Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Microbiol Biol Educ ; 23(3)2022 Dec.
Article in English | MEDLINE | ID: mdl-36532218

ABSTRACT

Laboratory exercises for undergraduates that involve authentic discovery and research have been shown to increase student learning and engagement. To bring these advantages into the microbiology curriculum, we developed a semester-long course-based undergraduate research experience for a laboratory based on brewing beer with wild yeast. This set of lab exercises uses many of the same protocols found in traditional microbiology lab curricula-isolating and maintaining pure cultures, staining and microscopy, use of aseptic technique, PCR, gel electrophoresis, and media preparation-and integrates them into a novel and exciting project that enables students to be active participants in the scientific method. Students are assessed on their ability to brew beer successfully and to stain and visualize microorganisms; they are also assessed for knowledge gains in the traditional portion of the course, their ability to use their brewing knowledge in other settings, and their attitudes about science. After completing the course, students showed gains in general microbiology knowledge and their engagement with science.

2.
PLoS Pathog ; 16(8): e1008685, 2020 08.
Article in English | MEDLINE | ID: mdl-32745153

ABSTRACT

Smallpox and monkeypox pose severe threats to human health. Other orthopoxviruses are comparably virulent in their natural hosts, including ectromelia, the cause of mousepox. Disease severity is linked to an array of immunomodulatory proteins including the B22 family, which has homologs in all pathogenic orthopoxviruses but not attenuated vaccine strains. We demonstrate that the ectromelia B22 member, C15, is necessary and sufficient for selective inhibition of CD4+ but not CD8+ T cell activation by immunogenic peptide and superantigen. Inhibition is achieved not by down-regulation of surface MHC- II or co-stimulatory protein surface expression but rather by interference with antigen presentation. The appreciable outcome is interference with CD4+ T cell synapse formation as determined by imaging studies and lipid raft disruption. Consequently, CD4+ T cell activating stimulus shifts to uninfected antigen-presenting cells that have received antigen from infected cells. This work provides insight into the immunomodulatory strategies of orthopoxviruses by elucidating a mechanism for specific targeting of CD4+ T cell activation, reflecting the importance of this cell type in control of the virus.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Histocompatibility Antigens Class II/immunology , Viral Proteins/immunology , Animals , Ectromelia, Infectious/metabolism , Ectromelia, Infectious/virology , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Proteins/metabolism , Virulence
3.
J Immunol ; 202(5): 1340-1349, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30700590

ABSTRACT

CD4+ T cells play critical roles in defending against poxviruses, both by potentiating cellular and humoral responses and by directly killing infected cells. Despite this central role, the basis for pox-specific CD4+ T cell activation, specifically the origin of the poxvirus-derived peptides (epitopes) that activate CD4+ T cells, remains poorly understood. In addition, because the current licensed poxvirus vaccines can cause serious adverse events and even death, elucidating the requirements for MHC class II (MHC-II) processing and presentation of poxviral Ags could be of great use. To address these questions, we explored the CD4+ T cell immunogenicity of ectromelia, the causative agent of mousepox. Having identified a large panel of novel epitopes via a screen of algorithm-selected synthetic peptides, we observed that immunization of mice with inactivated poxvirus primes a virtually undetectable CD4+ T cell response, even when adjuvanted, and is unable to provide protection against disease after a secondary challenge. We postulated that an important contributor to this outcome is the poor processability of whole virions for MHC-II-restricted presentation. In line with this hypothesis, we observed that whole poxvirions are very inefficiently converted into MHC-II-binding peptides by the APC as compared with subviral material. Thus, stability of the virion structure is a critical consideration in the rational design of a safe alternative to the existing live smallpox vaccine.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Immunogenicity, Vaccine/immunology , Poxviridae/immunology , Vaccines, Inactivated/immunology , Animals , Female , Mice , Mice, Inbred C57BL
4.
J Virol ; 88(16): 9472-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899206

ABSTRACT

We assessed several routes of immunization with vaccinia virus (VACV) in protecting mice against ectromelia virus (ECTV). By a wide margin, skin scarification provided the greatest protection. Humoral immunity and resident-memory T cells notwithstanding, several approaches revealed that circulating, memory CD8(+) T cells primed via scarification were functionally superior and conferred enhanced virus control. Immunization via the epithelial route warrants further investigation, as it may also provide enhanced defense against other infectious agents.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Epithelium/immunology , Vaccinia virus/immunology , Animals , Immunity, Humoral/immunology , Immunization/methods , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Vaccination/methods
5.
J Gen Virol ; 92(Pt 9): 1971-1980, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21715594

ABSTRACT

The vaccinia virus A56 protein was one of the earliest-described poxvirus proteins with an identifiable activity. While originally characterized as a haemagglutinin protein, A56 has other functions as well. The A56 protein is capable of binding two viral proteins, a serine protease inhibitor (K2) and the vaccinia virus complement control protein (VCP), and anchoring them to the surface of infected cells. This is important; while both proteins have biologically relevant functions at the cell surface, neither one can locate there on its own. The A56-K2 complex reduces the amount of virus superinfecting an infected cell and also prevents the formation of syncytia by infected cells; the A56-VCP complex can protect infected cells from complement attack. Deletion of the A56R gene results in varying effects on vaccinia virus virulence. In addition, since the gene encoding the A56 protein is non-essential, it can be used as an insertion point for foreign genes and has been deleted in some viruses that are in clinical development as oncolytic agents.


Subject(s)
Glycoproteins/metabolism , Membrane Proteins/metabolism , Vaccinia virus/physiology , Viral Proteins/metabolism , Virulence Factors/metabolism , Gene Deletion , Humans , Viral Proteins/genetics
6.
J Virol ; 85(6): 2547-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21191012

ABSTRACT

Complement activation is an important component of the innate immune response against viral infection and also shapes adaptive immune responses. Despite compelling evidence that complement activation enhances T cell and antibody (Ab) responses during viral infection, it is unknown whether inhibition of complement by pathogens alters these responses. Vaccinia virus (VACV) modulates complement activation by encoding a complement regulatory protein called the vaccinia virus complement control protein (VCP). Although VCP has been described as a virulence factor, the mechanisms by which VCP enhances VACV pathogenesis have not been fully defined. Since complement is necessary for optimal adaptive immune responses to several viruses, we hypothesized that VCP contributes to pathogenesis by modulating anti-VACV T cell and Ab responses. In this study, we used an intradermal model of VACV infection to compare pathogenesis of wild-type virus (vv-VCPwt) and a virus lacking VCP (vv-VCPko). vv-VCPko formed smaller lesions in wild-type mice but not in complement-deficient mice. Attenuation of vv-VCPko correlated with increased accumulation of T cells at the site of infection, enhanced neutralizing antibody responses, and reduced viral titers. Importantly, depleting CD8(+) T cells together with CD4(+) T cells, which also eliminated T helper cell-dependent Ab responses, restored vv-VCPko to wild-type levels of virulence. These results suggest that VCP contributes to virulence by dampening both antibody and T cell responses. This work provides insight into how modulation of complement by poxviruses contributes to virulence and demonstrates that a pathogen-encoded complement regulatory protein can modulate adaptive immunity.


Subject(s)
Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Antibodies, Neutralizing/analysis , Antibodies, Viral/analysis , Disease Models, Animal , Female , Gene Deletion , Mice , Skin/pathology , Skin/virology , T-Lymphocytes/immunology , Viral Load , Viral Proteins/genetics , Virulence Factors/genetics
7.
J Virol ; 84(21): 11245-54, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719953

ABSTRACT

The vaccinia virus (VACV) complement control protein (VCP) is an immunomodulatory protein that is both secreted from and expressed on the surface of infected cells. Surface expression of VCP occurs though an interaction with the viral transmembrane protein A56 and is dependent on a free N-terminal cysteine of VCP. Although A56 and VCP have been shown to interact in infected cells, the mechanism remains unclear. To investigate if A56 is sufficient for surface expression, we transiently expressed VCP and A56 in eukaryotic cell lines and found that they interact on the cell surface in the absence of other viral proteins. Since A56 contains three extracellular cysteines, we hypothesized that one of the cysteines may be unpaired and could therefore form a disulfide bridge with VCP. To test this, we generated a series of A56 mutants in which each cysteine was mutated to a serine, and we found that mutation of cysteine 162 abrogated VCP cell surface expression. We also tested the ability of other poxvirus complement control proteins to bind to VACV A56. While the smallpox homolog of VCP is able to bind VACV A56, the ectromelia virus (ECTV) VCP homolog is only able to bind the ECTV homolog of A56, indicating that these proteins may have coevolved. Surface expression of poxvirus complement control proteins may have important implications in viral pathogenesis, as a virus that does not express cell surface VCP is attenuated in vivo. This suggests that surface expression of VCP may contribute to poxvirus pathogenesis.


Subject(s)
Cell Membrane/virology , Poxviridae/pathogenicity , Viral Proteins/metabolism , Cell Line , Cysteine/genetics , Cysteine/metabolism , Disulfides , Humans , Mutagenesis, Site-Directed
8.
J Virol ; 82(9): 4205-14, 2008 May.
Article in English | MEDLINE | ID: mdl-18287241

ABSTRACT

The vaccinia virus (VACV) complement control protein (VCP) is the major protein secreted from VACV-infected cells. It has been reported that VCP binds to the surfaces of uninfected cells by interacting with heparan sulfate proteoglycans (HSPGs). In this study, we show that VCP is also expressed on the surfaces of infected cells and demonstrate that surface localization occurs independently of HSPGs. Since VCP does not contain a transmembrane domain, we hypothesized that VCP interacts with a membrane protein that localizes to the infected-cell surface. We show that the VACV A56 membrane protein is necessary for the cell surface expression of VCP and demonstrate that VCP and A56 interact in VACV-infected cells. Since the surface expression of VCP was abrogated by reducing agents, we examined the contribution of an unpaired cysteine residue on VCP to VCP surface expression and VCP's interaction with A56. To do this, we mutated the unpaired cysteine in VCP and generated a recombinant virus expressing the altered form of VCP. Following the infection of cells with the mutant virus, VCP was neither expressed on the cell surface nor able to interact with A56. Importantly, the cell surface expression of VCP was found to protect infected cells from complement-mediated lysis. Our findings suggest a new function for VCP that may be important for poxvirus pathogenesis and impact immune responses to VACV-based vaccines.


Subject(s)
Complement Activation , Vaccinia virus/pathogenicity , Vaccinia/immunology , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , Viral Proteins/physiology , Cell Line , Cysteine/genetics , Gene Expression Regulation, Viral/immunology , Heparin/analogs & derivatives , Humans , Mutagenesis, Site-Directed , Proteoglycans , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...