Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.222
Filter
1.
Viruses ; 16(9)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39339924

ABSTRACT

Rabies virus (RABV) is among the first recognized viruses of public health concern and has historically contributed to the development of viral vaccines. Despite these significances, the three-dimensional structure of the RABV virion remains unknown due to the challenges in isolating structurally homogenous virion samples in sufficient quantities needed for structural investigation. Here, by combining the capabilities of cryogenic electron tomography (cryoET) and microscopy (cryoEM), we determined the three-dimensional structure of the wild-type RABV virion. Tomograms of RABV virions reveal a high level of structural heterogeneity among the bullet-shaped virion particles encompassing the glycoprotein (G) trimer-decorated envelope and the nucleocapsid composed of RNA, nucleoprotein (N), and matrix protein (M). The structure of the trunk region of the virion was determined by cryoEM helical reconstruction, revealing a one-start N-RNA helix bound by a single layer of M proteins at an N:M ratio of 1. The N-M interaction differs from that in fellow rhabdovirus vesicular stomatitis virus (VSV), which features two layers of M stabilizing the N-RNA helix at an M:N ratio of 2. These differences in both M-N stoichiometry and binding allow RABV to flex its N-RNA helix more freely and point to different mechanisms of viral assembly between these two bullet-shaped rhabdoviruses.


Subject(s)
Cryoelectron Microscopy , Rabies virus , Virion , Rabies virus/ultrastructure , Rabies virus/chemistry , Virion/ultrastructure , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , Electron Microscope Tomography , Models, Molecular , Nucleocapsid/ultrastructure , Nucleocapsid/metabolism , Nucleocapsid/chemistry , Rabies/virology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/ultrastructure , Viral Matrix Proteins/genetics
2.
PLoS Pathog ; 20(9): e1012525, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39241017

ABSTRACT

Epstein-Barr virus (EBV) uses latency programs to colonize the memory B-cell reservoir, and each program is associated with human malignancies. However, knowledge remains incomplete of epigenetic mechanisms that maintain the highly restricted latency I program, present in memory and Burkitt lymphoma cells, in which EBNA1 is the only EBV-encoded protein expressed. Given increasing appreciation that higher order chromatin architecture is an important determinant of viral and host gene expression, we investigated roles of Wings Apart-Like Protein Homolog (WAPL), a host factor that unloads cohesin to control DNA loop size and that was discovered as an EBNA2-associated protein. WAPL knockout (KO) in Burkitt cells de-repressed LMP1 and LMP2A expression, but not other EBV oncogenes, to yield a viral program reminiscent of EBV latency II, which is rarely observed in B-cells. WAPL KO also increased LMP1/2A levels in latency III lymphoblastoid cells. WAPL KO altered EBV genome architecture, triggering formation of DNA loops between the LMP promoter region and the EBV origins of lytic replication (oriLyt). Hi-C analysis further demonstrated that WAPL KO reprogrammed EBV genomic DNA looping. LMP1 and LMP2A de-repression correlated with decreased histone repressive marks at their promoters. We propose that EBV coopts WAPL to negatively regulate latent membrane protein expression to maintain Burkitt latency I.


Subject(s)
Epstein-Barr Virus Infections , Gene Expression Regulation, Viral , Herpesvirus 4, Human , Viral Matrix Proteins , Virus Latency , Humans , Herpesvirus 4, Human/genetics , Virus Latency/physiology , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/genetics , B-Lymphocytes/virology , B-Lymphocytes/metabolism , Burkitt Lymphoma/virology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Cell Line, Tumor
3.
J Virol ; 98(9): e0086424, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39162567

ABSTRACT

Novel bat H17N10 and H18N11 influenza A viruses (IAVs) are incapable of reassortment with conventional IAVs during co-infection. To date, the underlying mechanisms that inhibit bat and conventional IAV reassortment remain poorly understood. Herein, we used the bat influenza M gene in the PR8 H1N1 virus genetic background to determine the molecular basis that restricts reassortment of segment 7. Our results showed that NEP and M1 from bat H17N10 and H18N11 can interact with PR8 M1 and NEP, resulting in mediating PR8 viral ribonucleoprotein (vRNP) nuclear export and formation of virus-like particles with single vRNP. Further studies demonstrated that the incompatible packaging signals (PSs) of H17N10 or H18N11 M segment led to the failure to rescue recombinant viruses in the PR8 genetic background. Recombinant PR8 viruses (rPR8psH18M and rPR8psH17M) containing bat influenza M coding region flanked with the PR8 M PSs were rescued but displayed lower replication in contrast to the parental PR8 virus, which is due to a low efficiency of recombinant virus uncoating correlating with the functions of the bat M2. Our studies reveal molecular mechanisms of the M gene that hinder reassortment between bat and conventional IAVs, which will help to understand the biology of novel bat IAVs. IMPORTANCE: Reassortment is one of the mechanisms in fast evolution of influenza A viruses (IAVs) and responsible for generating pandemic strains. To date, why novel bat IAVs are incapable of reassorting with conventional IAVs remains completely understood. Here, we attempted to rescue recombinant PR8 viruses with M segment from bat IAVs to understand the molecular mechanisms in hindering their reassortment. Results showed that bat influenza NEP and M1 have similar functions as respective counterparts of PR8 to medicating viral ribonucleoprotein nuclear export. Moreover, the incompatible packaging signals of M genes from bat and conventional IAVs and impaired bat M2 functions are the major reasons to hinder their reassortment. Recombinant PR8 viruses with bat influenza M open reading frames were generated but showed attenuation, which correlated with the functions of the bat M2 protein. Our studies provide novel insights into the molecular mechanisms that restrict reassortment between bat and conventional IAVs.


Subject(s)
Influenza A Virus, H1N1 Subtype , Reassortant Viruses , Humans , Reassortant Viruses/genetics , Animals , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Chiroptera/virology , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Influenza, Human/virology , Influenza, Human/metabolism , HEK293 Cells , Virus Replication , Virus Assembly/genetics , Madin Darby Canine Kidney Cells , Dogs , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics
4.
Virus Genes ; 60(5): 488-500, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39103702

ABSTRACT

Epstein-Barr virus (EBV) infection has a strong correlation with the development of nasopharyngeal carcinoma (NPC). Aquaporin 3 (AQP3), a member of the aquaporin family, plays an important role in tumor development, especially in epithelial-mesenchymal transition. In this study, the expression of AQP3 in EBV-positive NPC cells was significantly lower than that in EBV-negative NPC cells. Western blot and qRT-PCR analysis showed that LMP1 down-regulated the expression of AQP3 by activating the ERK pathway. Cell biology experiments have confirmed that AQP3 affects the development of tumor by promoting cell migration and proliferation in NPC cells. In addition, AQP3 can promote the lysis of EBV in EBV-positive NPC cells. The inhibition of AQP3 expression by EBV through LMP1 may be one of the mechanisms by which EBV maintains latent infection-induced tumor progression.


Subject(s)
Aquaporin 3 , Cell Movement , Down-Regulation , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Viral Matrix Proteins , Humans , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Aquaporin 3/metabolism , Aquaporin 3/genetics , Epstein-Barr Virus Infections/virology , Nasopharyngeal Neoplasms/virology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Cell Line, Tumor , Latent Infection/virology , Cell Proliferation , Carcinoma/virology , Carcinoma/genetics
5.
Viruses ; 16(8)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39205313

ABSTRACT

Feline morbillivirus (FeMV) has been associated with feline health, although its exact role in pathogenesis is still debated. In this study, an indirect enzyme-linked immunosorbent assay (i-ELISA) targeting a recombinant matrix protein of FeMV (rFeMV-M) was developed and assessed in comparison to a Western blotting (WB) assay. The i-ELISA was evaluated using blood samples from 136 cats that were additionally tested with real-time reverse-transcription PCR (RT-qPCR). The i-ELISA exhibited a sensitivity of 90.1%, specificity of 75.6%, positive predictive value of 88.2%, and negative predictive value of 79.1%. The agreement between i-ELISA and WB analyses was substantial (a κ coefficient of 0.664 with a 95% confidence interval of 0.529 to 0.799). Within the study group, 68.4% (93/136) of the cats were serologically positive in the i-ELISA and 66.9% (91/136) in the WB assay, with 11.8% (11/93) of false positivity with the i-ELISA. However, only 8.1% (11/136) of the cats tested positive for FeMV using RT-qPCR (p < 0.001). The developed i-ELISA proved effective in identifying FeMV-infected cats and indicated the prevalence of FeMV exposure. Combining FeMV antibody detection through i-ELISA with FeMV RT-qPCR could offer a comprehensive method to determine and monitor FeMV infection status. Nevertheless, this assay still requires refinement due to a significant number of false positive results, which can lead to the misdiagnosis of cats without antibodies as having antibodies. This study also provided the first evidence of seroprevalence against FeMV among cat populations in Thailand, contributing valuable insights into the geographic distribution and prevalence of this virus.


Subject(s)
Antibodies, Viral , Cat Diseases , Enzyme-Linked Immunosorbent Assay , Morbillivirus Infections , Morbillivirus , Sensitivity and Specificity , Animals , Cats , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/veterinary , Antibodies, Viral/blood , Antibodies, Viral/immunology , Morbillivirus/immunology , Cat Diseases/virology , Cat Diseases/diagnosis , Cat Diseases/immunology , Morbillivirus Infections/veterinary , Morbillivirus Infections/diagnosis , Morbillivirus Infections/immunology , Morbillivirus Infections/virology , Recombinant Proteins/immunology , Female , Blotting, Western/veterinary , Male , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics
6.
Indian J Med Microbiol ; 51: 100699, 2024.
Article in English | MEDLINE | ID: mdl-39111666

ABSTRACT

INTRODUCTION: Nasopharyngeal carcinoma (NPC), arising from nasopharyngeal epithelium is caused by Epstein-Barr virus (EBV). It is common in South China, South East Asia and North East India. The aim and objectives of this study were to determine the prevalence of EBV in formalin-fixed paraffin-embedded (FFPE) tissue sections of clinically suspected NPC patients, correlate the results of polymerase chain reaction (PCR) with histopathology findings, and to determine the utility of tissue EBV DNA as a diagnostic bio-marker. MATERIALS AND METHODS: 31 FFPE tissue samples were collected from clinically suspected NPC patients from April 2018-December 2019. Histopathological diagnosis was done by examination of Hematoxylin and Eosin stained slides. Presence of EBV was detected by EBNA-1 PCR. IHC was performed using EBV Latent Membrane Protein 1. RESULTS: Of the 31 clinically suspected NPC cases, 15 (48.4 %) were histopathological confirmed NPC. Of these15, 13 (86.6 %) were non-keratinising undifferentiated NPC, and one each were keratinising NPC and non-keratinising differentiated NPC respectively. EBV EBNA1 PCR was positive in 35.5 % (11/31) of clinically suspected NPC cases. Of the 11 PCR positive cases, 9 (81.8 %) were histopathological confirmed NPC. Of the 31 clinically suspected NPC cases, IHC was indicated in 23 biopsies. Of which, 12 (52.2 %) were positive for LMP1 in the abnormal cells. Of the 12 IHC positive samples, 10 were NPC cases. CONCLUSION: EBV DNA as an indicator towards NPC among clinically suspected cases had a sensitivity of 60 % and specificity of 87.5 %. In this study, addition of EBV DNA detection by PCR from FFPE tissue sections could confirm EBV association in 20 % of cases where it was not detected by EBV LMP1 IHC, thus helped in increasing the detection of EBV positivity in NPC cases. Early diagnosis of NPC will improve the cure rate and hence reduce the morbidity and mortality rates.


Subject(s)
DNA, Viral , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Tertiary Care Centers , Humans , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Nasopharyngeal Carcinoma/virology , India/epidemiology , Epstein-Barr Virus Infections/epidemiology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , Nasopharyngeal Neoplasms/virology , Male , Female , Middle Aged , DNA, Viral/genetics , Adult , Polymerase Chain Reaction , Epstein-Barr Virus Nuclear Antigens/genetics , Viral Matrix Proteins/genetics , Aged , Young Adult
7.
Front Immunol ; 15: 1439184, 2024.
Article in English | MEDLINE | ID: mdl-39104541

ABSTRACT

Introduction: Human Cytomegalovirus (HCMV) is a betaherpesvirus that causes severe disease in immunocompromised transplant recipients. Immunotherapy with CD8 T cells specific for HCMV antigens presented on HLA class-I molecules is explored as strategy for long-term relief to such patients, but the antiviral effectiveness of T cell preparations cannot be efficiently predicted by available methods. Methods: We developed an Assay for Rapid Measurement of Antiviral T-cell Activity (ARMATA) by real-time automated fluorescent microscopy and used it to study the ability of CD8 T cells to neutralize HCMV and control its spread. As a proof of principle, we used TCR-transgenic T cells specific for the immunodominant HLA-A02-restricted tegumental phosphoprotein pp65. pp65 expression follows an early/late kinetic, but it is not clear at which stage of the virus cycle it acts as an antigen. We measured control of HCMV infection by T cells as early as 6 hours post infection (hpi). Results: The timing of the antigen recognition indicated that it occurred before the late phase of the virus cycle, but also that virion-associated pp65 was not recognized during virus entry into cells. Monitoring of pp65 gene expression dynamics by reporter fluorescent genes revealed that pp65 was detectable as early as 6 hpi, and that a second and much larger bout of expression occurs in the late phase of the virus cycle by 48 hpi. Since transgenic (Tg)-pp65 specific CD8 T cells were activated even when DNA replication was blocked, our data argue that pp65 acts as an early virus gene for immunological purposes. Discussion: ARMATA does not only allow same day identification of antiviral T-cell activity, but also provides a method to define the timing of antigen recognition in the context of HCMV infection.


Subject(s)
CD8-Positive T-Lymphocytes , Cytomegalovirus Infections , Cytomegalovirus , Phosphoproteins , Viral Matrix Proteins , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Cytomegalovirus/genetics , Phosphoproteins/immunology , Phosphoproteins/genetics , Humans , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Gene Expression Regulation, Viral , Antigens, Viral/immunology , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics
8.
Nat Commun ; 15(1): 6802, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122688

ABSTRACT

Influenza virus infection remains a major global health problem and requires a universal vaccine with broad protection against different subtypes as well as a rapid-response vaccine to provide immediate protection in the event of an epidemic outbreak. Here, we show that intranasal administration of probiotic Escherichia coli Nissle 1917 activates innate immunity in the respiratory tract and provides immediate protection against influenza virus infection within 1 day. Based on this vehicle, a recombinant strain is engineered to express and secret five tandem repeats of the extracellular domain of matrix protein 2 from different influenza virus subtypes. Intranasal vaccination with this strain induces durable humoral and mucosal responses in the respiratory tract, and provides broad protection against the lethal challenge of divergent influenza viruses in female BALB/c mice. Our findings highlight a promising delivery platform for developing mucosal vaccines that provide immediate and sustained protection against respiratory pathogens.


Subject(s)
Administration, Intranasal , Escherichia coli , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Probiotics , Animals , Escherichia coli/genetics , Probiotics/administration & dosage , Female , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Mice , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza A virus/immunology , Influenza A virus/genetics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Immunity, Innate , Immunity, Mucosal , Humans , Antibodies, Viral/immunology , Viroporin Proteins
9.
Nat Commun ; 15(1): 6953, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138205

ABSTRACT

Filovirus-host interactions play important roles in all stages of the virus lifecycle. Here, we identify LATS1/2 kinases and YAP, key components of the Hippo pathway, as critical regulators of EBOV transcription and egress. Specifically, we find that when YAP is phosphorylated by LATS1/2, it localizes to the cytoplasm (Hippo "ON") where it sequesters VP40 to prevent egress. In contrast, when the Hippo pathway is "OFF", unphosphorylated YAP translocates to the nucleus where it transcriptionally activates host genes and promotes viral egress. Our data reveal that LATS2 indirectly modulates filoviral VP40-mediated egress through phosphorylation of AMOTp130, a positive regulator of viral egress, but more surprisingly that LATS1/2 kinases directly modulate EBOV transcription by phosphorylating VP30, an essential regulator of viral transcription. In sum, our findings highlight the potential to exploit the Hippo pathway/filovirus axis for the development of host-oriented countermeasures targeting EBOV and related filoviruses.


Subject(s)
Ebolavirus , Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , Transcription, Genetic , Virus Release , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Phosphorylation , Ebolavirus/physiology , Ebolavirus/genetics , Ebolavirus/metabolism , HEK293 Cells , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , YAP-Signaling Proteins/metabolism , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/metabolism , Host-Pathogen Interactions , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics
10.
Emerg Microbes Infect ; 13(1): 2389115, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39129566

ABSTRACT

Rabies is a lethal zoonotic disease that threatens human health. As the only viral surface protein, the rabies virus (RABV) glycoprotein (G) induces main neutralizing antibody (Nab) responses; however, Nab titre is closely correlated with the conformation of G. Virus-like particles (VLP) formed by the co-expression of RABV G and matrix protein (M) improve retention and antigen presentation, inducing broad, durable immune responses. RABV nucleoprotein (N) can elicit humoral and cellular immune responses. Hence, we developed a series of nucleoside-modified RABV mRNA vaccines encoding wild-type G, soluble trimeric RABV G formed by an artificial trimer motif (tG-MTQ), membrane-anchored prefusion-stabilized G (preG). Furthermore, we also developed RABV VLP mRNA vaccine co-expressing preG and M to generate VLPs, and VLP/N mRNA vaccine co-expressing preG, M, and N. The RABV mRNA vaccines induced higher humoral and cellular responses than inactivated rabies vaccine, and completely protected mice against intracerebral challenge. Additionally, the IgG and Nab titres in RABV preG, VLP and VLP/N mRNA groups were significantly higher than those in G and tG-MTQ groups. A single administration of VLP or VLP/N mRNA vaccines elicited protective Nab responses, the Nab titres were significantly higher than that in inactivated rabies vaccine group at day 7. Moreover, RABV VLP and VLP/N mRNA vaccines showed superior capacities to elicit potent germinal centre, long-lived plasma cell and memory B cell responses, which linked to high titre and durable Nab responses. In summary, our data demonstrated that RABV VLP and VLP/N mRNA vaccines could be promising candidates against rabies.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Immunity, Cellular , Immunity, Humoral , Rabies Vaccines , Rabies virus , Rabies , Vaccines, Virus-Like Particle , Animals , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies/prevention & control , Rabies/immunology , Rabies virus/immunology , Rabies virus/genetics , Mice , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Female , mRNA Vaccines/immunology , Mice, Inbred BALB C , Nucleosides/immunology , Glycoproteins/immunology , Glycoproteins/genetics , Humans , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Antigens, Viral/immunology , Antigens, Viral/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology
11.
Virology ; 598: 110194, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39096774

ABSTRACT

RSV infection remains a serious threat to the children all over the world, especially, in the low-middle income countries. Vaccine delivery via the mucosa holds great potential for inducing local immune responses in the respiratory tract. Previously, we reported the development of highly immunogenic RSV virus-like-particles (RSV-VLPs) based on the conformationally stable prefusogenic-F protein (preFg), glycoprotein and matrix protein. Here, to explore whether mucosal delivery of RSV-VLPs is an effective strategy to induce RSV-specific mucosal and systemic immunity, RSV-VLPs were administered via the nasal, sublingual and pulmonary routes to BALB/c mice. The results demonstrate that immunization with the VLPs via the mucosal routes induced minimal mucosal response and yet facilitated modest levels of serum IgG antibodies, enhanced T cell responses and the expression of the lung-homing marker CXCR3 on splenocytes. Immunization with VLPs via all three mucosal routes provided protection against RSV challenge with no signs of RSV induced pathology.


Subject(s)
Antibodies, Viral , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Vaccines, Virus-Like Particle , Viral Fusion Proteins , Viral Matrix Proteins , Animals , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/administration & dosage , Mice , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Viral Fusion Proteins/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/administration & dosage , Female , Viral Matrix Proteins/immunology , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics , Immunity, Mucosal , Immunoglobulin G/blood , Immunoglobulin G/immunology , Respiratory Syncytial Virus, Human/immunology , Lung/virology , Lung/immunology , Glycoproteins/immunology , Glycoproteins/administration & dosage , Administration, Mucosal , Respiratory Syncytial Viruses/immunology , T-Lymphocytes/immunology
12.
Cell Death Dis ; 15(8): 604, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164228

ABSTRACT

Natural killer/T cell lymphoma (NKTCL) exhibits highly aggressive clinical behavior, and the outcomes for relapsed/refractory patients are still poor. Recently, the mechanism underlying the effect of Epstein-Barr virus (EBV) infection, which has not been fully defined in NKTCL, has attracted great attention. We explored how LMP1 promoted aerobic glycolysis via metabolic sequencing combined with mRNA sequencing and immunoprecipitation coupled to mass spectrometry. Experimental assays were used to determine the effects of LMP1 and its downstream pathway on the function and glucose metabolism of NKTCL cells. The correlations between LMP1 expression in patients and their clinical features, treatment response, and prognosis were analyzed. Results show that LMP1 enhances NKTCL cell proliferation in vitro and in vivo, inhibits apoptosis, and decreases gemcitabine sensitivity. In addition, LMP1 also enhances aerobic glycolysis in NKTCL cells, as indicated by increases in glucose uptake, lactate production, and extracellular acidification rate. Clinically, LMP1 expression is correlated with risk stratification, treatment response, and prognosis, and higher LMP1 expression indicates greater SUVmax for NKTCL patients. Mechanistically, LMP1 competitively binds to TRAF3 to promote cell proliferation and aerobic glycolysis by regulating the noncanonical NF-κB pathway. The application of an NF-κB pathway inhibitor or reactivation of the NF-κB pathway affects aerobic glycolysis and the biological function of NKTCL cells. In summary, this study is the first to describe and define in detail how LMP1 affects glucose metabolism in NKTCL and might provide a novel perspective for further treatment.


Subject(s)
Cell Proliferation , Glycolysis , Viral Matrix Proteins , Humans , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Animals , Mice , Cell Line, Tumor , Male , Female , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/genetics , NF-kappa B/metabolism , Herpesvirus 4, Human/metabolism , Middle Aged , Apoptosis , Lymphoma, Extranodal NK-T-Cell/metabolism , Lymphoma, Extranodal NK-T-Cell/pathology , Lymphoma, Extranodal NK-T-Cell/genetics , Signal Transduction
13.
Microbiol Spectr ; 12(9): e0026924, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39078193

ABSTRACT

Filoviruses produce viral particles with characteristic filamentous morphology. The major viral matrix protein, VP40, is trafficked to the plasma membrane and promotes viral particle formation and subsequent viral egress. In the present study, we assessed the role of the small GTPase Rab11-mediated endocytic pathway in Marburg virus (MARV) particle formation and budding. Although Rab11 was predominantly localized in the perinuclear region, it exhibited a more diffuse distribution in the cytoplasm of cells transiently expressing MARV VP40. Rab11 was incorporated into MARV-like particles. Expression of the dominant-negative form of Rab11 and knockdown of Rab11 decreased the amount of VP40 fractions in the cell periphery. Moreover, downregulation of Rab11 moderately reduced the release of MARV-like particles and authentic MARV. We further demonstrated that VP40 induces the distribution of the microtubule network toward the cell periphery, which was partly associated with Rab11. Depolymerization of microtubules reduced the accumulation of VP40 in the cell periphery along with viral particle formation. VP40 physically interacted with α-tubulin, a major component of microtubules, but not with Rab11. Taken together, these results suggested that VP40 partly interacts with microtubules and facilitates their distribution toward the cell periphery, leading to the trafficking of transiently tethering Rab11-positive vesicles toward the cell surface. As we previously demonstrated the role of Rab11 in the formation of Ebola virus particles, the results here suggest that filoviruses in general exploit the vesicle-trafficking machinery for proper virus-particle formation and subsequent egress. These pathways may be a potential target for the development of pan-filovirus therapeutics.IMPORTANCEFiloviruses, including Marburg and Ebola viruses, produce distinct filamentous viral particles. Although it is well known that the major viral matrix protein of these viruses, VP40, is trafficked to the cell surface and promotes viral particle production, details regarding the associated molecular mechanisms remain unclear. To address this knowledge gap, we investigated the role of the small GTPase Rab11-mediated endocytic pathway in this process. Our findings revealed that Marburg virus exploits the Rab11-mediated vesicle-trafficking pathway for the release of virus-like particles and authentic virions in a microtubule network-dependent manner. Previous findings demonstrated that Rab11 is also involved in Ebola virus-particle production. Taken together, these data suggest that filoviruses, in general, may hijack the microtubule-dependent vesicle-trafficking machinery for productive replication. Therefore, this pathway presents as a potential target for the development of pan-filovirus therapeutics.


Subject(s)
Endocytosis , Marburgvirus , Viral Matrix Proteins , Virion , rab GTP-Binding Proteins , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Marburgvirus/physiology , Marburgvirus/genetics , Marburgvirus/metabolism , Humans , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Animals , Virion/metabolism , Microtubules/metabolism , Microtubules/virology , Virus Release , Cell Line , HEK293 Cells , Virus Replication
14.
Cell ; 187(18): 4996-5009.e14, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38996527

ABSTRACT

Latent membrane protein 1 (LMP1) is the primary oncoprotein of Epstein-Barr virus (EBV) and plays versatile roles in the EBV life cycle and pathogenesis. Despite decades of extensive research, the molecular basis for LMP1 folding, assembly, and activation remains unclear. Here, we report cryo-electron microscopy structures of LMP1 in two unexpected assemblies: a symmetric homodimer and a higher-order filamentous oligomer. LMP1 adopts a non-canonical and unpredicted fold that supports the formation of a stable homodimer through tight and antiparallel intermolecular packing. LMP1 dimers further assemble side-by-side into higher-order filamentous oligomers, thereby allowing the accumulation and specific organization of the flexible cytoplasmic tails for efficient recruitment of downstream factors. Super-resolution microscopy and cellular functional assays demonstrate that mutations at both dimeric and oligomeric interfaces disrupt LMP1 higher-order assembly and block multiple LMP1-mediated signaling pathways. Our research provides a framework for understanding the mechanism of LMP1 and for developing potential therapies targeting EBV-associated diseases.


Subject(s)
Herpesvirus 4, Human , Viral Matrix Proteins , Humans , Cryoelectron Microscopy , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , HEK293 Cells , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Models, Molecular , Mutation , Protein Multimerization , Signal Transduction , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
15.
Adv Sci (Weinh) ; 11(36): e2403161, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39049720

ABSTRACT

Nasopharyngeal carcinoma (NPC), a squamous cell carcinoma originating in the nasopharynx, is a leading malignancy in south China and other south and east Asia areas. It is frequently associated with Epstein-Barr virus (EBV) infection, while there are also some NPC patients without EBV infection. Here, it is shown that the EBV+ (EBV positive) and EBV- (EBV negative) NPCs contain both shared and distinct genetic abnormalities, among the latter are increased mutations in TP53. To investigate the functional roles of NPC-associated genetic alterations, primary, orthotopic, and genetically defined NPC models were developed in mice, a key tool missed in the field. These models, initiated with gene-edited organoids of normal nasopharyngeal epithelium, faithfully recapitulated the pathological features of human disease. With these models, it is found that Trp53 and Cdkn2a deficiency are crucial for NPC initiation and progression. And latent membrane protein1 (LMP1), an EBV-coding oncoprotein, significantly promoted the distal metastasis. Further, loss of TGFBR2, which is frequently disrupted both in EBV- and EBV+ NPCs, dramatically accelerated the progression and lung metastasis of NPC probably by altering tumor microenvironment. Taken together, this work establishes a platform to dissect the genetic mechanisms underlying NPC pathogenesis and might be of value for future translational studies.


Subject(s)
Disease Models, Animal , Disease Progression , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Animals , Mice , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Epstein-Barr Virus Infections/genetics , Humans , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Herpesvirus 4, Human/genetics
16.
Phys Chem Chem Phys ; 26(30): 20629-20644, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39037444

ABSTRACT

The M2 proteins of influenza A and B viruses form acid-activated proton channels that are essential for the virus lifecycle. Proton selectivity is achieved by a transmembrane (TM) histidine whereas gating is achieved by a tryptophan residue. Although this functional apparatus is conserved between AM2 and BM2 channels, AM2 conducts protons exclusively inward whereas BM2 conducts protons in either direction depending on the pH gradient. Previous studies showed that in AM2, mutations of D44 abolished inward rectification of AM2, suggesting that the tryptophan gate is destabilized. To elucidate how charged residues C-terminal to the tryptophan regulates channel gating, here we investigate the structure and dynamics of H19 and W23 in a BM2 mutant, GDR-BM2, in which three BM2 residues are mutated to the corresponding AM2 residues, S16G, G26D and H27R. Whole-cell electrophysiological data show that GDR-BM2 conducts protons with inward rectification, identical to wild-type (WT) AM2 but different from WT-BM2. Solid-state NMR 15N and 13C spectra of H19 indicate that the mutant BM2 channel contains higher populations of cationic histidine and neutral τ tautomers compared to WT-BM2 at acidic pH. Moreover, 19F NMR spectra of 5-19F-labeled W23 resolve three peaks at acidic pH, suggesting three tryptophan sidechain conformations. Comparison of these spectra with the tryptophan spectra of other M2 peptides suggests that these indole sidechain conformations arise from interactions with the C-terminal charged residues and with the N-terminal cationic histidine. Taken together, these solid-state NMR data show that inward rectification in M2 proton channels is accomplished by tryptophan interactions with charged residues on both its C-terminal and N-terminal sides. Gating of these M2 proton channels is thus accomplished by a multi-residue complex with finely tuned electrostatic and aromatic interactions.


Subject(s)
Histidine , Influenza B virus , Protons , Tryptophan , Viral Matrix Proteins , Tryptophan/chemistry , Histidine/chemistry , Histidine/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Influenza B virus/chemistry , Influenza B virus/genetics , Influenza A virus/chemistry , Influenza A virus/metabolism , Influenza A virus/genetics , Hydrogen-Ion Concentration , Ion Channels/chemistry , Ion Channels/metabolism , Ion Channels/genetics , Mutation , Molecular Dynamics Simulation , Viroporin Proteins
17.
Viruses ; 16(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39066255

ABSTRACT

A wide range of virus-like particles (VLPs) is extensively employed as carriers to display various antigens for vaccine development to fight against different infections. The plant-produced truncated variant of the hepatitis E virus (HEV) coat protein is capable of forming VLPs. In this study, we demonstrated that recombinant fusion proteins comprising truncated HEV coat protein with green fluorescent protein (GFP) or four tandem copies of the extracellular domain of matrix protein 2 (M2e) of influenza A virus inserted at the Tyr485 position could be efficiently expressed in Nicotiana benthamiana plants using self-replicating vector based on the potato virus X genome. The plant-produced fusion proteins in vivo formed VLPs displaying GFP and 4M2e. Therefore, HEV coat protein can be used as a VLP carrier platform for the presentation of relatively large antigens comprising dozens to hundreds of amino acids. Furthermore, plant-produced HEV particles could be useful research tools for the development of recombinant vaccines against influenza.


Subject(s)
Antigen Presentation , Capsid Proteins , Hepatitis E virus , Nicotiana , Recombinant Fusion Proteins , Viral Matrix Proteins , Hepatitis E virus/immunology , Hepatitis E virus/genetics , Nicotiana/virology , Nicotiana/genetics , Capsid Proteins/genetics , Capsid Proteins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Plants, Genetically Modified , Influenza A virus/immunology , Influenza A virus/genetics , Hepatitis E/immunology , Hepatitis E/prevention & control , Hepatitis E/virology , Viroporin Proteins
18.
Virology ; 597: 110162, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38955082

ABSTRACT

There is an urgent need for influenza vaccines that offer broad cross-protection. The highly conserved ectodomain of the influenza matrix protein 2 (M2e) is a promising candidate; however, its low immunogenicity can be addressed. In this study, we developed influenza vaccines using the Lumazine synthase (LS) platform. The primary objective of this study was to determine the protective potential of M2e proteins expressed on Lumazine synthase (LS) nanoparticles. M2e-LS proteins, produced through the E. coli system, spontaneously assemble into nanoparticles. The study investigated the efficacy of the M2e-LS nanoparticle vaccine in mice. Mice immunized with M2e-LS nanoparticles exhibited significantly higher levels of intracellular cytokines than those receiving soluble M2e proteins. The M2e-LS protein exhibited robust immunogenicity and provided 100% protection against cross-clade influenza.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Multienzyme Complexes , Nanoparticles , Orthomyxoviridae Infections , Viral Matrix Proteins , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Nanoparticles/chemistry , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Female , Mice, Inbred BALB C , Antibodies, Viral/immunology , Cytokines/metabolism , Cross Protection/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Escherichia coli/genetics , Escherichia coli/metabolism , Viroporin Proteins
19.
Vopr Virusol ; 69(2): 175-186, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38843023

ABSTRACT

INTRODUCTION: The COVID-19 pandemic caused by SARS-CoV-2 has created serious health problems worldwide. The most effective way to prevent the occurrence of new epidemic outbreaks is vaccination. One of the modern and effective approaches to vaccine development is the use of virus-like particles (VLPs). The aim of the study is to develop a technology for production of VLP based on recombinant SARS-CoV-2 proteins (E, M, N and S) in insect cells. MATERIALS AND METHODS: Synthetic genes encoding coronavirus proteins E, M, N and S were used. VLP with various surface proteins of strains similar to the Wuhan virus, Delta, Alpha and Omicron were developed and cloned into the pFastBac plasmid. The proteins were synthesized in the baculovirus expression system and assembled into VLP in the portable Trichoplusia ni cell. The presence of insertion in the baculovirus genome was determined by PCR. ELISA and immunoblotting were used to study the antigenic activity of VLP. VLP purification was performed by ultracentrifugation using 20% sucrose. Morphology was assessed using electron microscopy and dynamic light scattering. RESULTS: VLPs consisting of recombinant SARS-CoV-2 proteins (S, M, E and N) were obtained and characterized. The specific binding of antigenic determinants in synthesized VLPs with antibodies to SARS-CoV-2 proteins has been demonstrated. The immunogenic properties of VLPs have been studied. CONCLUSION: The production and purification of recombinant VLPs consisting of full-length SARS-CoV-2 proteins with a universal set of surface antigens have been developed and optimized. Self-assembling particles that mimic the coronavirus virion induce a specific immune response against SARS-CoV-2.


Subject(s)
Baculoviridae , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccines, Virus-Like Particle , Animals , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Humans , COVID-19/virology , COVID-19/immunology , Baculoviridae/genetics , Baculoviridae/metabolism , COVID-19 Vaccines/immunology , Antibodies, Viral/immunology , Coronavirus M Proteins/genetics , Coronavirus M Proteins/immunology , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Phosphoproteins
20.
Molecules ; 29(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38930866

ABSTRACT

The H5N1 avian influenza virus seriously affects the health of poultry and humans. Once infected, the mortality rate is very high. Therefore, accurate and timely detection of the H5N1 avian influenza virus is beneficial for controlling its spread. This article establishes a dual gene detection method based on dual RPA for simultaneously detecting the HA and M2 genes of H5N1 avian influenza virus, for the detection of H5N1 avian influenza virus. Design specific primers for the conserved regions of the HA and M2 genes. The sensitivity of the dual RT-RPA detection method for HA and M2 genes is 1 × 10-7 ng/µL. The optimal primer ratio is 1:1, the optimal reaction temperature is 40 °C, and the optimal reaction time is 20 min. Dual RT-RPA was used to detect 72 samples, and compared with RT-qPCR detection, the Kappa value was 1 (p value < 0.05), and the clinical sample detection sensitivity and specificity were both 100%. The dual RT-RPA method is used for the first time to simultaneously detect two genes of the H5N1 avian influenza virus. As an accurate and convenient diagnostic tool, it can be used to diagnose the H5N1 avian influenza virus.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza in Birds , Influenza A Virus, H5N1 Subtype/genetics , Animals , Influenza in Birds/virology , Influenza in Birds/diagnosis , Humans , Sensitivity and Specificity , Influenza, Human/virology , Influenza, Human/diagnosis , Viral Matrix Proteins/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Birds/virology , Viroporin Proteins
SELECTION OF CITATIONS
SEARCH DETAIL