Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Phuoc T. Vo is active.

Publication


Featured researches published by Phuoc T. Vo.


Journal of Clinical Investigation | 2002

Inhibition of rotavirus replication by a non-neutralizing, rotavirus VP6–specific IgA mAb

Ningguo Feng; Jeffrey A. Lawton; Joana Gilbert; Nelly A. Kuklin; Phuoc T. Vo; B. V. Venkataram Prasad; Harry B. Greenberg

Rotaviruses are the leading cause of severe diarrheal disease in young children. Intestinal mucosal IgA responses play a critical role in protective immunity against rotavirus reinfection. Rotaviruses consist of three concentric capsid layers surrounding a genome of 11 segments of double-stranded RNA. The outer layer proteins, VP4 and VP7, which are responsible for viral attachment and entry, are targets for protective neutralizing antibodies. However, IgA mAbs directed against the intermediate capsid protein VP6, which do not neutralize the virus, have also been shown to protect mice from rotavirus infection and clear chronic infection in SCID mice. We investigated whether the anti-VP6 IgA (7D9) mAb could inhibit rotavirus replication inside epithelial cells and found that 7D9 acted at an early stage of infection to neutralize rotavirus following antibody lipofection. Using electron cryomicroscopy, we determined the three-dimensional structure of the virus-antibody complex. The attachment of 7D9 IgA to VP6 introduces a conformational change in the VP6 trimer, rendering the particle transcriptionally incompetent and preventing the elongation of initiated transcripts. Based on these observations, we suggest that anti-VP6 IgA antibodies confers protection in vivo by inhibiting viral transcription at the start of the intracellular phase of the viral replication cycle.


Virology | 1986

Antigenic mapping of the surface proteins of rhesus rotavirus

Robert D. Shaw; Phuoc T. Vo; Paul A. Offit; Barbara S. Coulsont; Harry B. Greenberg

Monoclonal antibodies have been produced and used to map the functional topography of the surface proteins of rhesus rotavirus (RRV) that mediate viral neutralization. Ten monoclonal antibodies directed to VP7 were studied in neutralization assays and competitive binding studies. A large neutralization domain with several interrelated epitopes on VP7 was apparent. Twelve monoclonal antibodies directed to VP3 were used in similar studies and delineated at least 2 distinct neutralization domains on that protein. Neutralizing monoclonal antibodies directed at both VP3 and VP7 were used to isolate viral antigenic variants, which were than studied in neutralization and hemagglutination inhibition assays. The viral variant studies, while confirming the general conclusions obtained from the competitive binding studies, allowed the apparent distinction of two separate neutralization domains on VP7 and three on VP3. All VP7-specific monoclonal antibodies (mAb) mediated serotype-specific neutralization, but a VP3-specific mAb was identified that neutralized rotaviruses of three distinct serotypes. No alteration of viral virulence was apparent in studies of suckling mice orally inoculated with antigenic variant viruses selected with our panel of neutralizing VP3 or VP7-specific mAbs.


Virology | 1988

Characterization of homotypic and heterotypic VP7 neutralization sites of rhesus rotavirus

Erich R. Mackow; Robert D. Shaw; Suzanne M. Matsui; Phuoc T. Vo; David A. Benfield; Harry B. Greenberg

The gene 9 nucleotide sequence was determined for rhesus rotavirus and each of 14 viral variants selected for their resistance to neutralizing monoclonal antibodies. Each variant contains a single gene 9, VP7, mutation which permits viral growth in the presence of the antibody. Variant mutations were identified in two distinct neutralization regions. Region A was identified by monoclonal antibodies that are involved in both serotype-specific and serotype cross-reactive neutralization. Region C was identified by serotype-specific neutralizing monoclonal antibodies. Heterotypic neutralizing monoclonal antibody 57-8 selected variants with a mutation at amino acid 94 in the A region, the same amino acid location selected by serotype-specific monoclonal antibodies. Monoclonal antibody 3 selected a VP7 mutation at amino acid 99 resulting in additional N-linked glycosylation of the VP7 protein. Despite the added VP7 glycosylation, variant v3 was not broadly resistant to additional VP7-specific neutralizing monoclonal antibodies.


Journal of Virology | 2008

Role of Interferon in Homologous and Heterologous Rotavirus Infection in the Intestines and Extraintestinal Organs of Suckling Mice

Ningguo Feng; Bumseok Kim; Martijn Fenaux; H. Nguyen; Phuoc T. Vo; M. B. Omary; Harry B. Greenberg

ABSTRACT Recent studies demonstrated that viremia and extraintestinal rotavirus infection are common in acutely infected humans and animals, while systemic diseases appear to be rare. Intraperitoneal infection of newborn mice with rhesus rotavirus (RRV) results in biliary atresia (BA), and this condition is influenced by the host interferon response. We studied orally inoculated 5-day-old suckling mice that were deficient in interferon (IFN) signaling to evaluate the role of interferon on the outcome of local and systemic infection after enteric inoculation. We found that systemic replication of RRV, but not murine rotavirus strain EC, was greatly enhanced in IFN-α/β and IFN-γ receptor double-knockout (KO) or STAT1 KO mice but not in mice deficient in B- or T-cell immunity. The enhanced replication of RRV was associated with a lethal hepatitis, pancreatitis, and BA, while no systemic disease was observed in strain EC-infected interferon-deficient mice. In IFN-α/β receptor KO mice the extraintestinal infection and systemic disease were only moderately increased, while RRV infection was not augmented and systemic disease was not present in IFN-γ receptor KO mice. The increase of systemic infection in IFN-deficient mice was also observed during simian strain SA11 infection but not following bovine NCDV, porcine OSU, or murine strain EW infection. Our data indicate that the requirements for the interferon system to inhibit intestinal and extraintestinal viral replication in suckling mice vary among different heterologous and homologous rotavirus strains, and this variation is associated with lethal systemic disease.


Gastroenterology | 1987

Epitope-Specific Immune Responses to Rotavirus Vaccination

Robert D. Shaw; Karen J. Fong; Genevieve Losonsky; Myron M. Levine; Yvonne Maldonado; Robert H. Yolken; Albert Z. Kapikian; Phuoc T. Vo; Harry B. Greenberg

Rotavirus gastroenteritis is a leading cause of infant mortality in developing countries and an important cause of morbidity in children under 2 yr of age in the United States. Vaccine programs have evaluated animal rotavirus strains that are attenuated in humans but antigenically similar to some human strains. Whether a single vaccine strain can elicit protective immunity in humans to rotaviruses of the same or different serotypes is an important question in determining vaccine efficacy. We used characterized serotype-specific monoclonal antibodies directed at VP7 in a competitive solid-phase immunoassay to measure epitope-specific immune responses to serotypes 1, 2, and 3 in sera of children who received a candidate serotype-3 rotavirus vaccine. Antibodies to serotype 3 were detected in 72% of sera samples, and to serotype 1 and 2 in only 11% each. Also, a VP3-specific monoclonal antibody which neutralizes three serotypically distinct strains of rotavirus was used to detect the presence of similar antibodies in 56% of the test sera. This finding suggests a mechanism of heterotypic immunity.


Journal of Virology | 2009

Variation in Antagonism of the Interferon Response to Rotavirus NSP1 Results in Differential Infectivity in Mouse Embryonic Fibroblasts

Ningguo Feng; Adrish Sen; H. Nguyen; Phuoc T. Vo; Yasutaka Hoshino; Emily M. Deal; Harry B. Greenberg

ABSTRACT Rotavirus NSP1 has been shown to function as an E3 ubiquitin ligase that mediates proteasome-dependent degradation of interferon (IFN) regulatory factors (IRF), including IRF3, -5, and -7, and suppresses the cellular type I IFN response. However, the effect of rotavirus NSP1 on viral replication is not well defined. Prior studies used genetic analysis of selected reassortants to link NSP1 with host range restriction in the mouse, suggesting that homologous and heterologous rotaviruses might use their different abilities to antagonize the IFN response as the basis of their host tropisms. Using a mouse embryonic fibroblast (MEF) model, we demonstrate that heterologous bovine (UK and NCDV) and porcine (OSU) rotaviruses fail to effectively degrade cellular IRF3, resulting in IRF3 activation and beta IFN (IFN-β) secretion. As a consequence of this failure, replication of these viruses is severely restricted in IFN-competent wild-type, but not in IFN-deficient (IFN-α/β/γ receptor- or STAT1-deficient) MEFs. On the other hand, homologous murine rotaviruses (ETD or EHP) or the heterologous simian rotavirus (rhesus rotavirus [RRV]) efficiently degrade cellular IRF3, diminish IRF3 activation and IFN-β secretion and are not replication restricted in wild-type MEFs. Genetic reassortant analysis between UK and RRV maps the distinctive phenotypes of IFN antagonism and growth restriction in wild-type MEFs to NSP1. Therefore, there is a direct relationship between the replication efficiencies of different rotavirus strains in MEFs and strain-related variations in NSP1-mediated antagonism of the type I IFN response.


Cellular Immunology | 1982

Protection against pulmonary blastomycosis: Adoptive transfer with T lymphocytes, but not serum, from resistant mice

Elmer Brummer; Pius A. Morozumi; Phuoc T. Vo; David A. Stevens

Abstract A murine model of pulmonary blastomycosis, in which the portal of entry is the same as in natural human infection, was used to study resistance to infection subsequent to pulmonary challenge. Lymphoid cells or serum from mice rendered resistant to fatal pulmonary challenge by resolution of a subcutaneous nonlethal infection were tested for ability to adoptively or passively transfer resistance to the pulmonary compartment of naive recipients. As one example, spleen cells (88 × 10 6 ), lymph node cells (17 × 10 6 ), and peritoneal cells (2 × 10 6 ) from resistant mice, but not normal mice, given iv transferred significant resistance to an LD 100 pulmonary challenge of naive recipients. Serum from resistant mice (ELISA titer 1:80) did not transfer protection. T lymphocytes (18 × 10 6 ) were as effective as unfractionated spleen cells (75 × 10 6 ) in the adoptive transfer of resistance. Spleen cells from resistant mice, when treated with anti-mouse T-cell serum plus complement (20 × 10 6 viable cells), failed to transfer resistance. The transfer of resistance with T lymphocytes from resistant to naive mice was T-cell dose dependent.


Journal of Virology | 2011

Rhesus Rotavirus Entry into a Polarized Epithelium Is Endocytosis Dependent and Involves Sequential VP4 Conformational Changes

Marie Wolf; Phuoc T. Vo; Harry B. Greenberg

ABSTRACT Rotavirus (RV) cell entry is an incompletely understood process, involving VP4 and VP7, the viral proteins composing the outermost layer of the nonenveloped RV triple-layered icosahedral particle (TLP), encasing VP6. VP4 can exist in three conformational states: soluble, cleaved spike, and folded back. In order to better understand the events leading to RV entry, we established a detection system to image input virus by monitoring the rhesus RV (RRV) antigens VP4, VP6, and VP7 at very early times postinfection. We provide evidence that decapsidation occurs directly after cell membrane penetration. We also demonstrate that several VP4 and VP7 conformational changes take place during entry. In particular, we detected, for the first time, the generation of folded-back VP5 in the context of the initiation of infection. Folded-back VP5 appears to be limited to the entry step. We furthermore demonstrate that RRV enters the cell cytoplasm through an endocytosis pathway. The endocytosis hypothesis is supported by the colocalization of RRV antigens with the early endosome markers Rab4 and Rab5. Finally, we provide evidence that the entry process is likely dependent on the endocytic Ca2+ concentration, as bafilomycin A1 treatment as well as an augmentation of the extracellular calcium reservoir using CaEGTA, which both lead to an elevated intraendosomal calcium concentration, resulted in the accumulation of intact virions in the actin network. Together, these findings suggest that internalization, decapsidation, and cell membrane penetration involve endocytosis, calcium-dependent uncoating, and VP4 conformational changes, including a fold-back.


Journal of Virology | 2009

VP5* Rearranges when Rotavirus Uncoats

Joshua D. Yoder; Shane D. Trask; Phuoc T. Vo; Mawuena Binka; Ningguo Feng; Stephen C. Harrison; Harry B. Greenberg; Philip R. Dormitzer

ABSTRACT Trypsin primes rotavirus for efficient infectivity by cleaving the spike protein, VP4, into VP8* and VP5*. A recombinant VP5* fragment has a trimeric, folded-back structure. Comparison of this structure with virion spikes suggests that a rearrangement, analogous to those of enveloped virus fusion proteins, may mediate membrane penetration by rotavirus during entry. To detect this inferred rearrangement of virion-associated authentic VP5*, we raised conformation-specific monoclonal antibodies against the recombinant VP5* fragment in its putative post-membrane penetration conformation. Using one of these antibodies, we demonstrate that rotavirus uncoating triggers a conformational change in the cleaved VP4 spike to yield rearranged VP5*.


Journal of General Virology | 1996

Cleavage of rotavirus VP4 in vivo

Juan E. Ludert; Ajit A. Krishnaney; John W. Burns; Phuoc T. Vo; Harry B. Greenberg

The infectivity of rotavirus particles is dependent on proteolytic cleavage of the outer capsid protein, VP4, at a specific site. This cleavage event yields two fragments, identified as VP5* and VP8*. It has been hypothesized that the particle is more stable, but non-infectious, when VP4 is in the uncleaved state. Uncleaved VP4 and the resultant increased stability might be advantageous for the virus to resist environmental degradation until it infects a susceptible host. When VP4 is cleaved in the lumen of the hosts gastrointestinal tract, the virus particle would become less stable but more infectious. To test this hypothesis, a series of experiments was undertaken to analyse the cleavage state of VP4 on virus shed by an infected host into the environment. Immunoblots of intestinal wash solutions derived from infant and adult BALB/c mice infected with a virulent cell culture-adapted variant of the EDIM virus (EW) or wild-type murine rotavirus EDIM-Cambridge were analysed. Virtually all of the VP4 in these samples was in the cleaved form. Moreover, cell culture titration of trypsin-treated and untreated intestinal contents from pups infected with EW indicated that excreted virus is fully activated prior to trypsin addition. It was also observed that trypsin-activated virus has no disadvantage in initiating infection in naive animals over virions containing an intact VP4. These studies indicate that VP4 is cleaved upon release from the intestinal cell and that virus shed into the environment does not have an intact VP4.

Collaboration


Dive into the Phuoc T. Vo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasutaka Hoshino

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge