Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
PLoS One ; 18(7): e0289100, 2023.
Article in English | MEDLINE | ID: mdl-37490507

ABSTRACT

The M2-2 protein from the respiratory syncytial virus (RSV) is a 10 kDa protein expressed by the second ORF of the viral gene M2. During infection, M2-2 has been described as the polymerase cofactor responsible for promoting genome replication, which occurs by the induction of changes in interactions between the polymerase and other viral proteins at early stages of infection. Despite its well-explored role in the regulation of the polymerase activity, little has been made to investigate the relationship of M2-2 with cellular proteins. A previous report showed poor recruitment of M2-2 to viral structures, with the protein being mainly localized to the nucleus and cytoplasmic granules. To unravel which other functions M2-2 exerts during infection, we performed proteomic analysis of co-immunoprecipitated cellular partners, identifying enrichment of proteins involved with regulation of translation, protein folding and mRNA splicing. In approaches based on these data, we found that M2-2 expression downregulates eiF2α phosphorylation and inhibits both translation and stress granules assembly. Finally, we also verified that M2-2 is targeted for proteasome degradation, being localized to granules composed of defective ribosomal products at the cytoplasm. These results suggest that besides its functions in the replicative complex, M2-2 may exert additional functions to contribute to successful RSV infection.


Subject(s)
Proteasome Endopeptidase Complex , Respiratory Syncytial Virus, Human , Proteomics , Stress Granules , Viral Proteins/genetics , Respiratory Syncytial Virus, Human/genetics , Virus Replication/physiology
2.
Front Cell Infect Microbiol ; 12: 849017, 2022.
Article in English | MEDLINE | ID: mdl-35677658

ABSTRACT

SARS-CoV-2 is an emerging virus from the Coronaviridae family and is responsible for the ongoing COVID-19 pandemic. In this work, we explored the previously reported SARS-CoV-2 structural membrane protein (M) interaction with human Proliferating Cell Nuclear Antigen (PCNA). The M protein is responsible for maintaining virion shape, and PCNA is a marker of DNA damage which is essential for DNA replication and repair. We validated the M-PCNA interaction through immunoprecipitation, immunofluorescence co-localization, and PLA (Proximity Ligation Assay). In cells infected with SARS-CoV-2 or transfected with M protein, using immunofluorescence and cell fractioning, we documented a reallocation of PCNA from the nucleus to the cytoplasm and the increase of PCNA and γH2AX (another DNA damage marker) expression. We also observed an increase in PCNA and γH2AX expression in the lung of a COVID-19 patient by immunohistochemistry. In addition, the inhibition of PCNA translocation by PCNA I1 and Verdinexor led to a reduction of plaque formation in an in vitro assay. We, therefore, propose that the transport of PCNA to the cytoplasm and its association with M could be a virus strategy to manipulate cell functions and may be considered a target for COVID-19 therapy.


Subject(s)
COVID-19 Drug Treatment , Coronavirus M Proteins , Proliferating Cell Nuclear Antigen , Coronavirus M Proteins/metabolism , Humans , Proliferating Cell Nuclear Antigen/metabolism , SARS-CoV-2
3.
J Virol ; 95(22): e0091221, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34431698

ABSTRACT

Respiratory syncytial virus (RSV) is the main cause of acute respiratory infections in young children and also has a major impact on the elderly and immunocompromised people. In the absence of a vaccine or efficient treatment, a better understanding of RSV interactions with the host antiviral response during infection is needed. Previous studies revealed that cytoplasmic inclusion bodies (IBs), where viral replication and transcription occur, could play a major role in the control of innate immunity during infection by recruiting cellular proteins involved in the host antiviral response. We recently showed that the morphogenesis of IBs relies on a liquid-liquid-phase separation mechanism depending on the interaction between viral nucleoprotein (N) and phosphoprotein (P). These scaffold proteins are expected to play a central role in the recruitment of cellular proteins to IBs. Here, we performed a yeast two-hybrid screen using RSV N protein as bait and identified the cellular protein TAX1BP1 as a potential partner of this viral protein. This interaction was validated by pulldown and immunoprecipitation assays. We showed that TAX1BP1 suppression has only a limited impact on RSV infection in cell cultures. However, RSV replication is decreased in TAX1BP1-deficient (TAX1BP1 knockout [TAX1BP1KO]) mice, whereas the production of inflammatory and antiviral cytokines is enhanced. In vitro infection of wild-type or TAX1BP1KO alveolar macrophages confirmed that the innate immune response to RSV infection is enhanced in the absence of TAX1BP1. Altogether, our results suggest that RSV could hijack TAX1BP1 to restrain the host immune response during infection. IMPORTANCE Respiratory syncytial virus (RSV), which is the leading cause of lower respiratory tract illness in infants, remains a medical problem in the absence of a vaccine or efficient treatment. This virus is also recognized as a main pathogen in the elderly and immunocompromised people, and the occurrence of coinfections (with other respiratory viruses and bacteria) amplifies the risks of developing respiratory distress. In this context, a better understanding of the pathogenesis associated with viral respiratory infections, which depends on both viral replication and the host immune response, is needed. The present study reveals that the cellular protein TAX1BP1, which interacts with the RSV nucleoprotein N, participates in the control of the innate immune response during RSV infection, suggesting that the N-TAX1BP1 interaction represents a new target for the development of antivirals.


Subject(s)
Intracellular Signaling Peptides and Proteins/immunology , Neoplasm Proteins/immunology , Nucleocapsid Proteins/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Cell Line , Cricetinae , Humans , Immunity, Innate , Mice , Mice, Knockout , Virus Replication
4.
Viruses ; 13(2)2021 02 02.
Article in English | MEDLINE | ID: mdl-33540662

ABSTRACT

Human respiratory syncytial virus (HRSV) is the most frequent cause of severe respiratory disease in children. The main targets of HRSV infection are epithelial cells of the respiratory tract, and the great majority of the studies regarding HRSV infection are done in respiratory cells. Recently, the interest on respiratory virus infection of lymphoid cells has been growing, but details of the interaction of HRSV with lymphoid cells remain unknown. Therefore, this study was done to assess the relationship of HRSV with A3.01 cells, a human CD4+ T cell line. Using flow cytometry and fluorescent focus assay, we found that A3.01 cells are susceptible but virtually not permissive to HRSV infection. Dequenching experiments revealed that the fusion process of HRSV in A3.01 cells was nearly abolished in comparison to HEp-2 cells, an epithelial cell lineage. Quantification of viral RNA by RT-qPCR showed that the replication of HRSV in A3.01 cells was considerably reduced. Western blot and quantitative flow cytometry analyses demonstrated that the production of HRSV proteins in A3.01 was significantly lower than in HEp-2 cells. Additionally, using fluorescence in situ hybridization, we found that the inclusion body-associated granules (IBAGs) were almost absent in HRSV inclusion bodies in A3.01 cells. We also assessed the intracellular trafficking of HRSV proteins and found that HRSV proteins colocalized partially with the secretory pathway in A3.01 cells, but these HRSV proteins and viral filaments were present only scarcely at the plasma membrane. HRSV infection of A3.01 CD4+ T cells is virtually unproductive as compared to HEp-2 cells, as a result of defects at several steps of the viral cycle: Fusion, genome replication, formation of inclusion bodies, recruitment of cellular proteins, virus assembly, and budding.


Subject(s)
Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology , T-Lymphocytes/virology , Cell Line , Humans , Respiratory Syncytial Virus, Human/genetics , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Assembly , Virus Replication
5.
Genet Mol Biol ; 44(1 Suppl 1): e20200212, 2020.
Article in English | MEDLINE | ID: mdl-33237152

ABSTRACT

SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), as well as SARS-CoV from 2003 along with MERS-CoV from 2012, is a member of the Betacoronavirus genus of the Nidovirales order and is currently the cause of the pandemic called COVID-19 (or Coronavirus disease 2019). COVID-19, which is characterized by cough, fever, fatigue, and severe cases of pneumonia, has affected more than 23 million people worldwide until August 25th, 2020. Here, we present a review of the cellular mechanisms associated with human coronavirus replication, including the unique molecular events related to the replication transcription complex (RTC) of coronaviruses. We also present information regarding the interactions between each viral protein and cellular proteins associated to known host-pathogen implications for the coronavirus biology. Finally, a specific topic addresses the current attempts for pharmacological interventions against COVID-19, highlighting the possible effects of each drug on the molecular events of viral replication. This review intends to aid future studies for a better understanding of the SARS-CoV-2 replication cycle and the development of pharmacological approaches targeting COVID-19.

6.
mBio ; 11(5)2020 09 29.
Article in English | MEDLINE | ID: mdl-32994321

ABSTRACT

Human respiratory syncytial virus (HRSV) envelope glycoproteins traffic to assembly sites through the secretory pathway, while nonglycosylated proteins M and N are present in HRSV inclusion bodies but must reach the plasma membrane, where HRSV assembly happens. Little is known about how nonglycosylated HRSV proteins reach assembly sites. Here, we show that HRSV M and N proteins partially colocalize with the Golgi marker giantin, and the glycosylated F and nonglycosylated N proteins are closely located in the trans-Golgi, suggesting their interaction in that compartment. Brefeldin A compromised the trafficking of HRSV F and N proteins and inclusion body sizes, indicating that the Golgi is important for both glycosylated and nonglycosylated HRSV protein traffic. HRSV N and M proteins colocalized and interacted with sorting nexin 2 (SNX2), a retromer component that shapes endosomes in tubular structures. Glycosylated F and nonglycosylated N HRSV proteins are detected in SNX2-laden aggregates with intracellular filaments projecting from their outer surfaces, and VPS26, another retromer component, was also found in inclusion bodies and filament-shaped structures. Similar to SNX2, TGN46 also colocalized with HRSV M and N proteins in filamentous structures at the plasma membrane. Cell fractionation showed enrichment of SNX2 in fractions containing HRSV M and N proteins. Silencing of SNX1 and 2 was associated with reduction in viral proteins, HRSV inclusion body size, syncytium formation, and progeny production. The results indicate that HRSV structural proteins M and N are in the secretory pathway, and SNX2 plays an important role in the traffic of HRSV structural proteins toward assembly sites.IMPORTANCE The present study contributes new knowledge to understand HRSV assembly by providing evidence that nonglycosylated structural proteins M and N interact with elements of the secretory pathway, shedding light on their intracellular traffic. To the best of our knowledge, the present contribution is important given the scarcity of studies about the traffic of HRSV nonglycosylated proteins, especially by pointing to the involvement of SNX2, a retromer component, in the HRSV assembly process.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Host Microbial Interactions , Nucleocapsid Proteins/metabolism , Respiratory Syncytial Virus, Human/physiology , Viral Proteins/metabolism , Virus Assembly , Amyloid beta-Protein Precursor/genetics , Carrier Proteins , Golgi Apparatus/metabolism , Golgi Matrix Proteins/metabolism , HeLa Cells , Humans , Protein Transport
7.
Preprint in English | SciELO Preprints | ID: pps-1045

ABSTRACT

SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), as well as SARS-CoV from 2003 and MERS-CoV from 2012, is a member of the Betacoronavirus genus of the Nidovirales order and is currently the cause of the pandemic called COVID-19 (or Coronavirus disease 2019). COVID-19 is characterized by cough, fever, fatigue, and severe cases of pneumonia and has affected more than 9.1 million people worldwide until June 22nd, 2020. Here we present a review of the cellular mechanisms associated with human coronaviruses replication, including the unique molecular events related to the replication transcription complex (RTC) of coronaviruses. Information regarding the interactions between each viral protein and cellular proteins is presented, associating to known host-pathogen implications for the coronavirus biology. Finally, a specific topic addresses the current attempts for pharmacological interventions against COVID-19, highlighting the possible effects of each drug on the molecular events of viral replication. This review intends to contribute to future studies for a better understanding of the SARS-CoV-2 replication cycle and the development of pharmacological approaches targeting COVID-19.

8.
J Virol ; 89(2): 917-30, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25355874

ABSTRACT

UNLABELLED: The human respiratory syncytial virus (HRSV) core viral RNA polymerase comprises the large polymerase protein (L) and its cofactor, the phosphoprotein (P), which associate with the viral ribonucleoprotein complex to replicate the genome and, together with the M2-1 protein, transcribe viral mRNAs. While cellular proteins have long been proposed to be involved in the synthesis of HRSV RNA by associating with the polymerase complex, their characterization has been hindered by the difficulty of purifying the viral polymerase from mammalian cell culture. In this study, enhanced green fluorescent protein (EGFP)-tagged L- and P-protein expression was coupled with high-affinity anti-GFP antibody-based immunoprecipitation and quantitative proteomics to identify cellular proteins that interacted with either the L- or the P-proteins when expressed as part of a biologically active viral RNP. Several core groups of cellular proteins were identified that interacted with each viral protein including, in both cases, protein chaperones. Ablation of chaperone activity by using small-molecule inhibitors confirmed previously reported studies which suggested that this class of proteins acted as positive viral factors. Inhibition of HSP90 chaperone function in the current study showed that HSP90 is critical for L-protein function and stability, whether in the presence or absence of the P-protein. Inhibition studies suggested that HSP70 also disrupts virus biology and might help the polymerase remodel the nucleocapsid to allow RNA synthesis to occur efficiently. This indicated a proviral role for protein chaperones in HRSV replication and demonstrates that the function of cellular proteins can be targeted as potential therapeutics to disrupt virus replication. IMPORTANCE: Human respiratory syncytial virus (HRSV) represents a major health care and economic burden, being the main cause of severe respiratory infections in infants worldwide. No vaccine or effective therapy is available. This study focused on identifying those cellular proteins that potentially interact specifically with the viral proteins that are central to virus replication and transcription, with a view to providing potential targets for the development of a specific, transient therapeutic which disrupts virus biology but prevents the emergence of resistance, while maintaining cell viability. In particular, protein chaperones (heat shock proteins 70 and 90), which aid protein folding and function, were identified. The mechanism by which these chaperones contribute to virus biology was tested, and this study demonstrates to the field that cellular protein chaperones may be required for maintaining the correct folding and therefore functionality of specific proteins within the virus replication complex.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Host-Pathogen Interactions , Molecular Chaperones/metabolism , Protein Interaction Maps , Respiratory Syncytial Virus, Human/physiology , Viral Proteins/metabolism , Virus Replication , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Protein Binding , Protein Interaction Mapping , Protein Stability
9.
Intervirology ; 57(2): 55-64, 2014.
Article in English | MEDLINE | ID: mdl-24480928

ABSTRACT

OBJECTIVES: Since it has been reported that in humans there is a relationship between human respiratory syncytial virus (hRSV)-specific cytotoxic T lymphocytes and symptom reduction, and that the polymerase (structural L protein) is highly conserved among different strains, this work aimed to identify the CD8 T cell epitopes H-2(d) restricted within the L sequence for immunization purposes. METHODS: We screened the hRSV strain A2 L protein sequence using two independent algorithms, SYFPEITHI and PRED/(BALB/c), to predict CD8 T cell epitopes. The selected peptides were synthesized and used to immunize BALB/c mice for the evaluation of T cell response. The production of IFN-γ from splenocytes of hRSV-infected animals stimulated by these peptides was assayed by ELISPOT. RESULTS: Nine peptides showing the best binding scores to the BALB/c MHC-I molecules (H-2K(d), L(d) and D(d)) were selected. Sequence homology analysis showed that these sequences are conserved among different hRSV strains. Two of these peptides induced significant IFN-γ production by ex vivo-stimulated T cells. CONCLUSIONS: Our results indicate that the hRSV L protein contains H-2(d)-restricted epitopes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA-Directed RNA Polymerases/immunology , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Structural Proteins/immunology , Animals , Enzyme-Linked Immunospot Assay , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mice, Inbred BALB C
10.
Front Immunol ; 4: 487, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24432018

ABSTRACT

Native type I heat-labile toxins (LTs) produced by enterotoxigenic Escherichia coli (ETEC) strains exert strong adjuvant effects on both antibody and T cell responses to soluble and particulate antigens following co-administration via mucosal routes. However, inherent enterotoxicity and neurotoxicity (following intra-nasal delivery) had reduced the interest in the use of these toxins as mucosal adjuvants. LTs can also behave as powerful and safe adjuvants following delivery via parenteral routes, particularly for activation of cytotoxic lymphocytes. In the present study, we evaluated the adjuvant effects of a new natural LT polymorphic form (LT2), after delivery via intradermal (i.d.) and subcutaneous (s.c.) routes, with regard to both antibody and T cell responses. A recombinant HIV-1 p24 protein was employed as a model antigen for determination of antigen-specific immune responses while the reference LT (LT1), produced by the ETEC H10407 strain, and a non-toxigenic LT form (LTK63) were employed as previously characterized LT types. LT-treated mice submitted to a four dose-base immunization regimen elicited similar p24-specific serum IgG responses and CD4(+) T cell activation. Nonetheless, mice immunized with LT1 or LT2 induced higher numbers of antigen-specific CD8(+) T cells and in vivo cytotoxic responses compared to mice immunized with the non-toxic LT derivative. These effects were correlated with stronger activation of local dendritic cell populations. In addition, mice immunized with LT1 and LT2, but not with LTK63, via s.c. or i.d. routes developed local inflammatory reactions. Altogether, the present results confirmed that the two most prevalent natural polymorphic LT variants (LT1 or LT2) display similar and strong adjuvant effects for subunit vaccines administered via i.d. or s.c. routes.

11.
PLoS One ; 8(8): e71322, 2013.
Article in English | MEDLINE | ID: mdl-23951135

ABSTRACT

Millions of people worldwide are currently infected with human papillomavirus (HPV), herpes simplex virus (HSV) or human immunodeficiency virus (HIV). For this enormous contingent of people, the search for preventive and therapeutic immunological approaches represents a hope for the eradication of latent infection and/or virus-associated cancer. To date, attempts to develop vaccines against these viruses have been mainly based on a monovalent concept, in which one or more antigens of a virus are incorporated into a vaccine formulation. In the present report, we designed and tested an immunization strategy based on DNA vaccines that simultaneously encode antigens for HIV, HSV and HPV. With this purpose in mind, we tested two bicistronic DNA vaccines (pIRES I and pIRES II) that encode the HPV-16 oncoprotein E7 and the HIV protein p24 both genetically fused to the HSV-1 gD envelope protein. Mice i.m. immunized with the DNA vaccines mounted antigen-specific CD8⁺ T cell responses, including in vivo cytotoxic responses, against the three antigens. Under experimental conditions, the vaccines conferred protective immunity against challenges with a vaccinia virus expressing the HIV-derived protein Gag, an HSV-1 virus strain and implantation of tumor cells expressing the HPV-16 oncoproteins. Altogether, our results show that the concept of a trivalent HIV, HSV, and HPV vaccine capable to induce CD8⁺ T cell-dependent responses is feasible and may aid in the development of preventive and/or therapeutic approaches for the control of diseases associated with these viruses.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/prevention & control , Herpes Simplex/prevention & control , Papillomavirus Infections/prevention & control , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , AIDS Vaccines/immunology , AIDS Vaccines/therapeutic use , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Animals , Antigens, Viral/genetics , Female , HIV/genetics , HIV/immunology , HIV Infections/immunology , Herpes Simplex/immunology , Herpes Simplex Virus Vaccines/immunology , Herpes Simplex Virus Vaccines/therapeutic use , Humans , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Papillomavirus Infections/immunology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/therapeutic use , Simplexvirus/genetics , Simplexvirus/immunology , Vaccines, DNA/genetics
12.
Virus Res ; 177(1): 108-12, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23892143

ABSTRACT

Characterization of Human Respiratory Syncytial Virus (HRSV) protein interactions with host cell components is crucial to devise antiviral strategies. Viral nucleoprotein, phosphoprotein and matrix protein genes were optimized for human codon usage and cloned into expression vectors. HEK-293T cells were transfected with these vectors, viral proteins were immunoprecipitated, and co-immunoprecipitated cellular proteins were identified through mass spectrometry. Cell proteins identified with higher confidence scores were probed in the immunoprecipitation using specific antibodies. The results indicate that nucleoprotein interacts with arginine methyl-transferase, methylosome protein and Hsp70. Phosphoprotein interacts with Hsp70 and tropomysin, and matrix with tropomysin and nucleophosmin. Additionally, we performed immunoprecipitation of these cellular proteins in cells infected with HRSV, followed by detection of co-immunoprecipitated viral proteins. The results indicate that these interactions also occur in the context of viral infection, and their potential contribution for a HRSV replication model is discussed.


Subject(s)
Nucleoproteins/metabolism , Phosphoproteins/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/metabolism , Viral Matrix Proteins/metabolism , HEK293 Cells , HSP72 Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Nucleoproteins/genetics , Phosphoproteins/genetics , Protein Binding , Protein-Arginine N-Methyltransferases/metabolism , Respiratory Syncytial Virus, Human/genetics , Viral Matrix Proteins/genetics , Viral Structural Proteins
13.
Braz. j. microbiol ; 42(1): 340-345, Jan.-Mar. 2011. ilus
Article in English | LILACS | ID: lil-571408

ABSTRACT

Human Respiratory Syncytial Virus P protein plus the viral RNA, N and L viral proteins, constitute the viral replication complex. In this report we describe that HRSV P protein has putative intrinsically disordered domains predicted by in silico methods. These two domains, located at the amino and caboxi terminus, were identified by mass spectrometry analysis of peptides obtained from degradation fragments observed in purified P protein expressed in bacteria. The degradation is not occurring at the central oligomerization domain, since we also demonstrate that the purified fragments are able to oligomerize, similarly to the protein expressed in cells infected by HRSV. Disordered domains can play a role in protein interaction, and the present data contribute to the comprehension of HRSV P protein interactions in the viral replication complex.


Subject(s)
Humans , Chromatography, Liquid/methods , Mass Spectrometry/methods , Peptide Fragments/analysis , In Vitro Techniques , RNA, Viral , Virus Replication , Respiratory Syncytial Virus, Human/isolation & purification , Diagnostic Techniques and Procedures , Methods
14.
Braz J Microbiol ; 42(1): 340-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-24031640

ABSTRACT

Human Respiratory Syncytial Virus P protein plus the viral RNA, N and L viral proteins, constitute the viral replication complex. In this report we describe that HRSV P protein has putative intrinsically disordered domains predicted by in silico methods. These two domains, located at the amino and caboxi terminus, were identified by mass spectrometry analysis of peptides obtained from degradation fragments observed in purified P protein expressed in bacteria. The degradation is not occurring at the central oligomerization domain, since we also demonstrate that the purified fragments are able to oligomerize, similarly to the protein expressed in cells infected by HRSV. Disordered domains can play a role in protein interaction, and the present data contribute to the comprehension of HRSV P protein interactions in the viral replication complex.

15.
J Virol Methods ; 158(1-2): 93-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19428575

ABSTRACT

Human respiratory syncytial virus (HRSV) is the major pathogen leading to respiratory disease in infants and neonates worldwide. An effective vaccine has not yet been developed against this virus, despite considerable efforts in basic and clinical research. HRSV replication is independent of the nuclear RNA processing constraints, since the virus genes are adapted to the cytoplasmic transcription, a process performed by the viral RNA-dependent RNA polymerase. This study shows that meaningful nuclear RNA polymerase II dependent expression of the HRSV nucleoprotein (N) and phosphoprotein (P) proteins can only be achieved with the optimization of their genes, and that the intracellular localization of N and P proteins changes when they are expressed out of the virus replication context. Immunization tests performed in mice resulted in the induction of humoral immunity using the optimized genes. This result was not observed for the non-optimized genes. In conclusion, optimization is a valuable tool for improving expression of HRSV genes in DNA vaccines.


Subject(s)
Antibodies, Viral/blood , Nucleoproteins/immunology , Phosphoproteins/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Proteins/immunology , Animals , Female , Humans , Mice , Mice, Inbred BALB C , Nucleoproteins/biosynthesis , Nucleoproteins/genetics , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Respiratory Syncytial Virus Vaccines/biosynthesis , Respiratory Syncytial Virus Vaccines/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics
16.
Intervirology ; 51(3): 189-95, 2008.
Article in English | MEDLINE | ID: mdl-18724066

ABSTRACT

OBJECTIVES: To construct a recombinant baculovirus expressing the fiber knob domain of human adenovirus type 2 modified by the insertion of a foreign peptide, purify this protein after its production in insect cells, and to test its properties. METHODS: Recombinant baculoviruses expressing the fiber knob were produced in Sf9 cells. The recombinant fiber knob was recovered from culture supernatants of infected cells and purified by a combination of Ni-NTA and ion-exchange chromatography. RESULTS: Fiber knob was recovered from the culture media as a soluble protein. In the system used, the fiber knob is expressed fused with the V5 epitope and a histidine tag, which allowed purification by Ni-NTA chromatography. The protein was further purified by ion-exchange chromatography. We show that the recombinant fiber knob produced, with 31 extra amino acids in the C-terminus, can oligomerize and bind to the adenovirus receptor CAR, as it can block the infection of a recombinant type 5 adenovirus. CONCLUSIONS: The modified form of the fiber knob, produced in insect cells and purified by Ni-NTA and ion-exchange chromatography, retains the properties of oligomerization and binding to the fiber natural receptor, CAR. This construct has the potential to be a new adjuvant.


Subject(s)
Adenoviruses, Human/genetics , Capsid Proteins/biosynthesis , Animals , Baculoviridae/genetics , Capsid Proteins/genetics , Capsid Proteins/isolation & purification , Cell Line , Chromatography, Affinity , Chromatography, Ion Exchange , Genetic Vectors , Protein Binding , Receptors, Virus/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recoverin/metabolism , Spodoptera
17.
Protein Expr Purif ; 53(1): 209-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17292625

ABSTRACT

The nucleoprotein (N) and the phosphoprotein (P) of the human respiratory syncytial virus (HRSV), A2 strain, were cloned into pMAL-c2e vector. The proteins were expressed fused with the maltose-binding protein (MBP) and were preferentially found in the soluble fraction of the bacterial lysate. After their purification using amylose resin, almost no other protein was detected in SDS-PAGE. The fused proteins were cleaved by digestion with enterokinase and then used as antigens for BALB/c mice immunization. The obtained polyclonal antibodies were tested against HRSV infected cells in immunofluorescence assays. The results indicate that the antibodies generated against the recombinant proteins were able to recognize the virus proteins. We now intend to purify the cleaved N and P proteins and use them in structural studies. The recombinant proteins will also be tested as potential inducers of a protective immunity against the HRSV.


Subject(s)
Antibodies, Viral/biosynthesis , Nucleoproteins/metabolism , Phosphoproteins/metabolism , Respiratory Syncytial Virus, Human/chemistry , Respiratory Syncytial Virus, Human/metabolism , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Blotting, Western , Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Cell Line, Tumor , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Enteropeptidase/pharmacology , Escherichia coli/genetics , Female , Fluorescent Antibody Technique, Direct , Genetic Vectors , Humans , Liver Neoplasms/pathology , Maltose-Binding Proteins , Mice , Mice, Inbred BALB C , Nucleoproteins/genetics , Nucleoproteins/isolation & purification , Phosphoproteins/genetics , Phosphoproteins/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/genetics , Reverse Transcriptase Polymerase Chain Reaction , Solubility , Transformation, Genetic , Vaccination
18.
Rev Soc Bras Med Trop ; 39(2): 159-62, 2006.
Article in English | MEDLINE | ID: mdl-16699642

ABSTRACT

A digoxigenin-labeled probe was produced from the Pasteur virus strain for the detection of the rabies virus N gene. The probe hybridization was performed from amplified N gene obtained by reverse transcription polymerase chain reaction and the results by RT-PCR and hybridization showed 100% agreement. The hybridization, when carried out in products amplified by RT-PCR, increases the sensitivity of this technique even more and confers specificity to the diagnosis. The technique described in this work will be useful in rabies diagnosis laboratories, once the cost is compatible with traditional rabies diagnostic techniques.


Subject(s)
Digoxigenin , Genes, Viral/genetics , Nucleoproteins/genetics , Rabies virus/genetics , Rabies/diagnosis , Animals , Blotting, Southern , Cattle , Chiroptera , DNA Probes , DNA, Viral/genetics , Dogs , Horses , Humans , In Situ Hybridization , Luminescent Measurements , Rabies/virology , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Sheep
19.
Rev. Soc. Bras. Med. Trop ; 39(2): 159-162, mar.-abr. 2006. ilus, tab
Article in English | LILACS, Sec. Est. Saúde SP | ID: lil-426908

ABSTRACT

A digoxigenin-labeled probe was produced from the Pasteur virus strain for the detection of the rabies virus N gene. The probe hybridization was performed from amplified N gene obtained by reverse transcription polymerase chain reaction and the results by RT-PCR and hybridization showed 100 percent agreement. The hybridization, when carried out in products amplified by RT-PCR, increases the sensitivity of this technique even more and confers specificity to the diagnosis. The technique described in this work will be useful in rabies diagnosis laboratories, once the cost is compatible with traditional rabies diagnostic techniques.


Subject(s)
Humans , Animals , Cattle , Dogs , DNA, Viral/genetics , Digoxigenin , Nucleoproteins/genetics , Rabies virus/genetics , Rabies/diagnosis , Luminescent Measurements , Blotting, Southern , Chiroptera , DNA Probes , Horses , In Situ Hybridization , Rabies/virology , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Sheep
20.
Braz. j. microbiol ; 35(4): 348-351, Oct.-Dec. 2004. ilus, graf
Article in English | LILACS | ID: lil-402622

ABSTRACT

O Vírus Respiratório Sincicial Humano (VRSH) é descrito como o mais importante patógeno viral causador de doenças respiratórias agudas das vias respiratórias inferiores em crianças. Neste estudo 84 amostras de crianças com idade abaixo dos dois anos apresentando sintomas de doença respiratória aguda, foram obtidas no período de setembro de 2000 a novembro de 2001. Analise por imunofluorescência indireta e transcrição reversa seguida de PCR, revelou que 18 per center (15/84) das amostras foram positivas, sendo que em 80 per center (12/15) dos casos a detecção de VRSH foi observada em crianças abaixo dos seis meses, e também que os subgrupos A e B co-circularam. Estes são os primeiros dados obtidos para a cidade de Botucatu, sendo que a sazonalidade mostrou-se evidente pela maior circulação desse vírus entre os meses de maio e julho.


Subject(s)
Child , Respirovirus , Respirovirus Infections , Fluorescent Antibody Technique , Methods
SELECTION OF CITATIONS
SEARCH DETAIL
...