cryo-microscopy (cryo-EM), we unveil the dynamic changes of the UL30/UL42 complex with DNA in three distinct states. First, a pre-translocation state with an open fingers domain ready for nucleotide incorporation. Second, a halted elongation state where the fingers close, trapping dATP in the dNTP pocket. Third, a DNA-editing state involving significant conformational changes to allow DNA realignment for exonuclease activity. Additionally, the flexible UL30 C-terminal domain interacts with UL42, forming an extended positively charged surface binding to DNA, thereby enhancing processive synthesis. These findings highlight substantial structural shifts in the polymerase and its DNA interactions during replication, offering insights for future antiviral drug development.


Sujet(s)
Cryomicroscopie électronique , ADN viral , DNA-directed DNA polymerase , Herpèsvirus humain de type 1 , Protéines virales , DNA-directed DNA polymerase/métabolisme , DNA-directed DNA polymerase/composition chimique , DNA-directed DNA polymerase/génétique , Protéines virales/métabolisme , Protéines virales/composition chimique , Protéines virales/ultrastructure , Herpèsvirus humain de type 1/enzymologie , Herpèsvirus humain de type 1/génétique , ADN viral/métabolisme , ADN viral/biosynthèse , Réplication de l'ADN , Holoenzymes/composition chimique , Holoenzymes/métabolisme , Modèles moléculaires , Réplication virale , Liaison aux protéines , Exodeoxyribonucleases
5.
Biochem Biophys Res Commun ; 718: 149931, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38723415

RÉSUMÉ

Oncolytic viruses (OVs) have shown potential in converting a "cold" tumor into a "hot" one and exhibit effectiveness in various cancer types. However, only a subset of patients respond to oncolytic virotherapy. It is important to understand the resistance mechanisms to OV treatment in pancreatic ductal adenocarcinoma (PDAC) to engineer oncolytic viruses. In this study, we used transcriptome RNA sequencing (RNA-seq) to identify Visfatin, which was highly expressed in the responsive tumors following OV treatment. To explore the antitumor efficacy, we modified OV-mVisfatin, which effectively inhibited tumor growth. For the first time, we revealed that Visfatin promoted the antitumor efficacy of OV by remodeling the tumor microenvironment, which involved enhancing CD8+ T cell and DC cell infiltration and activation, repolarizing macrophages towards the M1-like phenotype, and decreasing Treg cells using single-cell RNA sequencing (scRNA-seq) and flow cytometry. Furthermore, PD-1 blockade significantly enhanced OV-mVisfatin antitumor efficacy, offering a promising new therapeutic strategy for PDAC.


Sujet(s)
Herpèsvirus humain de type 1 , Nicotinamide phosphoribosyltransferase , Thérapie virale de cancers , Virus oncolytiques , Tumeurs du pancréas , Microenvironnement tumoral , Animaux , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Souris , Thérapie virale de cancers/méthodes , Nicotinamide phosphoribosyltransferase/génétique , Nicotinamide phosphoribosyltransferase/métabolisme , Herpèsvirus humain de type 1/génétique , Lignée cellulaire tumorale , Virus oncolytiques/génétique , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Souris de lignée C57BL , Humains , Lymphocytes T CD8+/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , Femelle
6.
J Virol ; 98(6): e0071224, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38780246

RÉSUMÉ

Within the first 15 minutes of infection, herpes simplex virus 1 immediate early proteins repurpose cellular RNA polymerase (Pol II) for viral transcription. An important role of the viral-infected cell protein 27 (ICP27) is to facilitate viral pre-mRNA processing and export viral mRNA to the cytoplasm. Here, we use precision nuclear run-on followed by deep sequencing (PRO-seq) to characterize transcription of a viral ICP27 null mutant. At 1.5 and 3 hours post infection (hpi), we observed increased total levels of Pol II on the mutant viral genome and accumulation of Pol II downstream of poly A sites indicating increased levels of initiation and processivity. By 6 hpi, Pol II accumulation on specific mutant viral genes was higher than that on wild-type virus either at or upstream of poly A signals, depending on the gene. The PRO-seq profile of the ICP27 mutant on late genes at 6 hpi was similar but not identical to that caused by treatment with flavopiridol, a known inhibitor of RNA processivity. This pattern was different from PRO-seq profiles of other α gene mutants and upon inhibition of viral DNA replication with PAA. Together, these results indicate that ICP27 contributes to the repression of aberrant viral transcription at 1.5 and 3 hpi by inhibiting initiation and decreasing RNA processivity. However, ICP27 is needed to enhance processivity on most late genes by 6 hpi in a mechanism distinguishable from its role in viral DNA replication.IMPORTANCEWe developed and validated the use of a processivity index for precision nuclear run-on followed by deep sequencing data. The processivity index calculations confirm infected cell protein 27 (ICP27) induces downstream of transcription termination on certain host genes. The processivity indices and whole gene probe data implicate ICP27 in transient immediate early gene-mediated repression, a process that also requires ICP4, ICP22, and ICP0. The data indicate that ICP27 directly or indirectly regulates RNA polymerase (Pol II) initiation and processivity on specific genes at specific times post infection. These observations support specific and varied roles for ICP27 in regulating Pol II activity on viral genes in addition to its known roles in post transcriptional mRNA processing and export.


Sujet(s)
Génome viral , Herpèsvirus humain de type 1 , Protéines précoces immédiates , Réplication virale , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/physiologie , Protéines précoces immédiates/génétique , Protéines précoces immédiates/métabolisme , Humains , Mutation , RNA polymerase II/métabolisme , RNA polymerase II/génétique , ARN viral/génétique , ARN viral/métabolisme , Transcription virale/génétique , Animaux , Régulation de l'expression des gènes viraux , Cellules Vero , Chlorocebus aethiops , Herpès/virologie , Herpès/génétique , ARN messager/génétique , ARN messager/métabolisme
7.
J Dermatolog Treat ; 35(1): 2350232, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38724041

RÉSUMÉ

BACKGROUND/PURPOSE: Dystrophic epidermolysis bullosa (DEB), a rare genetic skin disease caused by loss-of-function mutations in COL7A1, the gene encoding type VII collagen (COL7), is characterized by skin blistering, scarring, and extracutaneous manifestations that markedly reduce patient quality-of-life. Beremagene geperpavec-svdt ('B-VEC') is a gene therapy employing a non-integrating, replication-defective herpes simplex virus type 1 (HSV-1)-based vector encoding two copies of full-length human COL7A1 to restore COL7 protein after topical administration to DEB wounds. B-VEC was approved in the United States in 2023 as the first topical gene therapy and the first approved treatment for DEB. However, few providers have experience with use of this gene therapy. METHODS: Data was obtained through literature review and the experience of providers who participated in the B-VEC clinical study or initiated treatment after B-VEC approval. RESULTS: This review discusses the burden of disease, describes the clinical trial outcomes of B-VEC, and provides physician and patient/caregiver recommendations as a practical guide for the real-world use of B-VEC, which can be administered in-office or at the patient's home. CONCLUSIONS: By continuing to optimize the practical aspects of B-VEC administration, the focus will continue to shift to patient-centric considerations and improved patient outcomes.


Sujet(s)
Collagène de type VII , Épidermolyse bulleuse dystrophique , Thérapie génétique , Humains , Épidermolyse bulleuse dystrophique/thérapie , Épidermolyse bulleuse dystrophique/génétique , Collagène de type VII/génétique , Vecteurs génétiques , Herpèsvirus humain de type 1/génétique , Résultat thérapeutique , Qualité de vie
8.
Nat Commun ; 15(1): 3969, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38730242

RÉSUMÉ

Encephalitis is a rare and potentially fatal manifestation of herpes simplex type 1 infection. Following genome-wide genetic analyses, we identified a previously uncharacterized and very rare heterozygous variant in the E3 ubiquitin ligase WWP2, in a 14-month-old girl with herpes simplex encephalitis. The p.R841H variant (NM_007014.4:c.2522G > A) impaired TLR3 mediated signaling in inducible pluripotent stem cells-derived neural precursor cells and neurons; cells bearing this mutation were also more susceptible to HSV-1 infection compared to control cells. The p.R841H variant increased TRIF ubiquitination in vitro. Antiviral immunity was rescued following the correction of p.R841H by CRISPR-Cas9 technology. Moreover, the introduction of p.R841H in wild type cells reduced such immunity, suggesting that this mutation is linked to the observed phenotypes.


Sujet(s)
Encéphalite à herpès simplex , Herpèsvirus humain de type 1 , Mutation , Ubiquitin-protein ligases , Humains , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Femelle , Encéphalite à herpès simplex/génétique , Nourrisson , Herpèsvirus humain de type 1/génétique , Cellules souches pluripotentes induites/métabolisme , Récepteur de type Toll-3/génétique , Récepteur de type Toll-3/métabolisme , Ubiquitination , Neurones/métabolisme , Cellules souches neurales/métabolisme , Cellules souches neurales/virologie , Systèmes CRISPR-Cas
9.
Nat Commun ; 15(1): 3669, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38693119

RÉSUMÉ

Oncolytic viruses (OVs) show promise as a cancer treatment by selectively replicating in tumor cells and promoting antitumor immunity. However, the current immunogenicity induced by OVs for tumor treatment is relatively weak, necessitating a thorough investigation of the mechanisms underlying its induction of antitumor immunity. Here, we show that HSV-1-based OVs (oHSVs) trigger ZBP1-mediated PANoptosis (a unique innate immune inflammatory cell death modality), resulting in augmented antitumor immune effects. Mechanistically, oHSV enhances the expression of interferon-stimulated genes, leading to the accumulation of endogenous Z-RNA and subsequent activation of ZBP1. To further enhance the antitumor potential of oHSV, we conduct a screening and identify Fusobacterium nucleatum outer membrane vesicle (Fn-OMV) that can increase the expression of PANoptosis execution proteins. The combination of Fn-OMV and oHSV demonstrates potent antitumor immunogenicity. Taken together, our study provides a deeper understanding of oHSV-induced antitumor immunity, and demonstrates a promising strategy that combines oHSV with Fn-OMV.


Sujet(s)
Fusobacterium nucleatum , Herpèsvirus humain de type 1 , Thérapie virale de cancers , Virus oncolytiques , Protéines de liaison à l'ARN , Herpèsvirus humain de type 1/immunologie , Herpèsvirus humain de type 1/génétique , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Animaux , Humains , Thérapie virale de cancers/méthodes , Souris , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/immunologie , Lignée cellulaire tumorale , Fusobacterium nucleatum/immunologie , Tumeurs/thérapie , Tumeurs/immunologie , Femelle , Immunité innée , Souris de lignée BALB C
10.
Front Cell Infect Microbiol ; 14: 1383811, 2024.
Article de Anglais | MEDLINE | ID: mdl-38808062

RÉSUMÉ

Introduction: While astrocytes participate in the CNS innate immunity against herpes simplex virus type 1 (HSV-1) infection, they are the major target for the virus. Therefore, it is of importance to understand the interplay between the astrocyte-mediated immunity and HSV-1 infection. Methods: Both primary human astrocytes and the astrocyte line (U373) were used in this study. RT-qPCR and Western blot assay were used to measure IFNs, the antiviral IFN-stimulated genes (ISGs), IFN regulatory factors (IRFs) and HSV-1 DNA. IRF1 knockout or knockdown was performed with CRISPR/Cas9 and siRNA transfection techniques. Results: Poly(dA:dT) could inhibit HSV-1 replication and induce IFN-ß/IFN-λs production in human astrocytes. Poly(dA:dT) treatment of astrocytes also induced the expression of the antiviral ISGs (Viperin, ISG56 and MxA). Among IRFs members examined, poly(dA:dT) selectively unregulated IRF1 and IRF9, particularly IRF1 in human astrocytes. The inductive effects of poly(dA:dT) on IFNs and ISGs were diminished in the IRF1 knockout cells. In addition, IRF1 knockout attenuated poly(dA:dT)-mediated HSV-1 inhibition in the cells. Conclusion: The DNA sensors activation induces astrocyte intracellular innate immunity against HSV-1. Therefore, targeting the DNA sensors has potential for immune activation-based HSV-1 therapy.


Sujet(s)
Astrocytes , Herpèsvirus humain de type 1 , Facteur-1 de régulation d'interféron , Réplication virale , Humains , Astrocytes/virologie , Astrocytes/métabolisme , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Herpèsvirus humain de type 1/immunologie , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/physiologie , Immunité innée , Poly DA-DT , Herpès/immunologie , Herpès/virologie , Cytosol/métabolisme , Lignée cellulaire , Cellules cultivées , ADN viral/génétique , Techniques de knock-out de gènes
11.
J Med Case Rep ; 18(1): 257, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38778387

RÉSUMÉ

BACKGROUND: Mucosal melanoma, an aggressive type of malignancy different from the cutaneous melanomas commonly seen in the head and neck region, represents < 1% of all malignant melanomas. The pathogenesis of mucosal melanoma is unknown. Targetable mutations commonly seen in cutaneous melanoma, such as in the BRAF and NRAS genes, have a lower incidence in mucosal melanoma. Mucosal melanoma carries a distinct mutational pattern from cutaneous melanoma. Surgery with negative margins is the first-line treatment for mucosal melanoma, and systemic therapy is not well defined. Talimogene laherparepvec, an oncolytic viral immunotherapy, is United States Food and Drug Administration approved for the treatment of advanced malignant cutaneous melanoma, with local therapeutic benefits. Mucosal melanoma was initially excluded from talimogene laherparepvec's initial phase III clinical trial. CASE PRESENTATION: We present the case of a white female patient in her 40s with past medical history of systemic lupus erythematous, scleroderma, and estrogen-receptor-positive invasive ductal breast carcinoma. Following a bilateral mastectomy, the patient was found to have BRAF-negative mucosal melanoma of her hard palate with a soft palate skip lesion. Owing to the presence of a skip mucosal lesion as well as the anticipated defect and need for free-flap reconstructive surgery, nonsurgical management was considered. The patient was referred to medical oncology, where-based on the patient's complicated medical history and the risk of immunotherapy possibly worsening her prior autoimmune diseases-local talimogene laherparepvec injections were chosen as the primary therapy for her mucosal lesions. Though talimogene laherparepvec is approved for the treatment of cutaneous melanoma, there are limited data available on the use of talimogene laherparepvec in mucosal melanomas. CONCLUSION: The patient had a complete local tumor response at both the primary lesion as well as the skip lesion with the local injections. She had no side effects and maintained a high quality of life during treatment.


Sujet(s)
Produits biologiques , Mélanome , Humains , Mélanome/thérapie , Femelle , Produits biologiques/usage thérapeutique , Produits biologiques/administration et posologie , Adulte , Herpèsvirus humain de type 1/génétique , Muqueuse de la bouche/anatomopathologie , Injections intralésionnelles , Résultat thérapeutique , Antinéoplasiques immunologiques/usage thérapeutique , Antinéoplasiques immunologiques/administration et posologie , Thérapie virale de cancers/méthodes , Tumeurs du palais/thérapie
12.
J Hematol Oncol ; 17(1): 36, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38783389

RÉSUMÉ

Oncolytic viruses (OVs) offer a novel approach to treat solid tumors; however, their efficacy is frequently suboptimal due to various limiting factors. To address this challenge, we engineered an OV containing targets for neuron-specific microRNA-124 and Granulocyte-macrophage colony-stimulating factor (GM-CSF), significantly enhancing its neuronal safety while minimally compromising its replication capacity. Moreover, we identified PARP1 as an HSV-1 replication restriction factor using genome-wide CRISPR screening. In models of glioblastoma (GBM) and triple-negative breast cancer (TNBC), we showed that the combination of OV and a PARP inhibitor (PARPi) exhibited superior efficacy compared to either monotherapy. Additionally, single-cell RNA sequencing (scRNA-seq) revealed that this combination therapy sensitized TNBC to immune checkpoint blockade, and the incorporation of an immune checkpoint inhibitor (ICI) further increased the survival rate of tumor-bearing mice. The combination of PARPi and ICI synergistically enhanced the ability of OV to establish durable tumor-specific immune responses. Our study effectively overcomes the inherent limitations of OV therapy, providing valuable insights for the clinical treatment of TNBC, GBM, and other malignancies.


Sujet(s)
Thérapie virale de cancers , Thérapie virale de cancers/méthodes , Animaux , Humains , Souris , Facteur de stimulation des colonies de granulocytes et de macrophages/génétique , Glioblastome/thérapie , Glioblastome/génétique , Virus oncolytiques/génétique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du sein triple-négatives/thérapie , Tumeurs du sein triple-négatives/génétique , Femelle , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Herpèsvirus humain de type 1/génétique , Lignée cellulaire tumorale , Clustered regularly interspaced short palindromic repeats/génétique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , microARN/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Systèmes CRISPR-Cas
13.
J Infect Public Health ; 17(7): 102456, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38820896

RÉSUMÉ

BACKGROUND: A. baumannii is an important and common clinical pathogen, especially in the intensive care unit (ICU). This study aimed to characterize one hypervirulent A. baumannii strain in a patient with community-acquired pneumonia and herpes simplex type 1 virus infection. METHODS: Minimum inhibitory concentrations (MICs) were determined using the Kirby-Bauer (K-B) and broth microdilution methods. Galleria mellonella infection model experiment was conducted. Whole-genome sequencing (WGS) was performed using the Illumina and Nanopore platforms. The resistance and virulence determinants were identified using the ABRicate program with ResFinder and the VFDB database. The capsular polysaccharide locus (K locus) and lipooligosaccharide outer core locus (OC locus) were identified using Kleborate with Kaptive. Phylogenetic analyses were conducted using the BacWGSTdb server. RESULTS: A. baumannii XH2146 strain belongs to ST10Pas and ST447Oxf. The strain was resistant to cefazolin, ciprofloxacin, and trimethoprim/sulfamethoxazole (TMP-SMX). Bautype and Kaptive analyses showed that XH2146 contains OCL2 and KL49. WGS analysis revealed that the strain harbored blaADC-76, blaOXA-68, ant(3'')-IIa, tet(B), and sul2. Notably, tet(B) and sul2, both were located within a 114,700-bp plasmid (designated pXH2146-1). Virulence assay revealed A. baumannii XH2146 possessed higher virulence than A. baumannii AB5075 at 12 h. Comparative genomic analysis showed that A. baumannii ST447 strains were mainly isolated from the USA and exhibited a relatively close genetic relationship. Importantly, 11 strains were observed to carry blaOXA-58; blaOXA-23 was identified in 11 isolates and three ST447 A. baumannii strains harbored blaNDM-1. CONCLUSIONS: Early detection of community-acquired hypervirulent Acinetobacter baumannii strains is recommended to prevent their extensive spread in hospitals.


Sujet(s)
Infections à Acinetobacter , Acinetobacter baumannii , Infections communautaires , Herpèsvirus humain de type 1 , Tests de sensibilité microbienne , Phylogenèse , Séquençage du génome entier , Infections communautaires/microbiologie , Infections communautaires/épidémiologie , Humains , Acinetobacter baumannii/génétique , Acinetobacter baumannii/pathogénicité , Acinetobacter baumannii/effets des médicaments et des substances chimiques , Acinetobacter baumannii/isolement et purification , Chine/épidémiologie , Infections à Acinetobacter/microbiologie , Infections à Acinetobacter/épidémiologie , Animaux , Virulence/génétique , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/pathogénicité , Herpèsvirus humain de type 1/isolement et purification , Antibactériens/pharmacologie , Facteurs de virulence/génétique , Herpès/virologie , Pneumopathie bactérienne/microbiologie , Mâle , Génome bactérien , Papillons de nuit/microbiologie , Papillons de nuit/virologie
14.
PLoS One ; 19(5): e0295088, 2024.
Article de Anglais | MEDLINE | ID: mdl-38776332

RÉSUMÉ

Yaws affects children in tropical regions, while syphilis primarily affects sexually active adults worldwide. Despite various campaigns towards the eradication of yaws and elimination of syphilis, these two diseases are still present in Ghana. The aetiological agents of both diseases, two Treponema pallidum subspecies, are genetically similar. This study aimed to assess the prevalence of these treponematoses and the occurrence of pathogens causing similar skin lesions in the Ashanti region of Ghana. A point-of-care test was used to determine the seroprevalence of the treponematoses. Both yaws and syphilis were identified in the Ashanti region of Ghana. Multiplex PCR was used to identify treponemes and other pathogens that cause similar skin lesions. The results indicated that the seroprevalences of T. pallidum in individuals with yaws-like and syphilis-like lesions were 17.2% and 10.8%, respectively. Multiplex PCR results showed that 9.1%, 1.8% and 0.9% of yaws-like lesions were positive for Haemophilus ducreyi, herpes simplex virus-1 (HSV-1) and T. pallidum respectively. Among syphilis-like lesions, 28.3% were positive for herpes simplex virus -2 (HSV-2) by PCR. To our knowledge, this is the first time HSV-I and HSV-2 have been reported from yaws-like and syphilis-like lesions, respectively, in Ghana. The presence of other organisms apart from T. pallidum in yaws-like and syphilis-like lesions could impede the total healing of these lesions and the full recovery of patients. This may complicate efforts to achieve yaws eradication by 2030 and the elimination of syphilis and warrants updated empirical treatment guidelines for skin ulcer diseases.


Sujet(s)
Haemophilus ducreyi , Syphilis , Treponema pallidum , Pian , Humains , Ghana/épidémiologie , Pian/épidémiologie , Pian/microbiologie , Syphilis/épidémiologie , Syphilis/microbiologie , Femelle , Adulte , Mâle , Haemophilus ducreyi/isolement et purification , Haemophilus ducreyi/génétique , Adolescent , Prévalence , Treponema pallidum/génétique , Treponema pallidum/isolement et purification , Enfant , Jeune adulte , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/isolement et purification , Adulte d'âge moyen , Études séroépidémiologiques , Peau/microbiologie , Peau/anatomopathologie , Peau/virologie , Enfant d'âge préscolaire , Tréponématoses/épidémiologie , Tréponématoses/microbiologie
15.
Virol J ; 21(1): 102, 2024 05 02.
Article de Anglais | MEDLINE | ID: mdl-38698421

RÉSUMÉ

Human parechovirus, a member of the Picornaviridae family (PeVs), can lead to severe infections, including severe meningitis, meningoencephalitis, and sepsis-like syndrome. We report a case of human parechovirus-related encephalitis in a 52-year-old woman diagnosed with glioblastoma multiforme. She underwent surgical resection in June 2022. Unfortunately, her disease recurred, and she underwent a second resection in August 2022, followed by radiation therapy and Temozolomide therapy. She presented to the hospital with acute confusion followed by seizures, necessitating intubation for airway support. A cerebrospinal fluid (CSF) sample was obtained and processed using the Biofire FilmArray, which reported the detection of HSV-1. Despite being on Acyclovir, the patient did not show signs of improvement. Consequently, a second CSF sample was obtained and sent for next-generation sequencing (NGS), which returned a positive result for Parechovirus. In this presented case, the patient exhibited symptoms of an unknown infectious cause. The utilization of NGS and metagenomic analysis helped identify Parechovirus as the primary pathogen present, in addition to previously identified HSV. This comprehensive approach facilitated a thorough assessment of the underlying infection and guided targeted treatment. In conclusion, the application of NGS techniques and metagenomic analysis proved instrumental in identifying the root cause of the infection.


Sujet(s)
Sujet immunodéprimé , Parechovirus , Infections à Picornaviridae , Humains , Femelle , Adulte d'âge moyen , Infections à Picornaviridae/virologie , Infections à Picornaviridae/diagnostic , Parechovirus/génétique , Parechovirus/isolement et purification , Parechovirus/classification , Arabie saoudite , Séquençage nucléotidique à haut débit , Glioblastome/virologie , Métagénomique , Encéphalite virale/virologie , Encéphalite virale/diagnostic , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/isolement et purification , Hospitalisation
16.
Nat Commun ; 15(1): 4018, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38740820

RÉSUMÉ

Anti-HSV therapies are only suppressive because they do not eliminate latent HSV present in ganglionic neurons, the source of recurrent disease. We have developed a potentially curative approach against HSV infection, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Gene editing performed with two anti-HSV-1 meganucleases delivered by a combination of AAV9, AAV-Dj/8, and AAV-Rh10 can eliminate 90% or more of latent HSV DNA in mouse models of orofacial infection, and up to 97% of latent HSV DNA in mouse models of genital infection. Using a pharmacological approach to reactivate latent HSV-1, we demonstrate that ganglionic viral load reduction leads to a significant decrease of viral shedding in treated female mice. While therapy is well tolerated, in some instances, we observe hepatotoxicity at high doses and subtle histological evidence of neuronal injury without observable neurological signs or deficits. Simplification of the regimen through use of a single serotype (AAV9) delivering single meganuclease targeting a duplicated region of the HSV genome, dose reduction, and use of a neuron-specific promoter each results in improved tolerability while retaining efficacy. These results reinforce the curative potential of gene editing for HSV disease.


Sujet(s)
Dependovirus , Édition de gène , Herpès , Herpèsvirus humain de type 1 , Charge virale , Excrétion virale , Animaux , Édition de gène/méthodes , Femelle , Dependovirus/génétique , Souris , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/physiologie , Herpès/génétique , Herpès/virologie , Herpès/thérapie , Modèles animaux de maladie humaine , Latence virale/génétique , Humains , Vecteurs génétiques/génétique , Cellules Vero , Thérapie génétique/méthodes , Herpès génital/thérapie , Herpès génital/virologie , ADN viral/génétique
17.
Viruses ; 16(5)2024 05 08.
Article de Anglais | MEDLINE | ID: mdl-38793628

RÉSUMÉ

Our current understanding of HSV latency is based on a variety of clinical observations, and in vivo, ex vivo, and in vitro model systems, each with unique advantages and drawbacks. The criteria for authentically modeling HSV latency include the ability to easily manipulate host genetics and biological pathways, as well as mimicking the immune response and viral pathogenesis in human infections. Although realistically modeling HSV latency is necessary when choosing a model, the cost, time requirement, ethical constraints, and reagent availability are also equally important. Presently, there remains a pressing need for in vivo models that more closely recapitulate human HSV infection. While the current in vivo, ex vivo, and in vitro models used to study HSV latency have limitations, they provide further insights that add to our understanding of latency. In vivo models have shed light on natural infection routes and the interplay between the host immune response and the virus during latency, while in vitro models have been invaluable in elucidating molecular pathways involved in latency. Below, we review the relative advantages and disadvantages of current HSV models and highlight insights gained through each.


Sujet(s)
Herpès , Latence virale , Humains , Herpès/virologie , Animaux , Simplexvirus/physiologie , Herpèsvirus humain de type 1/physiologie , Herpèsvirus humain de type 1/génétique , Modèles animaux de maladie humaine
18.
Compend Contin Educ Dent ; 45(4): 192-197; quiz 198, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38622078

RÉSUMÉ

Human herpes virus is a family of DNA viruses that includes herpes simplex virus (HSV) and varicella zoster virus (VZV). HSV-1 and HSV-2 are fairly common and result in oral and genital lesions. Recurrent infections of herpes include lesions on the lips resulting in pain and possibly societal stigma, making adequate treatment of these conditions crucial. VZV is the cause of chicken pox and shingles. Acyclovir and other nucleoside analogues have been the gold standard of treatment for HSV and VZV, but newer, more effective treatments are being developed, which is beneficial regarding the issue of resistance to standard antivirals. Human papillomavirus (HPV) is also a DNA virus with different subtypes that result in four common oral benign lesions. The significance and treatments of HSV, VZV, and HPV are discussed, along with certain developing treatments of herpes labialis (HSV).


Sujet(s)
Zona , Herpèsvirus humain de type 1 , Infections à papillomavirus , Humains , Herpèsvirus humain de type 3/génétique , Virus des Papillomavirus humains , Infections à papillomavirus/thérapie , Herpèsvirus humain de type 1/génétique
20.
J Virol ; 98(5): e0003224, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38651900

RÉSUMÉ

Critical stages of lytic herpes simplex virus type 1 (HSV-1) replication are marked by the sequential expression of immediate early (IE) to early (E), then late (L) viral genes. HSV-1 can also persist in neuronal cells via a non-replicative, transcriptionally repressed infection called latency. The regulation of lytic and latent transcriptional profiles is critical to HSV-1 pathogenesis and persistence. We sought a fluorescence-based approach to observe the outcome of neuronal HSV-1 infection at the single-cell level. To achieve this goal, we constructed and characterized a novel HSV-1 recombinant that enables discrimination between lytic and latent infection. The dual reporter HSV-1 encodes a human cytomegalovirus-immediate early (hCMV-IE) promoter-driven enhanced yellow fluorescent protein (eYFP) to visualize the establishment of infection and an endogenous mCherry-VP26 fusion to report lytic replication. We confirmed that viral gene expression, replication, and spread of infection are not altered by the incorporation of the fluorescent reporters, and fluorescent protein (FP) detection virtuously reports the progression of lytic replication. We demonstrate that the outcome of HSV-1 infection of compartmentalized primary neurons is determined by viral inoculating dose: high-dose axonal inoculation proceeds to lytic replication, whereas low-dose axonal inoculation establishes a latent HSV-1 infection. Interfering with low-dose axonal inoculation via small molecule drugs reports divergent phenotypes of eYFP and mCherry reporter detection, correlating with altered states of viral gene expression. We report that the transcriptional state of neuronal HSV-1 infection is variable in response to changes in the intracellular neuronal environment.IMPORTANCEHerpes simplex virus type 1 (HSV-1) is a prevalent human pathogen that infects approximately 67% of the global human population. HSV-1 invades the peripheral nervous system, where latent HSV-1 infection persists within the host for life. Immunological evasion, viral persistence, and herpetic pathologies are determined by the regulation of HSV-1 gene expression. Studying HSV-1 gene expression during neuronal infection is challenging but essential for the development of antiviral therapeutics and interventions. We used a recombinant HSV-1 to evaluate viral gene expression during infection of primary neurons. Manipulation of cell signaling pathways impacts the establishment and transcriptional state of HSV-1 latency in neurons. The work here provides critical insight into the cellular and viral factors contributing to the establishment of latent HSV-1 infection.


Sujet(s)
Herpès , Herpèsvirus humain de type 1 , Neurones , Animaux , Humains , Chlorocebus aethiops , Cytomegalovirus/génétique , Cytomegalovirus/physiologie , Régulation de l'expression des gènes viraux , Gènes rapporteurs , Herpès/virologie , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/physiologie , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Neurones/virologie , Neurones/métabolisme , Cellules Vero , Latence virale/génétique , Réplication virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...