Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 55(11): 2135-2148.e6, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36306784

RESUMEN

Epstein-Barr virus (EBV) is nearly ubiquitous in adults. EBV causes infectious mononucleosis and is associated with B cell lymphomas, epithelial cell malignancies, and multiple sclerosis. The EBV gH/gL glycoprotein complex facilitates fusion of virus membrane with host cells and is a target of neutralizing antibodies. Here, we examined the sites of vulnerability for virus neutralization and fusion inhibition within EBV gH/gL. We developed a panel of human monoclonal antibodies (mAbs) that targeted five distinct antigenic sites on EBV gH/gL and prevented infection of epithelial and B cells. Structural analyses using X-ray crystallography and electron microscopy revealed multiple sites of vulnerability and defined the antigenic landscape of EBV gH/gL. One mAb provided near-complete protection against viremia and lymphoma in a humanized mouse EBV challenge model. Our findings provide structural and antigenic knowledge of the viral fusion machinery, yield a potential therapeutic antibody to prevent EBV disease, and emphasize gH/gL as a target for herpesvirus vaccines and therapeutics.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Cricetinae , Ratones , Animales , Humanos , Proteínas del Envoltorio Viral , Cricetulus , Glicoproteínas de Membrana , Células CHO
2.
J Virol ; 96(8): e0007522, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35348362

RESUMEN

Epstein-Barr virus (EBV) is an oncogenic herpesvirus that is associated with 200,000 new cases of cancer and 140,000 deaths annually. To date, there are no available vaccines or therapeutics for clinical usage. Recently, the viral heterodimer glycoprotein gH/gL has become a promising target for the development of prophylactic vaccines against EBV. Here, we developed the anti-gH antibody 6H2 and its chimeric version C6H2, which had full neutralizing activity in epithelial cells and partial neutralizing activity in B cells. C6H2 exhibited potent protection against lethal EBV challenge in a humanized mouse model. The cryo-electron microscopy (cryo-EM) structure further revealed that 6H2 recognized a previously unidentified epitope on gH/gL D-IV that is critical for viral attachment and subsequent membrane fusion with epithelial cells. Our results suggest that C6H2 is a promising candidate in the prevention of EBV-induced lymphoproliferative diseases (LPDs) and may inform the design of an EBV vaccine. IMPORTANCE Epstein-Barr virus (EBV) is a ubiquitous gammaherpesvirus that establishes lifelong persistence and is related to multiple diseases, including cancers. Neutralizing antibodies (NAbs) have proven to be highly effective in preventing EBV infection and subsequent diseases. Here, we developed an anti-EBV-gH NAb, 6H2, which blocked EBV infection in vitro and in vivo. This 6H2 neutralizing epitope should be helpful to understand EBV infection mechanisms and guide the development of vaccines and therapeutics against EBV infection.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Proteínas del Envoltorio Viral , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Microscopía por Crioelectrón , Epítopos/química , Infecciones por Virus de Epstein-Barr/prevención & control , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/metabolismo , Ratones , Vacunas , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología
3.
J Virol ; 95(18): e0065721, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34160252

RESUMEN

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that causes severe clinical disease in immunosuppressed patients and congenitally infected newborn infants. Viral envelope glycoproteins represent attractive targets for vaccination or passive immunotherapy. To extend the knowledge of mechanisms of virus neutralization, monoclonal antibodies (MAbs) were generated following immunization of mice with HCMV virions. Hybridoma supernatants were screened for in vitro neutralization activity, yielding three potent MAbs, 6E3, 3C11, and 2B10. MAbs 6E3 and 3C11 blocked infection of all viral strains that were tested, while MAb 2B10 neutralized only 50% of the HCMV strains analyzed. Characterization of the MAbs using indirect immunofluorescence analyses demonstrated their reactivity with recombinantly derived gH. While MAbs 6E3 and 3C11 reacted with gH when expressed alone, 2B10 detected gH only when it was coexpressed with gB and gL. Recognition of gH by 3C11 was dependent on the expression of the entire ectodomain of gH, whereas 6E3 required residues 1 to 629 of gH. The strain-specific determinant for neutralization by Mab 2B10 was identified as a single Met→Ile amino acid polymorphism within gH, located within the central part of the protein. The polymorphism is evenly distributed among described HCMV strains. The 2B10 epitope thus represents a novel strain-specific antibody target site on gH of HCMV. The dependence of the reactivity of 2B10 on the simultaneous presence of gB/gH/gL will be of value in the structural definition of this tripartite complex. The 2B10 epitope may also represent a valuable tool for diagnostics to monitor infections/reinfections with different HCMV strains during pregnancy or after transplantation. IMPORTANCE HCMV infections are life threatening to people with compromised or immature immune systems. Understanding the antiviral antibody repertoire induced during HCMV infection is a necessary prerequisite to define protective antibody responses. Here, we report three novel anti-gH MAbs that potently neutralized HCMV infectivity. One of these MAbs (2B10) targets a novel strain-specific conformational epitope on gH that only becomes accessible upon coexpression of the minimal fusion machinery gB/gH/gL. Strain specificity is dependent on a single amino acid polymorphism within gH. Our data highlight the importance of strain-specific neutralizing antibody responses against HCMV. The 2B10 epitope may also represent a valuable tool for diagnostics to monitor infections/reinfections with different HCMV strains during pregnancy or after transplantation. In addition, the dependence of the reactivity of 2B10 on the simultaneous presence of gB/gH/gL will be of value in the structural definition of this tripartite complex.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Epítopos/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Citomegalovirus/clasificación , Infecciones por Citomegalovirus/virología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C
4.
Fetal Pediatr Pathol ; 41(1): 77-86, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32479132

RESUMEN

Objective: We determined the prevalence and relationship of glycoprotein B (gB), glycoprotein N (gN), and glycoprotein H (gH) genotypes of cytomegalovirus (CMV) in CMV-associated thrombocytopenia (CAP). Methods: CMV gB, gN, and gH strains were determined by nested PCR and restriction length polymorphism from 24 CAP and 20 asymptomatic CMV infected infants. Results: The order of prevalence was gB1 (70.8%,17/24), gN4 (45.8%,11/24) and gH2 (54.2%,13/24). There was a greater prevalence of gB1(75.0%,15/20), gN4(50.0%,10/20) and gN2 (35.0%,7/20) in moderate to severe infection (p = 0.014 and p = 0.003). By logistic regression, gH2 (p = 0.031) had an elevated risk of thrombocytopenia. Reduced risks of thrombocytopenia were associated with gB2 (p = 0.020), gN1 (p = 0.018) and gN3 (p = 0.008). The most virulent were gB1 (p = 0.033) and gN2 (p = 0.038). Conclusions: There may be a potential association between the gH2 genotype of CMV and infantile thrombocytopenia.


Asunto(s)
Infecciones por Citomegalovirus , Trombocitopenia , Citomegalovirus/genética , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/epidemiología , Femenino , Genotipo , Humanos , Lactante , Polimorfismo de Longitud del Fragmento de Restricción , Embarazo , Trombocitopenia/genética
5.
J Virol ; 93(16)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31142670

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV) is an emerging pathogen and is the causative infectious agent of Kaposi sarcoma and two malignancies of B cell origin. To date, there is no licensed KSHV vaccine. Development of an effective vaccine against KSHV continues to be limited by a poor understanding of how the virus initiates acute primary infection in vivo in diverse human cell types. The role of glycoprotein H (gH) in herpesvirus entry mechanisms remains largely unresolved. To characterize the requirement for KSHV gH in the viral life cycle and in determination of cell tropism, we generated and characterized a mutant KSHV in which expression of gH was abrogated. Using a bacterial artificial chromosome containing a complete recombinant KSHV genome and recombinant DNA technology, we inserted stop codons into the gH coding region. We used electron microscopy to reveal that the gH-null mutant virus assembled and exited from cells normally, compared to wild-type virus. Using purified virions, we assessed infectivity of the gH-null mutant in diverse mammalian cell types in vitro Unlike wild-type virus or a gH-containing revertant, the gH-null mutant was unable to infect any of the epithelial, endothelial, or fibroblast cell types tested. However, its ability to infect B cells was equivocal and remains to be investigated in vivo due to generally poor infectivity in vitro Together, these results suggest that gH is critical for KSHV infection of highly permissive cell types, including epithelial, endothelial, and fibroblast cells.IMPORTANCE All homologues of herpesvirus gH studied to date have been implicated in playing an essential role in viral infection of diverse permissive cell types. However, the role of gH in the mechanism of KSHV infection remains largely unresolved. In this study, we generated a gH-null mutant KSHV and provided evidence that deficiency of gH expression did not affect viral particle assembly or egress. Using the gH-null mutant, we showed that gH was indispensable for KSHV infection of epithelial, endothelial, and fibroblast cells in vitro This suggests that gH is an important target for the development of a KSHV prophylactic vaccine to prevent initial viral infection.


Asunto(s)
Células Endoteliales/virología , Células Epiteliales/virología , Fibroblastos/virología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/fisiología , Proteínas del Envoltorio Viral/genética , Tropismo Viral , Genoma Viral , Genómica/métodos , Humanos , Mutación , Proteínas del Envoltorio Viral/metabolismo , Virión , Internalización del Virus
6.
J Med Virol ; 92(12): 3674-3681, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32567060

RESUMEN

Human cytomegalovirus (HCMV) is the leading cause of congenital infection and an opportunistic pathogen capable of establishing lifelong latency. In the present study, we aimed to investigate the distribution of glycoprotein B, H, and N in infants of Shanghai and correlate the genotype with active and latent HCMV infection. A total of 129 urine samples were collected between August 2014 and December 2015 from infants under 3 years with HCMV infection. Nested PCR was used to amplify the regions of UL55 (gB), UL75 (gH), and UL73 (gN). Gene sequencing and phylogenetic analyses were used to classify the genotypes. Overall, regarding gB, gB1 (57.27%) was predominant, followed by gB3 (41.82%) and gB4 (0.91%). gH1 (54.33%) was the most prevalent genotype of gH, followed by gH2 (45.67%). Concerning gN, we detected gN1 (17.44%), gN2 (2.33%), gN3a (29.07%), gN3b (8.14%), gN4a (13.95%), gN4b (15.12%), and gN4c (13.95%), among which gN3a was the dominant genotype. All the expected genotypes were present except gB2 in children with active infection: gB1 (56.25%), gB3 (42.5%), gB4 (1.25%), gH1 (58.70%), gH2 (41.30%), gN1 (19.05%), gN2 (3.17%), gN3a (25.40%), gN3b (6.35%), gN4a (15.87%), gN4b (17.46%), and gN4c (12.70%). However, among latent cases, we detected gB1 (60%), gB3 (40%), gH1 (42.86%), gH2 (57.14%), gN1 (13.04%), gN3a (39.13%), gN3b (13.04%), gN4a (8.70%), gN4b (8.70%), and gN4c (17.39%), respectively. gB2, gB4, and gN2 were absent in this group. The results revealed that gB1, gH1, and gN3a were predominant in the infants of Shanghai. gH showed different trends among children with active and latent infection.

7.
BMC Vet Res ; 14(1): 255, 2018 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-30157854

RESUMEN

BACKGROUND: Duck Enteritis Virus (DEV), belonging to the α-herpesvirus subfamily, is a linear double-stranded DNA virus. Glycoprotein H and L (gH and gL), encoded by UL22 and UL1, are conserved in the family of herpesviruses. They play important roles as gH/gL dimers during viral entry into host cells through cell-cell fusion. The interaction between gH and gL has been confirmed in several human herpesviruses, such as Herpes Simplex Virus (HSV), Epstein-Barr virus (EBV) and Human Cytomegalovirus (HCMV). In this paper, we studied the interaction between DEV gH and gL. RESULTS: Recombinant plasmids pEGFP-N-gH and pDsRED-N-gL were constructed successfully. Expressions of both DEV gH and gL were observed after incubation of COS-7 cells transfected with pEGFP-N-gH and pDsRED-N-gL plasmids after 12 h, respectively. Also, the co-localization of a proportion of the gH and gL was detected in the cytoplasm of COS-7 cells after co-transfection for 24 h. Then, pCMV-Flag-gL and pCMV-Myc-gH recombinant plasmids were constructed and co-transfected into COS-7 cells. It was showed that both gH and gL were tested with positive results through co-immunoprecipitation and Western-blotting. CONCLUSIONS: Our results demonstrated not only the co-localization of DEV gH and gL in COS-7 cells, but also the interaction between them. It will provide an insight for the further studies in terms of protein-protein interaction in DEV.


Asunto(s)
Mardivirus/fisiología , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral/química , Animales , Células COS , Chlorocebus aethiops , Patos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
8.
Intervirology ; 59(5-6): 235-242, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28329739

RESUMEN

AIMS: The aim of this study was to design peptides derived from glycoproteins H (gH) and B (gB) of herpes simplex viruses type 1 (HSV-1) and type 2 (HSV-2) with the potential to block herpetic infection and to evaluate their ability to inhibit HSV-1 and HSV-2 infection in vitro. METHODS: A library of continuous 15-25 residue stretches (CRSs) located at the surface of gH and gB from HSV-1 and HSV-2 was created. These CRSs were analyzed, and only those that were highly flexible and rich in charged residues were selected for the design of the antiviral peptides (AVPs). The toxicity of the AVPs was evaluated by MTT reduction assays. Virucidal activity of the AVPs was determined by a plaque reduction assay, and their antiviral effect was measured by cell viability assays. RESULTS AND CONCLUSION: Four AVPs (CB-1, CB-2, U-1, and U-2) derived from gB and gH were designed and synthetized, none of which showed high levels of toxicity in Vero cells. The U-1 and U-2 gB-derived AVPs showed high virucidal and antiviral activities against both HSV-1 and HSV-2. The gH-derived peptide CB-1 showed high virucidal and antiviral activities against HSV-2, while CB-2 showed similar results against HSV-1. The peptides CB-1 and CB-2 showed higher IC50 values than the U-1 and U-2 peptides.

9.
Microbiol Immunol ; 59(6): 331-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25808324

RESUMEN

Herpes simplex virus 1 (HSV-1) envelope glycoprotein H (gH) is important for viral entry into cells and nuclear egress of nucleocapsids. To clarify additional novel roles of gH during HSV-1 replication, host cell proteins that interact with gH were screened for by tandem affinity purification coupled with mass spectrometry-based proteomics in 293T cells transiently expressing gH. This screen identified 123 host cell proteins as potential gH interactors. Of these proteins, general control nonderepressive-1 (GCN1), a trans-acting positive effector of GCN2 kinase that regulates phosphorylation of the α subunit of translation initiation factor 2 (eIF2α), was subsequently confirmed to interact with gH in HSV-1-infected cells. eIF2α phosphorylation is known to downregulate protein synthesis, and various viruses have evolved mechanisms to prevent the accumulation of phosphorylated eIF2α in infected cells. Here, it was shown that GCN1 knockdown reduces phosphorylation of eIF2α in HSV-1-infected cells and that the gH-null mutation increases eIF2α in HSV-1-infected cells, whereas gH overexpression in the absence of other HSV-1 proteins reduces eIF2α phosphorylation. These findings suggest that GCN1 can regulate eIF2α phosphorylation in HSV-1-infected cells and that the GCN1-binding viral partner gH is necessary and sufficient to prevent the accumulation of phosphorylated eIF2α. Our database of 123 host cell proteins potentially interacting with gH will be useful for future studies aimed at unveiling further novel functions of gH and the roles of cellular proteins in HSV-1-infected cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Herpesvirus Humano 1/fisiología , Mapas de Interacción de Proteínas , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Cromatografía de Afinidad , Células Epiteliales/química , Células Epiteliales/virología , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Espectrometría de Masas , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN , Transactivadores
10.
J Med Virol ; 86(8): 1421-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24615599

RESUMEN

Cytomegalovirus (CMV) is a leading cause of congenital infection and a leading infectious cause of hearing loss in children. The ORF UL75 gene encodes envelope glycoprotein H (gH), which is essential for CMV entry into host cells and the target of the immune response in humans. However, the distribution of gH variants and the relationship between the viral genotype, viral load, and sequelae in children infected with CMV is debated. The UL75 genetic variation of CMV isolates from 42 newborns infected congenitally with CMV and 93 infants with postnatal or unproven congenital CMV infection was analyzed. Genotyping was performed by analysis of PCR-amplified fragments, and the viral load was measured by quantitative real-time PCR. There were no differences in the distribution of gH genotypes in the children infected congenitally and postnatally. Mixed-genotype infections with both gH1 and gH2 variants were detected in approximately 25% of the examined patients. No relationship between UL75 gene polymorphisms and the symptoms at birth was observed. The results suggest that the infection with gH2 genotype diminishes the risk of hearing loss in children (P = 0.010). In addition, sensorineural hearing loss was associated with CMV gH1 genotype infection in infants (P = 0.032) and a high viral load in urine (P = 0.005). In conclusion, it was found that the gH genotype does not predict clinical sequelae in newborn infants following congenital CMV infection. However, these results suggest that the gH genotype might be associated with hearing loss in children.


Asunto(s)
Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/virología , Citomegalovirus/clasificación , Citomegalovirus/genética , Variación Genética , Pérdida Auditiva/epidemiología , Proteínas del Envoltorio Viral/genética , Adulto , Citomegalovirus/aislamiento & purificación , ADN Viral/genética , Femenino , Genotipo , Pérdida Auditiva/virología , Humanos , Lactante , Recién Nacido , Masculino , Reacción en Cadena de la Polimerasa , Carga Viral
11.
Mol Ther Oncol ; 32(1): 200778, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38596302

RESUMEN

To retarget oncolytic herpes simplex virus (oHSV) to cancer-specific antigens, we designed a novel, double-retargeted oHSV platform that uses single-chain antibodies (scFvs) incorporated into both glycoprotein H and a bispecific adapter expressed from the viral genome to mediate infection predominantly via tumor-associated antigens. Successful retargeting was achieved using a nectin-1-detargeted HSV that remains capable of interacting with herpesvirus entry mediator (HVEM), the second canonical HSV entry receptor, and is, therefore, recognized by the adapter consisting of the virus-binding N-terminal 82 residues of HVEM fused to the target-specific scFv. We tested both an epithelial cell adhesion molecule (EpCAM)- and a human epidermal growth factor receptor 2-specific scFv separately and together to target cells expressing one, the other, or both receptors. Our results show not only dose-dependent, target receptor-specific infection in vitro, but also enhanced virus spread compared with single-retargeted virus. In addition, we observed effective infection and spreading of the EpCAM double-retargeted virus in vivo. Remarkably, a single intravenous dose of the EpCAM-specific virus eliminated all detectable tumors in a subcutaneous xenograft model, and the same intravenous dose seemed to be harmless in immunocompetent FVB/N mice. Our findings suggest that our double-retargeted oHSV platform can provide a potent, versatile, and systemically deliverable class of anti-cancer therapeutics that specifically target cancer cells while ensuring safety.

12.
Front Pediatr ; 11: 1112645, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37009289

RESUMEN

Background: Cytomegalovirus (CMV) is the leading cause of congenital infections worldwide and contributes to long-term sequelae in neonates and children. CMV envelope glycoproteins play a vital role in virus entry and cell fusion. The association between CMV polymorphisms and clinical outcomes remains controversial. The present study aims to demonstrate the distribution of glycoprotein B (gB), H (gH) and N (gN) genotypes in congenitally CMV (cCMV) infected symptomatic infants and attempts to figure out the association between viral glycoprotein genotypes and clinical outcomes. Methods: Genotyping of gB, gH and gN was performed in 42 cCMV symptomatic infants and 149 infants with postnatal CMV (pCMV) infection in Children's hospital of Fudan university. Nested PCR, gene sequencing and phylogenetic analyses were used to identify the genotypes. Results: Our study demonstrated that: 1. The CMV gB1, gH1 and gN1 were the predominant genotypes among symptomatic cCMV infected infants, while gB1, gH1 and gN3a were more prevalent in pCMV group. gH1 genotype has a significant association with symptomatic cCMV infection (p = 0.006). 2. No significant correlation was found between CMV genotypes and hearing impairment. However, gH1 was more prevalent among cCMV infected infants with moderate/severe hearing loss although without statistical difference (p = 0.130). 3. gB3 was more prevalent among infants with skin petechiae (p = 0.049) and found to be associated with an increased risk of skin petechiae (OR = 6.563). The gN4a subtype was significantly associated with chorioretinitis due to cCMV infection (p = 0.007). 4. Urine viral loads were not significantly associated with different genotypes or hearing impairment among symptomatic cCMV infected infants. Conclusions: Our findings demonstrated the overall distribution of gB, gH and gN genotypes in infants with symptomatic cCMV infection in Shanghai for the first time. The findings in our study may suggest a possible association between gH1 genotype and early infancy hearing loss. gB3 genotype was associated with a 6.5-fold increased risk of petechiae while gN4a strongly correlated with chorioretinitis due to cCMV infection. No significant correlation was found between urine viral loads and CMV genotypes or hearing impairment in cCMV infected infants.

13.
mBio ; 14(1): e0336822, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36629412

RESUMEN

Enveloped virus entry requires fusion of the viral envelope with a host cell membrane. Herpes simplex virus 1 (HSV-1) entry is mediated by a set of glycoproteins that interact to trigger the viral fusion protein glycoprotein B (gB). In the current model, receptor-binding by gD signals a gH/gL heterodimer to trigger a refolding event in gB that fuses the membranes. To explore functional interactions between gB and gH/gL, we used a bacterial artificial chromosome (BAC) to generate two HSV-1 mutants that show a small plaque phenotype due to changes in gB. We passaged the viruses to select for restoration of plaque size and analyzed second-site mutations that arose in gH. HSV-1 gB was replaced either by gB from saimiriine herpesvirus 1 (SaHV-1) or by a mutant form of HSV-1 gB with three alanine substitutions in domain V (gB3A). To shift the selective pressure away from gB, the gB3A virus was passaged in cells expressing gB3A. Sequencing of passaged viruses identified two interesting mutations in gH, including gH-H789Y in domain IV and gH-S830N in the cytoplasmic tail (CT). Characterization of these gH mutations indicated they are responsible for the enhanced plaque size. Rather than being globally hyperfusogenic, both gH mutations partially rescued function of the specific gB version present during their selection. These sites may represent functional interaction sites on gH/gL for gB. gH-H789 may alter the positioning of a membrane-proximal flap in the gH ectodomain, whereas gH-S830 may contribute to an interaction between the gB and gH CTs. IMPORTANCE Enveloped viruses enter cells by fusing their envelope with the host cell membrane. Herpes simplex virus 1 (HSV-1) entry requires the coordinated interaction of several viral glycoproteins, including gH/gL and gB. gH/gL and gB are essential for virus replication and both proteins are targets of neutralizing antibodies. gB fuses the membranes after being activated by gH/gL, but the details of how gH/gL activates gB are not known. This study examined the gH/gL-gB interaction using HSV-1 mutants that displayed reduced virus entry due to changes in gB. The mutant viruses were grown over time to select for additional mutations that could partially restore entry. Two mutations in gH (H789Y and S830N) were identified. The positions of the mutations in gH/gL may represent sites that contribute to gB activation during virus entry.


Asunto(s)
Herpesvirus Humano 1 , Proteínas del Envoltorio Viral , Proteínas del Envoltorio Viral/metabolismo , Herpesvirus Humano 1/fisiología , Glicoproteínas/metabolismo , Proteínas Virales de Fusión/metabolismo , Unión Proteica , Internalización del Virus , Fusión de Membrana
14.
Viruses ; 13(8)2021 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-34452377

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV) is the causative agent of multiple cancers in immunocompromised patients including two lymphoproliferative disorders associated with KSHV infection of B lymphocytes. Despite many years of research into the pathogenesis of KSHV associated diseases, basic questions related to KSHV molecular virology remain unresolved. One such unresolved question is the cellular receptors and viral glycoproteins needed for KSHV entry into primary B lymphocytes. In this study, we assess the contributions of KSHV glycoprotein H (gH) and the cellular receptor DC-SIGN to KSHV infection in tonsil-derived B lymphocytes. Our results show that (1) neither KSHV-gH nor DC-SIGN are essential for entry into any B cell subset, (2) DC-SIGN does play a role in KSHV entry into tonsil-derived B cells, but in all B cell subtypes alternative entry mechanisms exist, (3) KSHV-gH can participate in KSHV entry into centrocytes via a DC-SIGN independent entry mechanism, and (4) in the absence of KSHV-gH, DC-SIGN is required for KSHV entry into centrocytes. Our results provide a first glimpse into the complexity of KSHV entry in the lymphocyte compartment and highlight that multiple subset-dependent entry mechanisms are employed by KSHV which depend upon multiple cellular receptors and multiple KSHV glycoproteins.


Asunto(s)
Linfocitos B/virología , Proteínas de la Cápside/genética , Moléculas de Adhesión Celular/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Interacciones Microbiota-Huesped , Lectinas Tipo C/genética , Receptores de Superficie Celular/genética , Internalización del Virus , Linfocitos B/inmunología , Células Cultivadas , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Tonsila Palatina/citología , Tonsila Palatina/inmunología
15.
Viruses ; 13(3)2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33802390

RESUMEN

BACKGROUND: Strain-specific antibodies to human cytomegalovirus (HCMV) glycoproteins B and H (gB and gH) have been proposed as a potential diagnostic tool for identifying reinfection. We investigated genotype-specific IgG antibody responses in parallel with defining the gB and gH genotypes of the infecting viral strains. METHODS: Subjects with primary (n = 20) or non-primary (n = 25) HCMV infection were studied. The seven gB (gB1-7) and two gH (gH1-2) genotypes were determined by real-time PCR and whole viral genome sequencing, and genotype-specific IgG antibodies were measured by a peptide-based enzyme-linked immunosorbent assay (ELISA). RESULTS: Among subjects with primary infection, 73% (n = 8) infected by gB1-HCMV and 63% (n = 5) infected by gB2/3-HCMV had genotype-specific IgG antibodies to gB (gB2 and gB3 are similar in the region tested). Peptides from the rarer gB4-gB7 genotypes had nonspecific antibody responses. All subjects infected by gH1-HCMV and 86% (n = 6) infected by gH2-HCMV developed genotype-specific responses. Among women with non-primary infection, gB and gH genotype-specific IgG antibodies were detected in 40% (n = 10) and 80% (n = 20) of subjects, respectively. CONCLUSIONS: Peptide-based ELISA is capable of detecting primary genotype-specific IgG responses to HCMV gB and gH, and could be adopted for identifying reinfections. However, about half of the subjects did not have genotype-specific IgG antibodies to gB.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/inmunología , Inmunoglobulina G/sangre , Proteínas del Envoltorio Viral/inmunología , Citomegalovirus/genética , Citomegalovirus/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Genotipo , Humanos , Masculino
16.
Viruses ; 13(10)2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34696402

RESUMEN

Cell-cell fusion is a fundamental and complex process that occurs during reproduction, organ and tissue growth, cancer metastasis, immune response, and infection. All enveloped viruses express one or more proteins that drive the fusion of the viral envelope with cellular membranes. The same proteins can mediate the fusion of the plasma membranes of adjacent cells, leading to the formation of multinucleated syncytia. While cell-cell fusion triggered by alpha- and gammaherpesviruses is well-studied, much less is known about the fusogenic potential of betaherpesviruses such as human cytomegalovirus (HCMV) and human herpesviruses 6 and 7 (HHV-6 and HHV-7). These are slow-growing viruses that are highly prevalent in the human population and associated with several diseases, particularly in individuals with an immature or impaired immune system such as fetuses and transplant recipients. While HHV-6 and HHV-7 are strictly lymphotropic, HCMV infects a very broad range of cell types including epithelial, endothelial, mesenchymal, and myeloid cells. Syncytia have been observed occasionally for all three betaherpesviruses, both during in vitro and in vivo infection. Since cell-cell fusion may allow efficient spread to neighboring cells without exposure to neutralizing antibodies and other host immune factors, viral-induced syncytia may be important for viral dissemination, long-term persistence, and pathogenicity. In this review, we provide an overview of the viral and cellular factors and mechanisms identified so far in the process of cell-cell fusion induced by betaherpesviruses and discuss the possible consequences for cellular dysfunction and pathogenesis.


Asunto(s)
Células Gigantes/fisiología , Infecciones por Herpesviridae/metabolismo , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betaherpesvirinae/metabolismo , Betaherpesvirinae/patogenicidad , Fusión Celular , Citomegalovirus/fisiología , Células Gigantes/virología , Herpesviridae/fisiología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 6/inmunología , Herpesvirus Humano 7/inmunología , Humanos , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
17.
Pediatr Neonatol ; 61(1): 63-67, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31324476

RESUMEN

BACKGROUND: Human cytomegalovirus (HCMV) is a common pathogen that causes many diseases in young children. HCMV glycoprotein H (gH) genotypes may be associated with the type and severity of some diseases. In this study, systematic surveillance was conducted on the prevalence of HCMV infections and HCMV gH genotypes in children. METHODS: Urine samples were collected from children admitted to the inpatient wards and outpatient departments between January 2015 and December 2016 in the Children's Hospital, Zhejiang University School of Medicine (Hangzhou, China), and these were tested by real-time PCR for HCMV DNA detection and HCMV gH genotyping. RESULTS: During the study period, a total of 32,542 urine samples were collected and analyzed using real-time PCR to confirm HCMV infection, and 5286 (16.2%) cases were positive for HCMV DNA. From our results, children aged less than one year were the most susceptible population to HCMV. Based on the data obtained from gH probes combined with real-time PCR assay, 964 HCMV-positive samples were genotyped for gH. Among them, 584 (60.6%) were positive for gH1 subtype, 307 (31.8%) for gH2 subtype, and 73 (7.6%) for both gH1 and gH2 subtypes. The gH2 rate of 42.9% indicated HCMV infection was the highest subtype in the group aged ≤28 days while gH1 rate of 77% was the highest in the group aged >3 years. The highest gH2 rate (36.4%) and lowest gH1 rate (50.0%) were detected in children with HCMV viremia, whereas the highest gH1 rates (65.0%) and lowest gH2 rates (28.8%) were found in children with HCMV hepatitis. CONCLUSION: The findings of our study show that children less than 1-year-old are the major population among HCMV-infected children. gH1 is the predominant genotype of HCMV in China, which occurs more frequently than gH2 genotype in the case of HCMV hepatitis. However, the opposite tendency is observed in HCMV viremia, where the gH2 genotype is predominant.


Asunto(s)
Infecciones por Citomegalovirus/epidemiología , Proteínas del Envoltorio Viral/genética , Adolescente , Niño , Preescolar , China/epidemiología , Citomegalovirus/clasificación , Citomegalovirus/genética , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Prevalencia
18.
Chinese Journal of Biologicals ; (12): 166-171, 2024.
Artículo en Zh | WPRIM | ID: wpr-1011472

RESUMEN

@#Objective To express glycoprotein H(gH)of pseudorabies virus(PRV)in mammalian cells and detect its immunogenicity.Methods The gH gene fragment of PRV-XiangA strain was amplified by PCR and inserted into mammalian cell expression vector pIRES-neo3 to construct recombinant expression plasmid pIRES-gH,which was transfected to HEK-293F cells and cultured in suspension for 5 d. The cell culture supernatant was identified by Western blot and purified by nickel ion chromatography column. The purified gH was emulsified with ISA 201 VG adjuvant to immunize 8 female ICR mice,and 8 mice in control group were immunized with the same amount of adjuvant,which was strengthened at 5 and 8weeks after the first dose respectively. The blood samples were collected at 4,7 and 10 weeks after the first dose and detected for the titer of specific antibody and neutralizing antibody in serum of mice;The mice were challenged with PRVXiangA strain(1. 5 × 104TCID50)by nasal drops 2 d after the third blood collection,and observed for the morbidity and mortality daily.Results The recombinant expression plasmid pIRES-gH was constructed correctly as identified by sequencing. The gH protein was successfully expressed and modified by glycosylation in mammalian cells with good reactivity,and about 625 μg purified protein was obtained under 100 mL culture volume. After three times of immunization,mice produced high level of specific antibody and showed the effect of neutralizing PRV,and the titer of neutralizing antibody reached 1∶256. In the challenge test,all the mice in control group became ill and died,while half of the mice in gH immunized group did not get sick with a survival rate of 50%.Conclusion PRV gH was successfully expressed in mammalian cells,and its immune protection was confirmed for the first time,which provided experimental basis for the further research and application of gH,and also provided a new idea for the development of PRV subunit vaccine.

19.
Viruses ; 11(11)2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31752107

RESUMEN

How viruses enter cells is of critical importance to pathogenesis in the host and for treatment strategies. Over the last several years, the herpesvirus field has made numerous and thoroughly fascinating discoveries about the entry of alpha-, beta-, and gamma-herpesviruses, giving rise to knowledge of entry at the amino acid level and the realization that, in some cases, researchers had overlooked whole sets of molecules essential for entry into critical cell types. Herpesviruses come equipped with multiple envelope glycoproteins which have several roles in many aspects of infection. For herpesvirus entry, it is usual that a collective of glycoproteins is involved in attachment to the cell surface, specific interactions then take place between viral glycoproteins and host cell receptors, and then molecular interactions and triggers occur, ultimately leading to viral envelope fusion with the host cell membrane. The fact that there are multiple cell and virus molecules involved with the build-up to fusion enhances the diversity and specificity of target cell types, the cellular entry pathways the virus commandeers, and the final triggers of fusion. This review will examine discoveries relating to how Kaposi's sarcoma-associated herpesvirus (KSHV) encounters and binds to critical cell types, how cells internalize the virus, and how the fusion may occur between the viral membrane and the host cell membrane. Particular focus is given to viral glycoproteins and what is known about their mechanisms of action.


Asunto(s)
Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Internalización del Virus , Animales , Susceptibilidad a Enfermedades , Células Endoteliales/virología , Humanos , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Tropismo Viral , Replicación Viral
20.
Int J Infect Dis ; 77: 87-89, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30339986

RESUMEN

A 27-year-old pregnant woman who was positive for anti-cytomegalovirus (CMV) antibodies gave birth to a congenitally CMV-infected neonate at 40 weeks of gestation. According to strain-specific serological analysis, which is able to determine the two types of CMV glycoprotein H (gH), the mother possessed anti-gH(To) antibodies only, but the neonate possessed anti-gH(AD) and anti-gH(To) antibodies at 4 weeks after birth. As the anti-gH(To) IgG was decreased in the neonate at 8 months post-delivery, these antibodies are thought to have been transferred from the mother as maternal antibodies. The anti-gH(AD) IgG level was maintained in the child even after 8 months post-delivery. Congenital infection with a CMV gH(AD) type strain was confirmed by strain-specific real-time PCR using a urine specimen from the child. On the other hand, anti-gH(AD) IgG was not detected even after 8 months post-delivery in a maternal specimen. The mother only produced antibodies against the CMV strain identified as the primary infection, which is characteristic of original antigenic sin.


Asunto(s)
Anticuerpos Antivirales/orina , Infecciones por Citomegalovirus/diagnóstico , Transmisión Vertical de Enfermedad Infecciosa , Complicaciones Infecciosas del Embarazo/diagnóstico , Adulto , Citomegalovirus/aislamiento & purificación , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/orina , Femenino , Humanos , Inmunoglobulina G/orina , Lactante , Embarazo , Complicaciones Infecciosas del Embarazo/orina , Reacción en Cadena en Tiempo Real de la Polimerasa , Manejo de Especímenes , Proteínas del Envoltorio Viral/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA