Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 870
Filtrar
1.
Nat Commun ; 15(1): 3669, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38693119

RESUMEN

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.


Asunto(s)
Fusobacterium nucleatum , Herpesvirus Humano 1 , Viroterapia Oncolítica , Virus Oncolíticos , Proteínas de Unión al ARN , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/genética , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Animales , Humanos , Viroterapia Oncolítica/métodos , Ratones , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/inmunología , Línea Celular Tumoral , Fusobacterium nucleatum/inmunología , Neoplasias/terapia , Neoplasias/inmunología , Femenino , Inmunidad Innata , Ratones Endogámicos BALB C
2.
Nature ; 623(7985): 157-166, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37853118

RESUMEN

Immunotherapy failures can result from the highly suppressive tumour microenvironment that characterizes aggressive forms of cancer such as recurrent glioblastoma (rGBM)1,2. Here we report the results of a first-in-human phase I trial in 41 patients with rGBM who were injected with CAN-3110-an oncolytic herpes virus (oHSV)3. In contrast to other clinical oHSVs, CAN-3110 retains the viral neurovirulence ICP34.5 gene transcribed by a nestin promoter; nestin is overexpressed in GBM and other invasive tumours, but not in the adult brain or healthy differentiated tissue4. These modifications confer CAN-3110 with preferential tumour replication. No dose-limiting toxicities were encountered. Positive HSV1 serology was significantly associated with both improved survival and clearance of CAN-3110 from injected tumours. Survival after treatment, particularly in individuals seropositive for HSV1, was significantly associated with (1) changes in tumour/PBMC T cell counts and clonal diversity, (2) peripheral expansion/contraction of specific T cell clonotypes; and (3) tumour transcriptomic signatures of immune activation. These results provide human validation that intralesional oHSV treatment enhances anticancer immune responses even in immunosuppressive tumour microenvironments, particularly in individuals with cognate serology to the injected virus. This provides a biological rationale for use of this oncolytic modality in cancers that are otherwise unresponsive to immunotherapy (ClinicalTrials.gov: NCT03152318 ).


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Herpesvirus Humano 1 , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Glioblastoma/inmunología , Glioblastoma/patología , Nestina/genética , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Virus Oncolíticos/fisiología , Reproducibilidad de los Resultados , Análisis de Supervivencia , Linfocitos T/citología , Linfocitos T/inmunología , Resultado del Tratamiento , Microambiente Tumoral/inmunología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología
3.
Microbiol Spectr ; 10(1): e0188321, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35196784

RESUMEN

Virus infection triggers intricate signal cascade reactions to activate the host innate immunity, which leads to the production of type I interferon (IFN-I). Herpes simplex virus 1 (HSV-1), a human-restricted pathogen, is capable of encoding over 80 viral proteins, and several of them are involved in immune evasion to resist the host antiviral response through the IFN-I signaling pathway. Here, we determined that HSV-1 UL31, which is associated with nuclear matrix and is essential for the formation of viral nuclear egress complex, could inhibit retinoic acid-inducible gene I (RIG-I)-like receptor pathway-mediated interferon beta (IFN-ß)-luciferase (Luc) and (PRDIII-I)4-Luc (an expression plasmid of IFN-ß positive regulatory elements III and I) promoter activation, as well as the mRNA transcription of IFN-ß and downstream interferon-stimulated genes (ISGs), such as ISG15, ISG54, ISG56, etc., to promote viral infection. UL31 was shown to restrain IFN-ß activation at the interferon regulatory factor 3 (IRF3)/IRF7 level. Mechanically, UL31 was demonstrated to interact with TANK binding kinase 1 (TBK1), inducible IκB kinase (IKKi), and IRF3 to impede the formation of the IKKi-IRF3 complex but not the formation of the IRF7-related complex. UL31 could constrain the dimerization and nuclear translocation of IRF3. Although UL31 was associated with the CREB binding protein (CBP)/p300 coactivators, it could not efficiently hamper the formation of the CBP/p300-IRF3 complex. In addition, UL31 could facilitate the degradation of IKKi and IRF3 by mediating their K48-linked polyubiquitination. Taken together, these results illustrated that UL31 was able to suppress IFN-ß activity by inhibiting the activation of IKKi and IRF3, which may contribute to the knowledge of a new immune evasion mechanism during HSV-1 infection. IMPORTANCE The innate immune system is the first line of host defense against the invasion of pathogens. Among its mechanisms, IFN-I is an essential cytokine in the antiviral response, which can help the host eliminate a virus. HSV-1 is a double-stranded DNA virus that can cause herpes and establish a lifelong latent infection, due to its possession of multiple mechanisms to escape host innate immunity. In this study, we illustrate for the first time that the HSV-1-encoded UL31 protein has a negative regulatory effect on IFN-ß production by blocking the dimerization and nuclear translocation of IRF3, as well as promoting the K48-linked polyubiquitination and degradation of both IKKi and IRF3. This study may be helpful for fully understanding the pathogenesis of HSV-1.


Asunto(s)
Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Interferón beta/genética , Interferón beta/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Animales , Chlorocebus aethiops , Citocinas , Proteína 58 DEAD Box , Células HEK293 , Células HeLa , Herpes Simple , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón , Interferón Tipo I , Interferón beta/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores Inmunológicos , Transducción de Señal , Células Vero , Proteínas Virales/metabolismo
4.
J Virol ; 96(4): e0151021, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34935440

RESUMEN

Recent studies have demonstrated that the signaling activity of the cytosolic pathogen sensor retinoic acid-inducible gene-I (RIG-I) is modulated by a variety of posttranslational modifications (PTMs) to fine-tune the antiviral type I interferon (IFN) response. Whereas K63-linked ubiquitination of the RIG-I caspase activation and recruitment domains (CARDs) catalyzed by TRIM25 or other E3 ligases activates RIG-I, phosphorylation of RIG-I at S8 and T170 represses RIG-I signal transduction by preventing the TRIM25-RIG-I interaction and subsequent RIG-I ubiquitination. While strategies to suppress RIG-I signaling by interfering with its K63-polyubiquitin-dependent activation have been identified for several viruses, evasion mechanisms that directly promote RIG-I phosphorylation to escape antiviral immunity are unknown. Here, we show that the serine/threonine (Ser/Thr) kinase US3 of herpes simplex virus 1 (HSV-1) binds to RIG-I and phosphorylates RIG-I specifically at S8. US3-mediated phosphorylation suppressed TRIM25-mediated RIG-I ubiquitination, RIG-I-MAVS binding, and type I IFN induction. We constructed a mutant HSV-1 encoding a catalytically-inactive US3 protein (K220A) and found that, in contrast to the parental virus, the US3 mutant HSV-1 was unable to phosphorylate RIG-I at S8 and elicited higher levels of type I IFNs, IFN-stimulated genes (ISGs), and proinflammatory cytokines in a RIG-I-dependent manner. Finally, we show that this RIG-I evasion mechanism is conserved among the alphaherpesvirus US3 kinase family. Collectively, our study reveals a novel immune evasion mechanism of herpesviruses in which their US3 kinases phosphorylate the sensor RIG-I to keep it in the signaling-repressed state. IMPORTANCE Herpes simplex virus 1 (HSV-1) establishes lifelong latency in the majority of the human population worldwide. HSV-1 occasionally reactivates to produce infectious virus and to facilitate dissemination. While often remaining subclinical, both primary infection and reactivation occasionally cause debilitating eye diseases, which can lead to blindness, as well as life-threatening encephalitis and newborn infections. To identify new therapeutic targets for HSV-1-induced diseases, it is important to understand the HSV-1-host interactions that may influence infection outcome and disease. Our work uncovered direct phosphorylation of the pathogen sensor RIG-I by alphaherpesvirus-encoded kinases as a novel viral immune escape strategy and also underscores the importance of RNA sensors in surveilling DNA virus infection.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Herpesvirus Humano 1/inmunología , Evasión Inmune , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas Virales/metabolismo , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/fisiología , Secuencia de Aminoácidos , Proteína 58 DEAD Box/química , Células HEK293 , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Receptores Inmunológicos/química , Proteínas Virales/genética
5.
Cell Physiol Biochem ; 55(6): 726-738, 2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34816678

RESUMEN

Viruses have been widely used to treat cancer for many years and they achieved tremendous success in clinical trials with outstanding results, which has led to the foundation of companies that develop recombinant viruses for a better tumor treatment. Even though there has been a great progress in the field of viral tumor immunotherapy, until now only one virus, the oncolytic virus talimogene laherparepvec (TVEC), a genetically modified herpes simplex virus type 1 (T-VEC), has been approved by the FDA for cancer treatment. Although oncolytic viruses showed progress in certain cancer types and patient populations but they have yet shown limited efficacy when it comes to solid tumors. Only recently it was demonstrated that the immune stimulatory aspect of oncolytic viruses can strongly contribute to their anti-tumoral activity. One specific example in this context are arenaviruses, which have been shown to be non-cytopathic in nature lead to the massive immune activation within the tumor resulting in strong anti-tumoral activity. This strong immune activation might be also linked to their noncytopathic features, as their immune stimulatory potential is not self-limiting as is the case for oncolytic viruses due to their fast eradication by anti-viral immune effects. Because of this strong immune activation, arenaviruses appear superior to oncolytic viruses when it comes to potent and long-lasting anti-tumor effects in a broad variety of tumor types. Currently one of the most promising therapeutics which has turned to be very much beneficial for the treatment of different cancer types is represented by antibodies targeting checkpoint inhibitors such as PD-1/PD-L-1. In this review, we will summarize anti-tumoral effects of arenaviruses, and will discuss their potential to be combined with checkpoint inhibitors for a more efficient tumor treatment, which further emphasizes that arenavirus therapy as a viroimmunotherapy can be an efficient tool for the better clearance of tumors.


Asunto(s)
Arenavirus/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Productos Biológicos/inmunología , Productos Biológicos/uso terapéutico , Herpesvirus Humano 1/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/terapia
6.
Nature ; 600(7887): 138-142, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34759314

RESUMEN

Pathogens use virulence factors to inhibit the immune system1. The guard hypothesis2,3 postulates that hosts monitor (or 'guard') critical innate immune pathways such that their disruption by virulence factors provokes a secondary immune response1. Here we describe a 'self-guarded' immune pathway in human monocytes, in which guarding and guarded functions are combined in one protein. We find that this pathway is triggered by ICP0, a key virulence factor of herpes simplex virus type 1, resulting in robust induction of anti-viral type I interferon (IFN). Notably, induction of IFN by ICP0 is independent of canonical immune pathways and the IRF3 and IRF7 transcription factors. A CRISPR screen identified the ICP0 target MORC34 as an essential negative regulator of IFN. Loss of MORC3 recapitulates the IRF3- and IRF7-independent IFN response induced by ICP0. Mechanistically, ICP0 degrades MORC3, which leads to de-repression of a MORC3-regulated DNA element (MRE) adjacent to the IFNB1 locus. The MRE is required in cis for IFNB1 induction by the MORC3 pathway, but is not required for canonical IFN-inducing pathways. As well as repressing the MRE to regulate IFNB1, MORC3 is also a direct restriction factor of HSV-15. Our results thus suggest a model in which the primary anti-viral function of MORC3 is self-guarded by its secondary IFN-repressing function-thus, a virus that degrades MORC3 to avoid its primary anti-viral function will unleash the secondary anti-viral IFN response.


Asunto(s)
Adenosina Trifosfatasas/inmunología , Proteínas de Unión al ADN/inmunología , Modelos Inmunológicos , Factores de Virulencia/inmunología , Adenosina Trifosfatasas/deficiencia , Adenosina Trifosfatasas/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Edición Génica , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Proteínas Inmediatas-Precoces/inmunología , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Monocitos/inmunología , Receptor de Interferón alfa y beta , Proteínas Represoras/deficiencia , Proteínas Represoras/inmunología , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Ubiquitina-Proteína Ligasas/inmunología
7.
Nat Med ; 27(10): 1789-1796, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34608333

RESUMEN

Talimogene laherparepvec (T-VEC) is a herpes simplex virus type 1-based intralesional oncolytic immunotherapy approved for the treatment of unresectable melanoma. The present, ongoing study aimed to estimate the treatment effect of neoadjuvant T-VEC on recurrence-free survival (RFS) of patients with advanced resectable melanoma. An open-label, phase 2 trial (NCT02211131) was conducted in 150 patients with resectable stage IIIB-IVM1a melanoma who were randomized to receive T-VEC followed by surgery (arm 1, n = 76) or surgery alone (arm 2, n = 74). The primary endpoint was a 2-year RFS in the intention-to-treat population. Secondary and exploratory endpoints included overall survival (OS), pathological complete response (pCR), safety and biomarker analyses. The 2-year RFS was 29.5% in arm 1 and 16.5% in arm 2 (overall hazard ratio (HR) = 0.75, 80% confidence interval (CI) = 0.58-0.96). The 2-year OS was 88.9% for arm 1 and 77.4% for arm 2 (overall HR = 0.49, 80% CI = 0.30-0.79). The RFS and OS differences between arms persisted at 3 years. In arm 1, 17.1% achieved a pCR. Increased CD8+ density correlated with clinical outcomes in an exploratory analysis. Arm 1 adverse events were consistent with previous reports for T-VEC. The present study met its primary endpoint and estimated a 25% reduction in the risk of disease recurrence for neoadjuvant T-VEC plus surgery versus upfront surgery for patients with resectable stage IIIB-IVM1a melanoma.


Asunto(s)
Productos Biológicos/administración & dosificación , Inmunoterapia , Melanoma/terapia , Terapia Neoadyuvante , Adulto , Anciano , Productos Biológicos/inmunología , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Humanos , Masculino , Melanoma/genética , Melanoma/patología , Melanoma/virología , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/terapia , Recurrencia Local de Neoplasia/virología , Estadificación de Neoplasias , Viroterapia Oncolítica/tendencias , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología
8.
PLoS Pathog ; 17(10): e1009999, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34653236

RESUMEN

Ocular HSV-1 infection is a major cause of eye disease and innate and adaptive immunity both play a role in protection and pathology associated with ocular infection. Previously we have shown that M1-type macrophages are the major and earliest infiltrates into the cornea of infected mice. We also showed that HSV-1 infectivity in the presence and absence of M2-macrophages was similar to wild-type (WT) control mice. However, it is not clear whether the absence of M1 macrophages plays a role in protection and disease in HSV-1 infected mice. To explore the role of M1 macrophages in HSV-1 infection, we used mice lacking M1 activation (M1-/- mice). Our results showed that macrophages from M1-/- mice were more susceptible to HSV-1 infection in vitro than were macrophages from WT mice. M1-/- mice were highly susceptible to ocular infection with virulent HSV-1 strain McKrae, while WT mice were refractory to infection. In addition, M1-/- mice had higher virus titers in the eyes than did WT mice. Adoptive transfer of M1 macrophages from WT mice to M1-/- mice reduced death and rescued virus replication in the eyes of infected mice. Infection of M1-/- mice with avirulent HSV-1 strain KOS also increased ocular virus replication and eye disease but did not affect latency-reactivation seen in WT control mice. Severity of virus replication and eye disease correlated with significantly higher inflammatory responses leading to a cytokine storm in the eyes of M1-/- infected mice that was not seen in WT mice. Thus, for the first time, our study illustrates the importance of M1 macrophages specifically in primary HSV-1 infection, eye disease, and survival but not in latency-reactivation.


Asunto(s)
Síndrome de Liberación de Citoquinas/inmunología , Queratitis Herpética/inmunología , Macrófagos/inmunología , Animales , Herpesvirus Humano 1/inmunología , Ratones , Activación Viral/inmunología , Latencia del Virus/inmunología
9.
Nat Commun ; 12(1): 5908, 2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34625564

RESUMEN

Oncolytic herpes simplex virus-1 is capable of lysing tumor cells while alerting the immune system. CD47, in collaboration with SIRPα, represents an important immune checkpoint to inhibit phagocytosis by innate immune cells. Here we show locoregional control of glioblastoma by an oncolytic herpes virus expressing a full-length anti(α)-human CD47 IgG1 or IgG4 antibody. The antibodies secreted by the virus-infected glioblastoma cells block the CD47 'don't eat me' signal irrespective of the subclass; however, αCD47-IgG1 has a stronger tumor killing effect than αCD47-IgG4 due to additional antibody-dependent cellular phagocytosis by macrophages and antibody-dependent cellular cytotoxicity by NK cells. Intracranially injected αCD47-IgG1-producing virus continuously releases the respective antibody in the tumor microenvironment but not into systemic circulation; additionally, αCD47-IgG1-producing virus also improves the survival of tumor-bearing mice better than control oncolytic herpes virus combined with topical αCD47-IgG1. Results from immunocompetent mouse tumor models further confirm that macrophages, and to a lesser extent NK cells, mediate the anti-tumor cytotoxicity of antibody-producing oncolytic herpesviruses. Collectively, oncolytic herpes simplex virus-1 encoding full-length antibodies could improve immune-virotherapy for glioblastoma.


Asunto(s)
Anticuerpos/farmacología , Glioblastoma/inmunología , Glioblastoma/terapia , Inmunidad Innata , Virus Oncolíticos/inmunología , Animales , Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígeno CD47 , Modelos Animales de Enfermedad , Femenino , Herpesvirus Humano 1/inmunología , Humanos , Inmunoglobulina G , Inmunoterapia , Células Asesinas Naturales , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Viroterapia Oncolítica/métodos , Fagocitosis , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
PLoS Pathog ; 17(9): e1009950, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34591940

RESUMEN

STING is a nodal point for cellular innate immune response to microbial infections, autoimmunity and cancer; it triggers the synthesis of the antiviral proteins, type I interferons. Many DNA viruses, including Herpes Simplex Virus 1 (HSV1), trigger STING signaling causing inhibition of virus replication. Here, we report that HSV1 evades this antiviral immune response by inducing a cellular microRNA, miR-24, which binds to the 3' untranslated region of STING mRNA and inhibits its translation. Expression of the gene encoding miR-24 is induced by the transcription factor AP1 and activated by MAP kinases in HSV1-infected cells. Introduction of exogenous miR-24 or prior activation of MAPKs, causes further enhancement of HSV1 replication in STING-expressing cells. Conversely, transfection of antimiR-24 inhibits virus replication in those cells. HSV1 infection of mice causes neuropathy and death; using two routes of infection, we demonstrated that intracranial injection of antimiR-24 alleviates both morbidity and mortality of the infected mice. Our studies reveal a new immune evasion strategy adopted by HSV1 through the regulation of STING and demonstrates that it can be exploited to enhance STING's antiviral action.


Asunto(s)
Herpes Simple/inmunología , Evasión Inmune/inmunología , Proteínas de la Membrana/inmunología , MicroARNs/inmunología , Animales , Regulación de la Expresión Génica/inmunología , Herpes Simple/metabolismo , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Celular/inmunología , Proteínas de la Membrana/metabolismo , Ratones
11.
Viruses ; 13(9)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34578328

RESUMEN

We report on the efficacy of the non-attenuated HER2-retargeted oHSV named R-337 against the immunologically hot CT26-HER2 tumor, and an insight into the basis of the immune protection. Preliminarily, we conducted an RNA immune profiling and immune cell content characterization of CT26-HER2 tumor in comparison to the immunologically cold LLC1-HER2 tumor. CT26-HER2 tumor was implanted into HER2-transgenic BALB/c mice. Hallmarks of R-337 effects were the protection from primary tumor, long-term adaptive vaccination directed to both HER2 and CT26-wt cell neoantigens. The latter effect differentiated R-337 from OncoVEXGM-CSF. As to the basis of the immune protection, R-337 orchestrated several changes to the tumor immune profile, which cumulatively reversed the immunosuppression typical of this tumor (graphical abstract). Thus, Ido1 (inhibitor of T cell anticancer immunity) levels and T regulatory cell infiltration were decreased; Cd40 and Cd27 co-immunostimulatory markers were increased; the IFNγ cascade was activated. Of note was the dampening of IFN-I response, which we attribute to the fact that R-337 is fully equipped with genes that contrast the host innate response. The IFN-I shut-down likely favored viral replication and the expression of the mIL-12 payload, which, in turn, boosted the antitumor response. The results call for a characterization of tumor immune markers to employ oncolytic herpesviruses more precisely.


Asunto(s)
Genotipo , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Inmunidad , Inmunoterapia/métodos , Neoplasias/inmunología , Virus Oncolíticos/inmunología , Animales , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Virus Oncolíticos/patogenicidad , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Replicación Viral
12.
Neurosci Lett ; 765: 136172, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34433098

RESUMEN

BACKGROUND: Herpes simplex virus encephalitis (HSE) is an acute central nervous system infectious disease caused by herpes simplex virus (HSV). Currently, there is no effective treatment for HSE infection, which produces many pro-inflammatory factors. Kaempferol-3-O-rhamnoside (K-3-rh) is a plant flavonoid. This study was investigated the anti-inflammatory effect of K-3-rh on encephalitis induced by HSV-1. METHODS: HSV-1 was co-cultured with VERO cells. Cells were divided into four groups, including the control group, virus group, K-3-rh group, Astragalus polysaccharide (APS) group and dexamethasone group. Flow cytometry were utilized to determine cell apoptosis, respectively. Proteins and mRNAs were estimated by western blot and qRT-PCR, respectively. RESULTS: After viral infection, the cytokines were significantly increased. After K-3-rh intervention, the expression of tumor necrosis factor-α (TNF-α), interleukin-1 beta (IL-1ß), and nitric oxide (NO) in microglia were reduced contrast with those in the virus group, and the expression of interleukin-10 (IL-10) did not change. After viral infection, the apoptotic rate increased significantly, and K-3-rh could inhibit viral-induced apoptosis in the microglial cell line. The induction of microglia apoptosis was achieved by cytochrome c and caspase-9-mediated mitochondrial pathway. Also, the pathological changes of brain tissue in mice of each drug intervention group were alleviated. CONCLUSIONS: In conclusion, K-3-rh had the potential to reduce HSV-1-induced brain injury by reducing the secretion of microglial pro-inflammatory factors, inducing apoptosis of microglia cells, and through cytochrome C and caspase-3 pathway.


Asunto(s)
Encefalitis por Herpes Simple/tratamiento farmacológico , Glicósidos/farmacología , Herpesvirus Humano 1/inmunología , Quempferoles/farmacología , Microglía/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Caspasa 3/metabolismo , Línea Celular , Chlorocebus aethiops , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Encefalitis por Herpes Simple/inmunología , Encefalitis por Herpes Simple/patología , Encefalitis por Herpes Simple/virología , Glicósidos/uso terapéutico , Humanos , Quempferoles/uso terapéutico , Ratones , Microglía/inmunología , Microglía/patología , Microglía/virología , Células Vero
13.
Viruses ; 13(7)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34372614

RESUMEN

Oncolytic herpes simplex virus (oHSV) is a therapeutic modality that has seen substantial success for the treatment of cancer, though much remains to be improved. Commonly attenuated through the deletion or alteration of the γ134.5 neurovirulence gene, the basis for the success of oHSV relies in part on the malignant silencing of cellular pathways critical for limiting these viruses in healthy host tissue. However, only recently have the molecular mechanisms underlying the success of these treatments begun to emerge. Further clarification of these mechanisms can strengthen rational design approaches to develop the next generation of oHSV. Herein, we review our current understanding of the molecular basis for tumor susceptibility to γ134.5-attenuated oHSV, with particular focus on the malignant suppression of nucleic acid sensing, along with strategies meant to improve the clinical efficacy of these therapeutic viruses.


Asunto(s)
Herpesvirus Humano 1/fisiología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Animales , Citocinas/inmunología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Interacciones Microbiota-Huesped/inmunología , Humanos , Ratones
14.
J Virol ; 95(22): e0105021, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34431697

RESUMEN

The viral fusion protein glycoprotein B (gB) is conserved in all herpesviruses and is essential for virus entry. During entry, gB fuses viral and host cell membranes by refolding from a prefusion to a postfusion form. We previously introduced three structure-based mutations (gB-I671A/H681A/F683A) into the domain V arm of the gB ectodomain that resulted in reduced cell-cell fusion. A virus carrying these three mutations (called gB3A) displayed a small-plaque phenotype and remarkably delayed entry into cells. To identify mutations that could counteract this phenotype, we serially passaged the gB3A virus and selected for revertant viruses with increased plaque sizes. Genomic sequencing revealed that the revertant viruses had second-site mutations in gB, including E187A, M742T, and S383F/G645R/V705I/V880G. Using expression constructs encoding these mutations, only gB-V880G was shown to enhance cell-cell fusion. In contrast, all of the revertant viruses showed enhanced entry kinetics, underscoring the fact that cell-cell fusion and virus-cell fusion are different. The results indicate that mutations in three different regions of gB (domain I, the membrane proximal region, and the cytoplasmic tail domain) can counteract the slow-entry phenotype of gB3A virus. Mapping these compensatory mutations to prefusion and postfusion structural models suggests sites of intramolecular functional interactions with the gB domain V arm that may contribute to the gB fusion function. IMPORTANCE The nine human herpesviruses are ubiquitous and cause a range of diseases in humans. Glycoprotein B (gB) is an essential viral fusion protein that is conserved in all herpesviruses. During host cell entry, gB mediates virus-cell membrane fusion by undergoing a conformational change. Structural models for the prefusion and postfusion forms of gB exist, but the details of how the protein converts from one to the other are unclear. We previously introduced structure-based mutations into gB that inhibited virus entry and fusion. By passaging this entry-deficient virus over time, we selected second-site mutations that partially restore virus entry. The locations of these mutations suggest regulatory sites that contribute to fusion and gB refolding during entry. gB is a target of neutralizing antibodies, and defining how gB refolds during entry could provide a basis for the development of fusion inhibitors for future research or clinical use.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1 , Proteínas del Envoltorio Viral , Internalización del Virus , Animales , Células CHO , Chlorocebus aethiops , Cricetulus , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Fusión de Membrana , Modelos Moleculares , Mutación , Conformación Proteica , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
15.
J Immunol ; 207(2): 613-625, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34272232

RESUMEN

Alphaherpesviruses are large dsDNA viruses with an ability to establish persistent infection in hosts, which rely partly on their ability to evade host innate immune responses, notably the type I IFN response. However, the relevant molecular mechanisms are not well understood. In this study, we report the UL42 proteins of alphaherpesvirus pseudorabies virus (PRV) and HSV type 1 (HSV1) as a potent antagonist of the IFN-I-induced JAK-STAT signaling pathway. We found that ectopic expression of UL42 in porcine macrophage CRL and human HeLa cells significantly suppresses IFN-α-mediated activation of the IFN-stimulated response element (ISRE), leading to a decreased transcription and expression of IFN-stimulated genes (ISGs). Mechanistically, UL42 directly interacts with ISRE and interferes with ISG factor 3 (ISGF3) from binding to ISRE for efficient gene transcription, and four conserved DNA-binding sites of UL42 are required for this interaction. The substitution of these DNA-binding sites with alanines results in reduced ISRE-binding ability of UL42 and impairs for PRV to evade the IFN response. Knockdown of UL42 in PRV remarkably attenuates the antagonism of virus to IFN in porcine kidney PK15 cells. Our results indicate that the UL42 protein of alphaherpesviruses possesses the ability to suppress IFN-I signaling by preventing the association of ISGF3 and ISRE, thereby contributing to immune evasion. This finding reveals UL42 as a potential antiviral target.


Asunto(s)
ADN Polimerasa Dirigida por ADN/inmunología , Exodesoxirribonucleasas/inmunología , Herpesvirus Suido 1/inmunología , Interferón Tipo I/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Proteínas Virales/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Células HEK293 , Células HeLa , Herpesvirus Humano 1/inmunología , Humanos , Evasión Inmune/inmunología , Inmunidad Innata/inmunología , Seudorrabia/inmunología , Elementos de Respuesta/inmunología , Transducción de Señal/inmunología , Porcinos , Transcripción Genética/inmunología
16.
mSphere ; 6(4): e0060821, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34319127

RESUMEN

Atopic dermatitis (AD) is a condition affecting 30 million persons in the United States. AD patients are heavily infected with Staphylococcus aureus on the skin. A particularly severe form of AD is eczema herpeticum (ADEH), where the patients' AD is complicated by S. aureus and herpes simplex virus (HSV) infection. This study examined the S. aureus strains from 15 ADEH patients, provided blinded, and showed a high association of ADEH with strains that produce toxic shock syndrome toxin-1 (TSST-1; 73%) compared to 10% production by typical AD isolates from patients without EH and those from another unrelated condition, cystic fibrosis. The ADEH isolates produced the superantigens associated with TSS (TSST-1 and staphylococcal enterotoxins A, B, and C). This association may in part explain the potential severity of ADEH. We also examined the effect of TSST-1 and HSV-1 on human epithelial cells and keratinocytes. TSST-1 used CD40 as its receptor on epithelial cells, and HSV-1 either directly or indirectly interacted with CD40. The consequence of these interactions was chemokine production, which is capable of causing harmful inflammation, with epidermal/keratinocyte barrier disruption. Human epithelial cells treated first with TSST-1 and then HSV-1 resulted in enhanced chemokine production. Finally, we showed that TSST-1 modestly increased HSV-1 replication but did not increase viral plaque size. Our data suggest that ADEH is associated with production of the major TSS-associated superantigens, together with HSV reactivation. The superantigens plus HSV may damage the skin barrier by causing harmful inflammation, thereby leading to increased symptoms. IMPORTANCE Atopic dermatitis (eczema, AD) with concurrent herpes simplex virus infection (eczema herpeticum, ADEH) is a severe form of AD. We show that ADEH patients are colonized with Staphylococcus aureus that primarily produces the superantigen toxic shock syndrome toxin-1 (TSST-1); however, significantly but to a lesser extent the superantigens staphylococcal enterotoxins A, B, and C are also represented in ADEH. Our studies showed that TSST-1 uses the immune costimulatory molecule CD40 as its epithelial cell receptor. Herpes simplex virus (HSV) also interacted directly or indirectly with CD40 on epithelial cells. Treatment of epithelial cells with TSST-1 and then HSV-1 resulted in enhanced chemokine production. We propose that this combination of exposures (TSST-1 and then HSV) leads to opening of epithelial and skin barriers to facilitate potentially serious ADEH.


Asunto(s)
Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Enterotoxinas/genética , Enterotoxinas/metabolismo , Herpesvirus Humano 1/metabolismo , Erupción Variceliforme de Kaposi/microbiología , Staphylococcus aureus/patogenicidad , Superantígenos/genética , Superantígenos/metabolismo , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/farmacología , Antígenos CD40/inmunología , Quimiocinas/inmunología , Enterotoxinas/inmunología , Enterotoxinas/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Células Epiteliales/virología , Células HaCaT , Herpesvirus Humano 1/inmunología , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/microbiología , Queratinocitos/virología , Staphylococcus aureus/metabolismo , Superantígenos/inmunología , Superantígenos/farmacología
17.
Cells ; 10(5)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34064516

RESUMEN

Sphingolipids are important structural membrane components and, together with cholesterol, are often organized in lipid rafts, where they act as signaling molecules in many cellular functions. They play crucial roles in regulating pathobiological processes, such as cancer, inflammation, and infectious diseases. The bioactive metabolites ceramide, sphingosine-1-phosphate, and sphingosine have been shown to be involved in the pathogenesis of several microbes. In contrast to ceramide, which often promotes bacterial and viral infections (for instance, by mediating adhesion and internalization), sphingosine, which is released from ceramide by the activity of ceramidases, kills many bacterial, viral, and fungal pathogens. In particular, sphingosine is an important natural component of the defense against bacterial pathogens in the respiratory tract. Pathologically reduced sphingosine levels in cystic fibrosis airway epithelial cells are normalized by inhalation of sphingosine, and coating plastic implants with sphingosine prevents bacterial infections. Pretreatment of cells with exogenous sphingosine also prevents the viral spike protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) from interacting with host cell receptors and inhibits the propagation of herpes simplex virus type 1 (HSV-1) in macrophages. Recent examinations reveal that the bactericidal effect of sphingosine might be due to bacterial membrane permeabilization and the subsequent death of the bacteria.


Asunto(s)
Infecciones Bacterianas/inmunología , Micosis/inmunología , Transducción de Señal/inmunología , Esfingosina/metabolismo , Virosis/inmunología , Animales , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Pared Celular/efectos de los fármacos , Ceramidas/metabolismo , Modelos Animales de Enfermedad , Herpesvirus Humano 1/inmunología , Humanos , Lisofosfolípidos/metabolismo , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Micosis/tratamiento farmacológico , Micosis/metabolismo , Micosis/microbiología , SARS-CoV-2/inmunología , Esfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología , Esfingosina/uso terapéutico , Virosis/tratamiento farmacológico , Virosis/metabolismo , Virosis/virología
18.
Cancer Lett ; 518: 49-58, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34139284

RESUMEN

Oncolytic viruses as cancer vaccines modulate the tumor microenvironment and act synergistically with immune checkpoint inhibitors to overcome resistance. Taking advantage of the loading capacity for exogenous genes, we generated a recombinant herpes simplex virus type 1 (HSV-1), HSV-aPD-1, carrying a full-length humanized anti-PD-1 monoclonal antibody (anti-PD-1 mAb) that replicates and expresses anti-PD-1 mAbs in tumor cells in vitro and in vivo. Its anti-tumor effect was assessed in human PD-1 knock-in mice by analyzing tumor inhibition, cell populations and RNA expression in tumors, and serum cytokine levels. Enhanced anti-tumor immune responses and T-cell infiltration were induced by HSV-aPD-1 compared with unloaded virus or anti-PD-1 therapy in both MC38 and B16-F10 models, resulting in improved treatment efficacy in the latter. Moreover, compared with unloaded HSV-1 or HSV-1 loaded with GM-CSF/IL-2 combined with anti-PD-1 mAbs, HSV-aPD-1 displayed similar therapeutic control of tumor growth. Finally, tumor RNAseq analysis in the B16-F10 model showed upregulated IFN pathway and antigen processing and presentation genes, and downregulated angiogenesis and cell adhesion genes, which all contribute to tumor inhibition. These findings indicate the clinical potential of HSV-aPD-1 as monotherapy or combination therapy, especially in tumors resistant to immune checkpoint inhibitors.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Herpesvirus Humano 1/inmunología , Neoplasias/inmunología , Virus Oncolíticos/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Células A549 , Animales , Presentación de Antígeno/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Terapia Combinada/métodos , Femenino , Células Hep G2 , Humanos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/virología , Neovascularización Patológica/inmunología , Neovascularización Patológica/virología , Microambiente Tumoral/inmunología , Células Vero
19.
J Neuroimmunol ; 358: 577636, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34174587

RESUMEN

Herpesviruses like Epstein-Barr virus, human herpesvirus (HHV)-6, HHV-1, VZV, and human endogenous retroviruses, have an age-old clinical association with multiple sclerosis (MS). MS is an autoimmune disease of the nervous system wherein the myelin sheath deteriorates. The most popular mode of virus mediated immune system manipulation is molecular mimicry. Numerous herpesvirus antigens are similar to myelin proteins. Other mechanisms described here include the activity of cytokines and autoantibodies produced by the autoreactive T and B cells, respectively, viral déjà vu, epitope spreading, CD46 receptor engagement, impaired remyelination etc. Overall, this review addresses the host-parasite association of viruses with MS.


Asunto(s)
Autoanticuerpos/inmunología , Herpesviridae/inmunología , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/inmunología , Autoanticuerpos/sangre , Herpesviridae/metabolismo , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 3/inmunología , Herpesvirus Humano 3/metabolismo , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 6/inmunología , Herpesvirus Humano 6/metabolismo , Humanos , Esclerosis Múltiple/sangre
20.
Front Immunol ; 12: 662234, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34012447

RESUMEN

Herpes simplex virus type 1 (HSV-1) infection is highly prevalent in humans, with approximately two-thirds of the world population living with this virus. However, only a fraction of those carrying HSV-1, which elicits lifelong infections, are symptomatic. HSV-1 mainly causes lesions in the skin and mucosae but reaches the termini of sensory neurons innervating these tissues and travels in a retrograde manner to the neuron cell body where it establishes persistent infection and remains in a latent state until reactivated by different stimuli. When productive reactivations occur, the virus travels back along axons to the primary infection site, where new rounds of replication are initiated in the skin, in recurrent or secondary infections. During this process, new neuron infections occur. Noteworthy, the mechanisms underlying viral reactivations and the exit of latency are somewhat poorly understood and may be regulated by a crosstalk between the infected neurons and components of the immune system. Here, we review and discuss the immune responses that occur at the skin during primary and recurrent infections by HSV-1, as well as at the interphase of latently-infected neurons. Moreover, we discuss the implications of neuronal signals over the priming and migration of immune cells in the context of HSV-1 infection.


Asunto(s)
Células Epiteliales/metabolismo , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Células Receptoras Sensoriales/metabolismo , Enfermedades Cutáneas Virales/inmunología , Animales , Técnicas de Cultivo de Célula , Células Epiteliales/inmunología , Regulación Viral de la Expresión Génica , Herpesvirus Humano 1/genética , Humanos , Ratones , Células Receptoras Sensoriales/inmunología , Activación Viral , Latencia del Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA