Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Virol ; 86(5): 2715-28, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190722

ABSTRACT

Genome sequences of transmitted/founder (T/F) HIV-1 have been inferred by analyzing single genome amplicons of acute infection plasma viral RNA in the context of a mathematical model of random virus evolution; however, few of these T/F sequences have been molecularly cloned and biologically characterized. Here, we describe the derivation and biological analysis of ten infectious molecular clones, each representing a T/F genome responsible for productive HIV-1 clade B clinical infection. Each of the T/F viruses primarily utilized the CCR5 coreceptor for entry and replicated efficiently in primary human CD4(+) T lymphocytes. This result supports the conclusion that single genome amplification-derived sequences from acute infection allow for the inference of T/F viral genomes that are consistently replication competent. Studies with monocyte-derived macrophages (MDM) demonstrated various levels of replication among the T/F viruses. Although all T/F viruses replicated in MDM, the overall replication efficiency was significantly lower compared to prototypic "highly macrophage-tropic" virus strains. This phenotype was transferable by expressing the env genes in an isogenic proviral DNA backbone, indicating that T/F virus macrophage tropism mapped to Env. Furthermore, significantly higher concentrations of soluble CD4 were required to inhibit T/F virus infection compared to prototypic macrophage-tropic virus strains. Our findings suggest that the acquisition of clinical HIV-1 subtype B infection occurs by mucosal exposure to virus that is not highly macrophage tropic and that the generation and initial biological characterization of 10 clade B T/F infectious molecular clones provides new opportunities to probe virus-host interactions involved in HIV-1 transmission.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/physiology , Macrophages/virology , Virus Replication , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Genome, Viral , HIV Infections/immunology , HIV-1/genetics , Humans , Macrophages/immunology , Male , Molecular Sequence Data
2.
Med Sci Educ ; 33(6): 1465-1471, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38188385

ABSTRACT

Medical students are the future of academic medicine. They will serve as admissions committee members, deans, and program directors with responsibility for selecting future physicians. At the same time, schools and programs are working to diversify the physician workforce to care for diverse patient populations. At UAB Heersink School of Medicine, we developed an elective course, Holistic Review: Creating a Mission-Driven Physician Workforce, to train learners in bias, diversity, and data-informed decision-making. Students applied lessons to the admissions process by participating in application screening and multiple-mini interview rating. Student reflections demonstrated course applicability for careers in medicine and beyond.

3.
J Virol ; 84(10): 4998-5006, 2010 May.
Article in English | MEDLINE | ID: mdl-20200250

ABSTRACT

Control of HIV-1 replication following nonsterilizing HIV-1 vaccination could be achieved by vaccine-elicited CD8(+) T-cell-mediated antiviral activity. To date, neither the functional nor the phenotypic profiles of CD8(+) T cells capable of this activity are clearly understood; consequently, little is known regarding the ability of vaccine strategies to elicit them. We used multiparameter flow cytometry and viable cell sorts from phenotypically defined CD8(+) T-cell subsets in combination with a highly standardized virus inhibition assay to evaluate CD8(+) T-cell-mediated inhibition of viral replication. Here we show that vaccination against HIV-1 Env and Gag-Pol by DNA priming followed by recombinant adenovirus type 5 (rAd5) boosting elicited CD8(+) T-cell-mediated antiviral activity against several viruses with either lab-adapted or transmitted virus envelopes. As it did for chronically infected virus controllers, this activity correlated with HIV-1-specific CD107a or macrophage inflammatory protein 1beta (MIP-1beta) expression from HIV-1-specific T cells. Moreover, for vaccinees or virus controllers, purified memory CD8(+) T cells from a wide range of differentiation stages were capable of significantly inhibiting virus replication. Our data define attributes of an antiviral CD8(+) T-cell response that may be optimized in the search for an efficacious HIV-1 vaccine.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , T-Lymphocyte Subsets/immunology , Adenoviridae/genetics , Flow Cytometry , Humans , Immunization, Secondary , Transduction, Genetic , Vaccination/methods , Vaccines, DNA/immunology
4.
AIDS Res Hum Retroviruses ; 31(12): 1278-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26101895

ABSTRACT

We previously developed replication-competent reporter HIV-1 (referred to herein as LucR.T2A reporter viruses), utilizing a "ribosome skipping" T2A peptide strategy to link Renilla luciferase (LucR) with Nef expression. The demonstrated utility for HIV-1 vaccine and transmission study applications included measurement of neutralizing antibody (NAb) activity in vaccine sera, improved cell-mediated virus inhibition assays, such as T cell-mediated virus inhibition and antibody-dependent cell-mediated cytotoxicity (ADCC) assays, and humanized mouse models. Herein, we extend our prior work and introduce reporter virus technology for applications that require fully functional Nef. We demonstrate that in CD4(+) T cells productively infected with LucR.T2A reporter viruses, T2A peptide-driven Nef expression and function, such as down-regulation of surface CD4 and MHC-I, were impaired. We overcame this limitation of LucR.T2A reporter viruses and achieved physiological Nef expression and function by engineering novel LucR reporter HIV-1 comprising 11 different internal ribosome entry site (IRES) elements chosen for size and relative activity. A range of Nef expression was observed in 293T cells transfected with the different LucR.IRES reporter virus constructs. Iteratively, we identified IRES reporter genomes that expressed Nef closest to physiological levels and produced virus with infectivity, titers, and replication kinetics similar to nonreporter viruses. Our results demonstrated that LucR reporter activity was stable over multiple replication cycles in peripheral blood mononuclear cells (PBMCs). Furthermore, we analyzed Nef functionality, i.e., down-modulation of MHC-I and CD4, following infection of T cell lines and PBMCs. Unlike LucR.T2A reporter virus, one of the redesigned LucR.IRES reporter viruses [containing the modified encephalomyocarditis virus (EMCV) 6ATR IRES element, "6ATRi"] demonstrated Nef expression and function similar to parental "nonreporter" virus. In a previously validated (nef-independent) T cell-based NAb neutralization assay, LucR.6ATRi reporter virus performed indistinguishably from LucR.T2A reporter virus. In summary, reporter viruses comprising the "6ATRi" element promise to augment HIV-1 vaccine and transmission research approaches requiring a sensitive reporter readout combined with wild-type Nef function.


Subject(s)
Gene Expression Profiling , HIV-1/physiology , Internal Ribosome Entry Sites , Luciferases, Renilla/analysis , Protein Biosynthesis , Virus Replication , nef Gene Products, Human Immunodeficiency Virus/biosynthesis , CD4-Positive T-Lymphocytes/virology , Down-Regulation , Genes, Reporter , HIV-1/genetics , Histocompatibility Antigens Class I/biosynthesis , Humans , Luciferases, Renilla/genetics , nef Gene Products, Human Immunodeficiency Virus/metabolism
5.
J Immunol Methods ; 409: 161-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24291126

ABSTRACT

Emergence of SIV and HIV specific CD8 T cells has been shown to correlate with control of in vivo replication. Poor correlation between IFN-γ ELISPOT responses and in vivo control of the virus has triggered the development of more relevant assays to assess functional HIV-1 specific CD8 T-cell responses for the evaluation and prioritization of new HIV-1 vaccine candidates. We previously established a viral inhibition assay (VIA) that measures the ability of vaccine-induced CD8 T-cell responses to inhibit viral replication in autologous CD4 T cells. In this assay, viral replication is determined by measuring p24 in the culture supernatant. Here we describe the development of a novel VIA, referred to as IMC LucR VIA that exploits replication-competent HIV-1 infectious molecular clones (IMCs) in which the complete proviral genome is strain-specific and which express the Renilla luciferase (LucR) gene to determine viral growth and inhibition. The introduction of the luciferase readout does provide significant improvement of the read out time. In addition to switching to the LucR read out, changes made to the overall protocol resulted in the miniaturization of the assay from a 48 to a 96-well plate format, which preserved sample and allowed for the introduction of replicates. The overall assay time was reduced from 13 to 8 days. The assay has a high degree of specificity, and the previously observed non-specific background inhibition in cells from HIV-1 negative volunteers has been reduced dramatically. Importantly, we observed an increase in positive responses, indicating an improvement in sensitivity compared to the original VIA. Currently, only a limited number of "whole-genome" IMC-LucR viruses are available and our efforts will focus on expanding the panel to better evaluate anti-viral breadth. Overall, we believe the IMC LucR VIA provides a platform to assess functional CD8 T-cell responses in large-scale clinical trial testing, which will enhance the ability to select the most promising HIV-1 vaccine candidates capable of controlling HIV-1 replication in vivo.


Subject(s)
AIDS Vaccines/therapeutic use , CD8-Positive T-Lymphocytes/virology , Genes, Reporter , HIV Infections/therapy , HIV-1/growth & development , High-Throughput Screening Assays/standards , Luciferases, Renilla/biosynthesis , Virus Replication , Automation, Laboratory/standards , CD8-Positive T-Lymphocytes/immunology , Cell Line , Guideline Adherence/standards , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , HIV Infections/blood , HIV Infections/diagnosis , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Luciferases, Renilla/genetics , Miniaturization/standards , Observer Variation , Practice Guidelines as Topic/standards , Predictive Value of Tests , Quality Control , Reproducibility of Results , Time Factors , Transfection , Treatment Outcome
6.
AIDS Res Hum Retroviruses ; 26(3): 279-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20218881

ABSTRACT

Bacterial lipopolysaccharide (endotoxin) is a frequent contaminant of biological specimens and is also known to be a potent inducer of beta-chemokines and other soluble factors that inhibit HIV-1 infection in vitro. Though lipopolysaccharide (LPS) has been shown to stimulate the production of soluble HIV-1 inhibitors in cultures of monocyte-derived macrophages, the ability of LPS to induce similar inhibitors in other cell types is poorly characterized. Here we show that LPS exhibits potent anti-HIV activity in phytohemagglutinin-stimulated peripheral blood mononuclear cells (PBMCs) but has no detectable anti-HIV-1 activity in TZM-bl cells. The anti-HIV-1 activity of LPS in PBMCs was strongly associated with the production of beta-chemokines from CD14-positive monocytes. Culture supernatants from LPS-stimulated PBMCs exhibited potent anti-HIV-1 activity when added to TZM-bl cells but, in this case, the antiviral activity appeared to be related to IFN-gamma rather than to beta-chemokines. These observations indicate that LPS stimulates PBMCs to produce a complex array of soluble HIV-1 inhibitors, including beta-chemokines and IFN-gamma, that differentially inhibit HIV-1 depending on the target cell type. The results also highlight the need to use endotoxin-free specimens to avoid artifacts when assessing HIV-1-specific neutralizing antibodies in PBMC-based assays.


Subject(s)
Chemokines, CC/biosynthesis , HIV-1/drug effects , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/pharmacology , Antibodies, Monoclonal/drug effects , Antibodies, Neutralizing/drug effects , Cell Line, Tumor , Equipment Contamination , Escherichia coli/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/metabolism , Neutralization Tests , Salmonella enterica/metabolism
7.
Virology ; 408(1): 1-13, 2010 Dec 05.
Article in English | MEDLINE | ID: mdl-20863545

ABSTRACT

Effective vaccine development for human immunodeficiency virus type 1 (HIV-1) will require assays that ascertain the capacity of vaccine immunogens to elicit neutralizing antibodies (NAb) to diverse HIV-1 strains. To facilitate NAb assessment in peripheral blood mononuclear cell (PBMC)-based assays, we developed an assay-adaptable platform based on a Renilla luciferase (LucR) expressing HIV-1 proviral backbone. LucR was inserted into pNL4-3 DNA, preserving all viral open reading frames. The proviral genome was engineered to facilitate expression of diverse HIV-1 env sequences, allowing analysis in an isogenic background. The resulting Env-IMC-LucR viruses are infectious, and LucR is stably expressed over multiple replications in PBMC. HIV-1 neutralization, targeting TZM-bl cells, was highly correlative comparing virus (LucR) and cell (firefly luciferase) readouts. In PBMC, NAb activity can be analyzed either within a single or multiple cycles of replication. These results represent advancement toward a standardizable PBMC-based neutralization assay for assessing HIV-1 vaccine immunogen efficacy.


Subject(s)
HIV Antibodies/immunology , HIV-1/growth & development , Leukocytes, Mononuclear/virology , Luciferases, Renilla/biosynthesis , Staining and Labeling/methods , Virology/methods , Virus Replication , AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Luciferases, Renilla/genetics , Neutralization Tests/methods , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
8.
J Exp Med ; 207(4): 763-76, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20368576

ABSTRACT

Traditional antibody-mediated neutralization of HIV-1 infection is thought to result from the binding of antibodies to virions, thus preventing virus entry. However, antibodies that broadly neutralize HIV-1 are rare and are not induced by current vaccines. We report that four human anti-phospholipid monoclonal antibodies (mAbs) (PGN632, P1, IS4, and CL1) inhibit HIV-1 CCR5-tropic (R5) primary isolate infection of peripheral blood mononuclear cells (PBMCs) with 80% inhibitory concentrations of <0.02 to approximately 10 microg/ml. Anti-phospholipid mAbs inhibited PBMC HIV-1 infection in vitro by mechanisms involving binding to monocytes and triggering the release of MIP-1alpha and MIP-1beta. The release of these beta-chemokines explains both the specificity for R5 HIV-1 and the activity of these mAbs in PBMC cultures containing both primary lymphocytes and monocytes.


Subject(s)
Antibodies, Antiphospholipid/pharmacology , Antibodies, Monoclonal/pharmacology , Chemokines, CC/metabolism , HIV-1/physiology , Receptors, CCR5/physiology , Viral Tropism/physiology , Virus Internalization/drug effects , Antibodies, Antiphospholipid/genetics , Antibodies, Antiphospholipid/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cardiolipins/immunology , Cell Fusion , Chemokine CCL3/immunology , Chemokine CCL3/metabolism , Chemokine CCL4/immunology , Chemokine CCL4/metabolism , Chemokines/metabolism , Complementarity Determining Regions/genetics , Culture Media, Conditioned/pharmacology , Endotoxins/pharmacology , Epithelial Cells/virology , Giant Cells/cytology , HIV-1/classification , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fc Fragments/immunology , Kinetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Mutation/genetics , Mutation/immunology , Phosphatidylethanolamines/immunology , Phosphatidylserines/immunology , beta 2-Glycoprotein I/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL