Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.561
Filtrar
1.
Rev Med Virol ; 34(5): e2584, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39304923

RESUMO

Neurotropic viruses have been implicated in altering the central nervous system microenvironment and promoting brain metastasis of breast cancer through complex interactions involving viral entry mechanisms, modulation of the blood-brain barrier, immune evasion, and alteration of the tumour microenvironment. This narrative review explores the molecular mechanisms by which neurotropic viruses such as Herpes Simplex Virus, Human Immunodeficiency Virus, Japanese Encephalitis Virus, and Rabies Virus facilitate brain metastasis, focusing on their ability to disrupt blood-brain barrier integrity, modulate immune responses, and create a permissive environment for metastatic cell survival and growth within the central nervous system. Current therapeutic implications and challenges in targeting neurotropic viruses to prevent or treat brain metastasis are discussed, highlighting the need for innovative strategies and multidisciplinary approaches in virology, oncology, and immunology.


Assuntos
Neoplasias Encefálicas , Neoplasias da Mama , Humanos , Neoplasias da Mama/patologia , Neoplasias da Mama/virologia , Neoplasias da Mama/terapia , Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/virologia , Neoplasias Encefálicas/terapia , Feminino , Barreira Hematoencefálica/virologia , Animais , Microambiente Tumoral , Vírus da Raiva/fisiologia , Vírus da Raiva/patogenicidade , Vírus da Raiva/imunologia , Simplexvirus/fisiologia
2.
Rev Med Virol ; 34(5): e2574, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39090526

RESUMO

Herpes simplex virus (HSV) infections in allogeneic haematopoietic stem cell transplantation (HSCT) recipients pose significant challenges, with higher incidence, severity, and risk of emergence of resistance to antivirals due to impaired T-cell mediated immunity. This literature review focuses on acyclovir-refractory/resistant HSV infections in HSCT recipients. The review addresses the efficacy of antiviral prophylaxis, the incidence of acyclovir-refractory/resistant HSV infections, and the identification of risk factors and potential prognostic impact associated with those infections. Additionally, alternative therapeutic options are discussed. While acyclovir prophylaxis demonstrates a significant benefit in reducing HSV infections in HSCT recipients and, in some cases, overall mortality, concerns arise about the emergence of drug-resistant HSV strains. Our systematic review reports a median incidence of acyclovir-resistant HSV infections of 16.1%, with an increasing trend in recent years. Despite limitations in available studies, potential risk factors of emergence of HSV resistance to acyclovir include human leucocyte antigen (HLA) mismatches, myeloid neoplasms and acute leukaemias, and graft-versus-host disease (GVHD). Limited evidences suggest a potentially poorer prognosis for allogeneic HSCT recipients with acyclovir-refractory/resistant HSV infection. Alternative therapeutic approaches, such as foscarnet, cidofovir, topical cidofovir, optimised acyclovir dosing, and helicase-primase inhibitors offer promising options but require further investigations. Overall, larger studies are needed to refine preventive and therapeutic strategies for acyclovir-refractory/resistant HSV infections in allogeneic HSCT recipients and to identify those at higher risk.


Assuntos
Aciclovir , Antivirais , Farmacorresistência Viral , Transplante de Células-Tronco Hematopoéticas , Herpes Simples , Humanos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Herpes Simples/tratamento farmacológico , Herpes Simples/virologia , Herpes Simples/terapia , Antivirais/uso terapêutico , Aciclovir/uso terapêutico , Simplexvirus/efeitos dos fármacos , Simplexvirus/fisiologia , Fatores de Risco , Transplantados , Incidência
3.
Mol Biomed ; 5(1): 35, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39207577

RESUMO

Herpes simplex virus (HSV), an epidemic human pathogen threatening global public health, gains notoriety for its complex pathogenesis that encompasses lytic infection of mucosal cells, latent infection within neurons, and periodic reactivation. This intricate interplay, coupled with HSV's sophisticated immune evasion strategies, gives rise to various diseases, including genital lesions, neonatal encephalitis, and cancer. Despite more than 70 years of relentless research, an effective preventive or therapeutic vaccine against HSV has yet to emerge, primarily due to the limited understanding of virus-host interactions, which in turn impedes the identification of effective vaccine targets. However, HSV's unique pathological features, including its substantial genetic load capacity, high replicability, transmissibility, and neurotropism, render it a promising candidate for various applications, spanning oncolytic virotherapy, gene and immune therapies, and even as an imaging tracer in neuroscience. In this review, we comprehensively update recent breakthroughs in HSV pathogenesis and immune evasion, critically summarize the progress made in vaccine candidate development, and discuss the multifaceted applications of HSV as a biological tool. Importantly, we highlight both success and challenges, emphasizing the critical need for intensified research into HSV, with the aim of providing deeper insights that can not only advance HSV treatment strategies but also broaden its application horizons.


Assuntos
Herpes Simples , Desenvolvimento de Vacinas , Humanos , Herpes Simples/imunologia , Herpes Simples/prevenção & controle , Herpes Simples/virologia , Animais , Simplexvirus/patogenicidade , Simplexvirus/imunologia , Simplexvirus/fisiologia , Vacinas contra o Vírus do Herpes Simples/imunologia , Evasão da Resposta Imune
4.
Viruses ; 16(8)2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39205282

RESUMO

The cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-dependent Cl- channel, is closely associated with multiple pathogen infections, such as SARS-CoV-2. However, whether the function of the CFTR is involved in herpes simplex virus (HSV) infection has not been reported. To evaluate the association of CFTR activity with HSV infection, the antiviral effect of CFTR inhibitors in epithelial cells and HSV-infected mice was tested in this study. The data showed that treatment with CFTR inhibitors in different concentrations, Glyh-101 (5-20 µM), CFTRi-172 (5-20 µM) and IOWH-032 (5-20 µM), or the gene silence of the CFTR could suppress herpes simplex virus 1 (HSV-1) and herpes simplex virus 2 (HSV-2) replication in human HaCaT keratinocytes cells, and that a CFTR inhibitor, Glyh-101 (10-20 µM), protected mice from HSV-1 and HSV-2 infection. Intracellular Cl- concentration ([Cl-]i) was decreased after HSV infection via the activation of adenylyl cyclase (AC)-cAMP signaling pathways. CFTR inhibitors (20 µM) increased the reduced [Cl-]i caused by HSV infection in host epithelial cells. Additionally, CFTR inhibitors reduced the activity and phosphorylation of SGK1 in infected cells and tissues (from the eye and vagina). Our study found that CFTR inhibitors can effectively suppress HSV-1 and HSV-2 infection, revealing a previously unknown role of CFTR inhibitors in HSV infection and suggesting new perspectives on the mechanisms governing HSV infection in host epithelial cells, as well as leading to potential novel treatments.


Assuntos
Antivirais , Regulador de Condutância Transmembrana em Fibrose Cística , Herpes Simples , Herpesvirus Humano 1 , Herpesvirus Humano 2 , Replicação Viral , Animais , Camundongos , Antivirais/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Humanos , Herpes Simples/tratamento farmacológico , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Replicação Viral/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Herpesvirus Humano 2/fisiologia , Feminino , Linhagem Celular , Células Epiteliais/virologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células HaCaT , Queratinócitos/virologia , Queratinócitos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Chlorocebus aethiops , Simplexvirus/efeitos dos fármacos , Simplexvirus/fisiologia
5.
Viruses ; 16(5)2024 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-38793628

RESUMO

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.


Assuntos
Herpes Simples , Latência Viral , Humanos , Herpes Simples/virologia , Animais , Simplexvirus/fisiologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/genética , Modelos Animais de Doenças
6.
Int J Mol Sci ; 25(9)2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38732185

RESUMO

Herpes simplex virus (HSV) infections are highly widespread among humans, producing symptoms ranging from ulcerative lesions to severe diseases such as blindness and life-threatening encephalitis. At present, there are no vaccines available, and some existing antiviral treatments can be ineffective or lead to adverse effects. As a result, there is a need for new anti-HSV drugs. In this report, the in vitro anti-HSV effect of 9,9'-norharmane dimer (nHo-dimer), which belongs to the ß-carboline (ßC) alkaloid family, was evaluated. The dimer exhibited no virucidal properties and did not impede either the attachment or penetration steps of viral particles. The antiviral effect was only exerted under the constant presence of the dimer in the incubation media, and the mechanism of action was found to involve later events of virus infection. Analysis of fluorescence lifetime imaging data showed that the nHo-dimer internalized well into the cells when present in the extracellular incubation medium, with a preferential accumulation into perinuclear organelles including mitochondria. After washing the host cells with fresh medium free of nHo-dimer, the signal decreased, suggesting the partial release of the compound from the cells. This agrees with the observation that the antiviral effect is solely manifested when the alkaloid is consistently present in the incubation media.


Assuntos
Antivirais , Antivirais/farmacologia , Antivirais/química , Chlorocebus aethiops , Humanos , Células Vero , Animais , Simplexvirus/efeitos dos fármacos , Simplexvirus/fisiologia , Herpes Simples/tratamento farmacológico , Herpes Simples/virologia , Carbolinas/farmacologia , Carbolinas/química , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Harmina/farmacologia , Harmina/química , Harmina/análogos & derivados
7.
A A Pract ; 17(10): e01719, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37870295

RESUMO

A 36-year-old woman with no significant past medical history underwent a sphenopalatine ganglion block for treatment of a month-long migraine headache refractory to conservative treatment protocols. The headache resolved initially, but 1 day following the procedure, the headache recurred. The patient also developed an erythematous and edematous rash which cultures confirmed to be herpes simplex virus (HSV). Following several unsuccessful treatment modalities, the patient received valacyclovir, which resulted in resolution of her headache. Underlying HSV-1 infection may cause intractable migraine headache and nerve blocks may potentiate reactivation of latent HSV infection that caused the skin lesion in this case.


Assuntos
Herpes Simples , Transtornos de Enxaqueca , Bloqueio do Gânglio Esfenopalatino , Feminino , Humanos , Adulto , Simplexvirus/fisiologia , Herpes Simples/tratamento farmacológico , Herpes Simples/etiologia , Transtornos de Enxaqueca/terapia , Cefaleia
8.
Viruses ; 15(1)2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36680194

RESUMO

Herpes simplex virus (HSV) and varicella zoster virus (VZV) rely on transport of virus particles in neuronal axons to spread from sites of viral latency in sensory ganglia to peripheral tissues then on to other hosts. This process of anterograde axonal transport involves kinesin motors that move virus particles rapidly along microtubules. α-herpesvirus anterograde transport has been extensively studied by characterizing the porcine pseudorabies virus (PRV) and HSV, with most studies focused on two membrane proteins: gE/gI and US9. It was reported that PRV and HSV US9 proteins bind to kinesin motors, promoting tethering of virus particles on the motors, and furthering anterograde transport within axons. Alternatively, other models have argued that HSV and PRV US9 and gE/gI function in the cytoplasm and not in neuronal axons. Specifically, HSV gE/gI and US9 mutants are defective in the assembly of virus particles in the cytoplasm of neurons and the subsequent sorting of virus particles to cell surfaces and into axons. However, PRV US9 and gE/gI mutants have not been characterized for these cytoplasmic defects. We examined neurons infected with PRV mutants, one lacking both gE/gI and US9 and the other lacking just US9, by electron microscopy. Both PRV mutants exhibited similar defects in virus assembly and cytoplasmic sorting of virus particles to cell surfaces. As well, the mutants exhibited reduced quantities of infectious virus in neurons and in cell culture supernatants. We concluded that PRV US9 primarily functions in neurons to promote cytoplasmic steps in anterograde transport.


Assuntos
Herpesvirus Suídeo 1 , Animais , Suínos , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/metabolismo , Transporte Axonal/fisiologia , Proteínas do Envelope Viral/metabolismo , Cinesinas/metabolismo , Linhagem Celular , Axônios , Simplexvirus/fisiologia , Citoplasma/metabolismo , Vírion/metabolismo
11.
Viruses ; 14(2)2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35215799

RESUMO

Critically ill patients, such as those in intensive care units (ICUs), can develop herpes simplex virus (HSV) pneumonitis. Given the high prevalence of acute respiratory distress syndrome (ARDS) and multiple pre-existing conditions among ICU patients with HSV pneumonitis, factors predicting mortality in this patient population require further investigation. In this retrospective study, the bronchoalveolar lavage or sputum samples of ICU patients were cultured or subjected to a polymerase chain reaction for HSV detection. Univariable and multivariable Cox regressions were conducted for mortality outcomes. The length of hospital stay was plotted against mortality on Kaplan-Meier curves. Among the 119 patients with HSV pneumonitis (age: 65.8 ± 14.9 years), the mortality rate was 61.34% (73 deaths). The mortality rate was significantly lower among patients with diabetes mellitus (odds ratio [OR] 0.12, 95% confidence interval [CI]: 0.02-0.49, p = 0.0009) and significantly higher among patients with ARDS (OR: 4.18, 95% CI: 1.05-17.97, p < 0.0001) or high (≥30) Acute Physiology and Chronic Health Evaluation II scores (OR: 1.08, 95% CI: 1.00-1.18, p = 0.02). Not having diabetes mellitus (DM), developing ARDS, and having a high Acute Physiology and Chronic Health Evaluation II (APACHE II) score were independent predictors of mortality among ICU patients with HSV pneumonitis.


Assuntos
Estado Terminal/mortalidade , Herpes Simples/mortalidade , Pneumonia Viral/mortalidade , Síndrome do Desconforto Respiratório/complicações , Simplexvirus/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Herpes Simples/etiologia , Herpes Simples/virologia , Mortalidade Hospitalar , Humanos , Unidades de Terapia Intensiva/estatística & dados numéricos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Pneumonia Viral/etiologia , Pneumonia Viral/virologia , Estudos Retrospectivos , Simplexvirus/genética
12.
Viruses ; 14(2)2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35215908

RESUMO

The transcription factors IRF3 and NF-κB are crucial in innate immune signalling in response to many viral and bacterial pathogens. However, mechanisms leading to their activation remain incompletely understood. Viral RNA can be detected by RLR receptors, such as RIG-I and MDA5, and the dsRNA receptor TLR3. Alternatively, the DExD-Box RNA helicases DDX1-DDX21-DHX36 activate IRF3/NF-κB in a TRIF-dependent manner independent of RIG-I, MDA5, or TLR3. Here, we describe DDX50, which shares 55.6% amino acid identity with DDX21, as a non-redundant factor that promotes activation of the IRF3 signalling pathway following its stimulation with viral RNA or infection with RNA and DNA viruses. Deletion of DDX50 in mouse and human cells impaired IRF3 phosphorylation and IRF3-dependent endogenous gene expression and cytokine/chemokine production in response to cytoplasmic dsRNA (polyIC transfection), and infection by RNA and DNA viruses. Mechanistically, whilst DDX50 co-immunoprecipitated TRIF, it acted independently to the previously described TRIF-dependent RNA sensor DDX1. Indeed, shRNA-mediated depletion of DDX1 showed DDX1 was dispensable for signalling in response to RNA virus infection. Importantly, loss of DDX50 resulted in a significant increase in replication and dissemination of virus following infection with vaccinia virus, herpes simplex virus, or Zika virus, highlighting its important role as a broad-ranging viral restriction factor.


Assuntos
RNA Helicases DEAD-box/metabolismo , Herpes Simples/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Simplexvirus/fisiologia , Vaccinia virus/fisiologia , Vacínia/metabolismo , Infecção por Zika virus/metabolismo , Zika virus/fisiologia , Animais , RNA Helicases DEAD-box/genética , Herpes Simples/genética , Herpes Simples/virologia , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon/genética , Camundongos , Fosforilação , Transdução de Sinais , Simplexvirus/genética , Vacínia/genética , Vacínia/virologia , Vaccinia virus/genética , Zika virus/genética , Infecção por Zika virus/genética , Infecção por Zika virus/virologia
13.
Viruses ; 14(1)2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-35062295

RESUMO

Primate simplex viruses, including Herpes simplex viruses 1 and 2, form a group of closely related herpesviruses, which establish latent infections in neurons of their respective host species. While neuropathogenic infections in their natural hosts are rare, zoonotic transmission of Macacine alphaherpesvirus 1 (McHV1) from macaques to humans is associated with severe disease. Human infections with baboon-derived Papiine alphaherpesvirus 2 (PaHV2) have not been reported, although PaHV2 and McHV1 share several biological properties, including neuropathogenicity in mice. The reasons for potential differences in PaHV2 and McHV1 pathogenicity are presently not understood, and answering these questions will require mutagenic analysis. Here, we report the development of a recombinant system, which allows rescue of recombinant PaHV2. In addition, we used recombineering to generate viruses carrying reporter genes (Gaussia luciferase or enhanced green fluorescent protein), which replicate with similar efficiency as wild-type PaHV2. We demonstrate that these viruses can be used to analyze susceptibility of cells to infection and inhibition of infection by neutralizing antibodies and antiviral compounds. In summary, we created a recombinant system for PaHV2, which in the future will be invaluable for molecular analyses of neuropathogenicity of PaHV2.


Assuntos
Clonagem Molecular , Genoma Viral , Recombinação Genética , Simplexvirus/genética , Animais , Anticorpos Antivirais/imunologia , Antivirais/farmacologia , Linhagem Celular , Genes Reporter , Humanos , Papio/imunologia , Simplexvirus/imunologia , Simplexvirus/patogenicidade , Simplexvirus/fisiologia
14.
Viruses ; 13(12)2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34960625

RESUMO

Herpesvirus capsids are assembled in the nucleus and undergo a two-step process to cross the nuclear envelope. Capsids bud into the inner nuclear membrane (INM) aided by the nuclear egress complex (NEC) proteins UL31/34. At that stage of egress, enveloped virions are found for a short time in the perinuclear space. In the second step of nuclear egress, perinuclear enveloped virions (PEVs) fuse with the outer nuclear membrane (ONM) delivering capsids into the cytoplasm. Once in the cytoplasm, capsids undergo re-envelopment in the Golgi/trans-Golgi apparatus producing mature virions. This second step of nuclear egress is known as de-envelopment and is the focus of this review. Compared with herpesvirus envelopment at the INM, much less is known about de-envelopment. We propose a model in which de-envelopment involves two phases: (i) fusion of the PEV membrane with the ONM and (ii) expansion of the fusion pore leading to release of the viral capsid into the cytoplasm. The first phase of de-envelopment, membrane fusion, involves four herpes simplex virus (HSV) proteins: gB, gH/gL, gK and UL20. gB is the viral fusion protein and appears to act to perturb membranes and promote fusion. gH/gL may also have similar properties and appears to be able to act in de-envelopment without gB. gK and UL20 negatively regulate these fusion proteins. In the second phase of de-envelopment (pore expansion and capsid release), an alpha-herpesvirus protein kinase, US3, acts to phosphorylate NEC proteins, which normally produce membrane curvature during envelopment. Phosphorylation of NEC proteins reverses tight membrane curvature, causing expansion of the membrane fusion pore and promoting release of capsids into the cytoplasm.


Assuntos
Capsídeo/metabolismo , Infecções por Herpesviridae/virologia , Herpesviridae/fisiologia , Proteínas Virais de Fusão/metabolismo , Núcleo Celular/virologia , Citoplasma/virologia , Herpesviridae/genética , Herpesviridae/ultraestrutura , Humanos , Fusão de Membrana , Membrana Nuclear/virologia , Fosforilação , Simplexvirus/genética , Simplexvirus/fisiologia , Envelope Viral , Proteínas Virais de Fusão/genética , Vírion , Rede trans-Golgi/virologia
15.
Viruses ; 13(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34834914

RESUMO

Viral skin infections often affect the sports community. The aim of this study was to assess the rates, location sites, and seasons of appearance of common viral cutaneous diseases in beach volleyball athletes in Greece. Five hundred and forty-nine beach volleyball athletes participated in this study. The average age was 28.4 years. The viral infections were herpes simplex (type 1), molluscum contagiosum and warts. The measured parameters included: gender, age, the season when athletes may be more susceptible to infections and the location of infection in the body. Practicing information such as the number of training years, number of weekly trainings, and average hours of daily training was also recorded. Incidence rates correlated in relation to age: (a) warts (p < 0.001), molluscum contagiosum (p < 0.001), and herpes simplex (p = 0.001); (b) years of training: warts (p < 0.001), molluscum contagiosum (p < 0.001), and herpes simplex (p = 0.004); (c) average hours of daily training: molluscum contagiosum (p = 0.006) and herpes simplex (p < 0.010). The skin is the largest organ, and the risk of infection should not be underestimated. Prevention, early detection, recognition, and treatment are related to health and athletic performance, but also to the risk of transmission.


Assuntos
Atletas/estatística & dados numéricos , Herpes Simples/epidemiologia , Molusco Contagioso/epidemiologia , Vírus do Molusco Contagioso/isolamento & purificação , Dermatopatias/epidemiologia , Verrugas/epidemiologia , Adulto , Feminino , Grécia/epidemiologia , Herpes Simples/virologia , Humanos , Masculino , Molusco Contagioso/virologia , Vírus do Molusco Contagioso/classificação , Vírus do Molusco Contagioso/genética , Vírus do Molusco Contagioso/fisiologia , Filogenia , Simplexvirus/genética , Simplexvirus/isolamento & purificação , Simplexvirus/fisiologia , Dermatopatias/virologia , Voleibol , Verrugas/virologia , Adulto Jovem
16.
Sci Rep ; 11(1): 21171, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34707143

RESUMO

Herpes simplex virus type 1 (HSV-1) is the predominant cause of herpes simplex encephalitis (HSE), a condition characterized by acute inflammation and viral replication in the brain. Host genetics contribute to HSE onset, including monogenic defects in type I interferon signaling in cases of childhood HSE. Mouse models suggest a further contribution of immune cell-mediated inflammation to HSE pathogenesis. We have previously described a truncating mutation in the c-Rel transcription factor (RelC307X) that drives lethal HSE in 60% of HSV-1-infected RelC307X mice. In this study, we combined dual host-virus RNA sequencing with flow cytometry to explore cell populations and mechanisms involved in RelC307X-driven HSE. At day 5 postinfection, prior to HSE clinical symptom onset, elevated HSV-1 transcription was detected together with augmented host interferon-stimulated and inflammatory gene expression in the brainstems of high-responding RelC307X mice, predictive of HSE development. This early induction of host gene expression preceded pathological infiltration of myeloid and T cells in RelC307X mice at HSE onset by day 7. Thus, we establish c-Rel as an early regulator of viral and host responses during mouse HSE. These data further highlight the importance of achieving a balanced immune response and avoiding excess interferon-driven inflammation to promote HSE resistance.


Assuntos
Encefalite por Herpes Simples/metabolismo , Interferon Tipo I/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Animais , Encefalite por Herpes Simples/virologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas Proto-Oncogênicas c-rel/genética , Transdução de Sinais , Simplexvirus/genética , Simplexvirus/patogenicidade , Simplexvirus/fisiologia , Linfócitos T/metabolismo , Linfócitos T/virologia
17.
J Immunol ; 207(7): 1824-1835, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34470854

RESUMO

This report evaluates how HSV enters the brain to cause herpes simplex encephalitis following infection at a peripheral site. We demonstrate that encephalitis regularly occurred when BALB/c mice were infected with HSV and treated daily with 2-deoxy-d-glucose (2DG), which inhibits glucose use via the glycolysis pathway. The outcome of infection in the trigeminal ganglion (TG), the site to which the virus spreads, replicates, and establishes latency, showed marked differences in viral and cellular events between treated and untreated animals. In control-untreated mice, the replicating virus was present only during early time points, whereas in 2DG recipients, replicating virus remained for the 9-d observation period. This outcome correlated with significantly reduced numbers of innate inflammatory cells as well as T cells in 2DG-treated animals. Moreover, T cells in the TG of treated animals were less activated and contained a smaller fraction of expressed IFN-γ production compared with untreated controls. The breakdown of latency was accelerated when cultures of TG cells taken from mice with established HSV latency were cultured in the presence of 2DG. Taken together, the results of both in vivo and in vitro investigations demonstrate that the overall effects of 2DG therapy impaired the protective effects of one or more inflammatory cell types in the TG that normally function to control productive infection and prevent spread of virus to the brain.


Assuntos
Encéfalo/patologia , Encefalite por Herpes Simples/metabolismo , Glucose/metabolismo , Simplexvirus/fisiologia , Linfócitos T/imunologia , Animais , Células Cultivadas , Desoxiglucose/administração & dosagem , Humanos , Imunidade Inata , Interferon gama/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Latência Viral
18.
Epidemiol Infect ; 149: e145, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34130765

RESUMO

The appearance on the skin of herpes virus lesions, concomitantly with the coronavirus disease 2019 (COVID-19) pandemic, leads us to suspect an underlying infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Diagnostic reverse transcriptase polymerase chain reaction tests and immunoglobulin M (IgM) and IgG seroconversion studies have therefore been carried out. We present three cases of herpes virus infections in immunocompetent patients: one of the infections was herpes simplex 1 in a 40-year-old woman, and the other two were herpes varicella-zoster infections in a 62-year-old man and a 25-year-old woman. The patients were in the care of the southern health district of Seville of the SAS (Andalusian Health Service) during the Spanish state of alarm over the COVID-19 pandemic. The SARS-CoV-2 infection was confirmed in only one of the three cases. In this study, we briefly review the etiopathogenic role of the COVID-19 pandemic situation, whereby immunodeficiencies are generated that favour the appearance of other viral infections, such as herpes virus infections.


Assuntos
COVID-19/complicações , Herpes Simples/etiologia , Herpes Zoster/etiologia , Herpesvirus Humano 3/fisiologia , Simplexvirus/fisiologia , Ativação Viral , Adulto , COVID-19/epidemiologia , COVID-19/virologia , Feminino , Herpes Simples/diagnóstico , Herpes Simples/virologia , Herpes Zoster/diagnóstico , Herpes Zoster/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Espanha/epidemiologia
19.
Int J Cancer ; 149(1): 214-227, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33687756

RESUMO

Oncolytic viruses (OVs) remodel the tumor microenvironment by switching a "cold" tumor into a "hot" tumor with high CD8+ T-cell infiltration. CD8+ T-cell activity plays an essential role in the antitumor efficacy of OVs. However, the activity of T cells is impaired by the programmed cell death protein-1/programmed cell death-ligand 1 (PD-1/PD-L1) interaction. To date, it remains unclear why OVs alone have a significant antitumor activity even when PD-L1 expression persists on tumor or immune cells. In this study, we found that canerpaturev (C-REV) treatment significantly suppressed tumor growth, even though it induced a significant increase in PD-L1 expression in tumors in vivo as well as persistence of high PD-L1 expression on antigen-presenting cells (macrophage and dendritic cells [DCs]). Surprisingly, we observed that C-REV treatment increased the abundance of activated CD8+ PD-1- tumor-infiltrating lymphocytes (TILs) in the tumor on both the injected and contralateral sides, although infiltration of CD8+ PD-1high TILs into the tumor was observed in the control group. Moreover, the difference in PD-1 expression was observed only in tumors after treatment with C-REV, whereas most CD8+ T cells in the spleen, tumor-draining lymph nodes and blood were PD-1-negative, and this did not change after C-REV treatment. In addition, changes in expression of T-cell immunoglobulin and mucin-domain containing-3 and T-cell immune-receptor with Ig and ITIM domains were not observed on CD8+ TILs after C-REV treatment. Taken together, our findings may reveal mechanisms that allow OVs to trigger an antitumor immune response, irrespective of a PD-L1-enriched tumor microenvironment, by recruitment of CD8+ PD-1- TILs.


Assuntos
Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/imunologia , Carcinoma de Células Escamosas/imunologia , Herpes Simples/imunologia , Neoplasias Pancreáticas/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Microambiente Tumoral/imunologia , Animais , Antígeno B7-H1/imunologia , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Feminino , Herpes Simples/virologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptor de Morte Celular Programada 1/imunologia , Simplexvirus/fisiologia
20.
Methods Mol Biol ; 2193: 183-196, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32808270

RESUMO

Corneal infections by viruses and bacteria can result in ocular surface defects, ulcers, or wounds. Herpes simplex virus type-1 (HSV-1) is a human virus with global seroprevalence in the range of 60-90%. While the virus more commonly causes mucocutaneous lesions including ulcers on the face and mouth, it is also a leading cause of infection-associated blindness. In this chapter, we discuss an in-depth protocol required to evaluate corneal damage due to HSV-1 infection using porcine models of ex vivo infection. Our methods can be adapted to study similar infections caused by other viruses and bacteria.


Assuntos
Lesões da Córnea/virologia , Modelos Animais de Doenças , Ceratite Herpética/virologia , Simplexvirus/fisiologia , Técnicas de Cultura de Tecidos/métodos , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Córnea/citologia , Córnea/virologia , Lesões da Córnea/etiologia , Lesões da Córnea/patologia , Interações Hospedeiro-Patógeno , Ceratite Herpética/patologia , Simplexvirus/efeitos dos fármacos , Simplexvirus/patogenicidade , Suínos , Células Vero , Internalização do Vírus , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA