Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Acad Radiol ; 29 Suppl 5: S82-S88, 2022 05.
Article in English | MEDLINE | ID: mdl-34987000

ABSTRACT

RATIONALE AND OBJECTIVES: We aim to compare Choose Your Own Adventure (CYOA) presentation format with linear case format as educational methods for teaching a radiology small group session to medical students. MATERIALS AND METHODS: A radiology small group session was held for preclinical second-year medical students in the pulmonary course, whereby eight classrooms of students and eight radiology facilitators were each randomized to do either the linear case format or the nonlinear CYOA presentation format. All students in attendance were administered a survey at the end of the session, which assessed students' perceptions using five-point Likert-type questions. The survey also contained a four-question knowledge quiz on chest radiology. The facilitators were administered a qualitative survey as well. Between-group analyses were performed using Student's t-test. RESULTS: Of the 144 students who attended the small group sessions, 143 students completed the survey (99.3%). The CYOA format group reported significantly greater engagement in the cases (4.5 ± 0.7 vs. 3.8 ± 0.7, p < 0.001), satisfaction with the format (4.6 ± 0.6 vs. 3.7 ± 0.9, p < 0.001), and enhancement of clinical decision making skills (4.5 ± 0.6 vs. 3.5 ± 0.9, p < 0.001). The linear format group reported a greater role for the facilitator to add value (4.6 ± 0.5 vs. 4.3 ± 1.1, p = 0.033). There was no significant difference between groups in performance on the knowledge quiz. CONCLUSION: Medical students reported higher satisfaction, engagement, and enhanced clinical decision making skills with the CYOA presentation method compared to linear case format for radiology small group learning.


Subject(s)
Radiology , Students, Medical , Humans , Learning , Radiography , Radiology/education , Surveys and Questionnaires , Teaching
2.
Cell ; 168(5): 755-757, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235193

ABSTRACT

Three recent studies find that the single-pass transmembrane protein HAP2 mediates gamete fusion and is remarkably similar to class II fusion proteins found in viruses such as dengue and Zika.


Subject(s)
Germ Cells/metabolism , Membrane Proteins/metabolism , Dengue , Humans , Zika Virus , Zika Virus Infection
3.
Proc Natl Acad Sci U S A ; 113(24): E3413-22, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27247400

ABSTRACT

Most simian-human immunodeficiency viruses (SHIVs) bearing envelope (Env) glycoproteins from primary HIV-1 strains fail to infect rhesus macaques (RMs). We hypothesized that inefficient Env binding to rhesus CD4 (rhCD4) limits virus entry and replication and could be enhanced by substituting naturally occurring simian immunodeficiency virus Env residues at position 375, which resides at a critical location in the CD4-binding pocket and is under strong positive evolutionary pressure across the broad spectrum of primate lentiviruses. SHIVs containing primary or transmitted/founder HIV-1 subtype A, B, C, or D Envs with genotypic variants at residue 375 were constructed and analyzed in vitro and in vivo. Bulky hydrophobic or basic amino acids substituted for serine-375 enhanced Env affinity for rhCD4, virus entry into cells bearing rhCD4, and virus replication in primary rhCD4 T cells without appreciably affecting antigenicity or antibody-mediated neutralization sensitivity. Twenty-four RMs inoculated with subtype A, B, C, or D SHIVs all became productively infected with different Env375 variants-S, M, Y, H, W, or F-that were differentially selected in different Env backbones. Notably, SHIVs replicated persistently at titers comparable to HIV-1 in humans and elicited autologous neutralizing antibody responses typical of HIV-1. Seven animals succumbed to AIDS. These findings identify Env-rhCD4 binding as a critical determinant for productive SHIV infection in RMs and validate a novel and generalizable strategy for constructing SHIVs with Env glycoproteins of interest, including those that in humans elicit broadly neutralizing antibodies or bind particular Ig germ-line B-cell receptors.


Subject(s)
CD4 Antigens/metabolism , HIV Infections , HIV-1/physiology , Mutation, Missense , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus/physiology , Virus Replication/genetics , env Gene Products, Human Immunodeficiency Virus , Amino Acid Substitution , Animals , HIV Infections/genetics , HIV Infections/metabolism , Humans , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/metabolism , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism
4.
Viruses ; 8(5)2016 05 13.
Article in English | MEDLINE | ID: mdl-27187446

ABSTRACT

The identification of host cellular genes that act as either proviral or antiviral factors has been aided by the development of an increasingly large number of high-throughput screening approaches. Here, we review recent advances in which these new technologies have been used to interrogate host genes for the ability to impact bunyavirus infection, both in terms of technical advances as well as a summary of biological insights gained from these studies.


Subject(s)
Bunyaviridae Infections/immunology , Bunyaviridae Infections/pathology , Host-Pathogen Interactions , Orthobunyavirus/pathogenicity , Genetic Testing/methods , High-Throughput Screening Assays , Humans
5.
AIDS Res Hum Retroviruses ; 32(7): 705-17, 2016 07.
Article in English | MEDLINE | ID: mdl-26847431

ABSTRACT

Platelet factor 4 (PF4) has been recently shown to inhibit infection by a broad range of human immunodeficiency virus type 1 (HIV-1) isolates in vitro. We found that the inhibitory effects of PF4 are limited to a defined concentration range where PF4 exists largely in a monomeric state. Under these conditions, PF4 bound the HIV-1 envelope protein and inhibited HIV-1 attachment to the cell surface. However, as concentrations increased to the point where PF4 exists largely in tetrameric or higher-order forms, viral infection in vitro was enhanced. Enhancement could be inhibited by mutations in PF4 that shift the oligomeric equilibrium toward the monomeric state, or by using soluble glycosaminoglycans (GAGs) to which tetrameric PF4 avidly binds. We conclude that at physiologically relevant concentrations, oligomeric PF4 enhances infection by HIV-1 by interacting with the viral envelope protein as well as cell surface GAGs, enhancing virus attachment to the cell surface. This effect was not specific to HIV-1, as enhancement was seen with some but not all other viruses tested. The biphasic effects of PF4 on HIV-1 infection suggest that native PF4 will not be a useful antiviral agent and that PF4 could contribute to the hematologic abnormalities commonly seen in HIV-infected individuals by enhancing virus infection in the bone marrow.


Subject(s)
HIV-1/physiology , Host-Pathogen Interactions , Platelet Factor 4/metabolism , Virus Attachment , env Gene Products, Human Immunodeficiency Virus/metabolism , Humans , Protein Binding
6.
J Virol ; 90(3): 1414-23, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26581979

ABSTRACT

UNLABELLED: Rift Valley fever virus (RVFV) causes recurrent insect-borne epizootics throughout the African continent, and infection of humans can lead to a lethal hemorrhagic fever syndrome. Deep mutagenesis of haploid human cells was used to identify host factors required for RVFV infection. This screen identified a suite of enzymes involved in glycosaminoglycan (GAG) biogenesis and transport, including several components of the cis-oligomeric Golgi (COG) complex, one of the central components of Golgi complex trafficking. In addition, disruption of PTAR1 led to RVFV resistance as well as reduced heparan sulfate surface levels, consistent with recent observations that PTAR1-deficient cells exhibit altered Golgi complex morphology and glycosylation defects. A variety of biochemical and genetic approaches were utilized to show that both pathogenic and attenuated RVFV strains require GAGs for efficient infection on some, but not all, cell types, with the block to infection being at the level of virion attachment. Examination of other members of the Bunyaviridae family for GAG-dependent infection suggested that the interaction with GAGs is not universal among bunyaviruses, indicating that these viruses, as well as RVFV on certain cell types, employ additional unidentified virion attachment factors and/or receptors. IMPORTANCE: Rift Valley fever virus (RVFV) is an emerging pathogen that can cause severe disease in humans and animals. Epizootics among livestock populations lead to high mortality rates and can be economically devastating. Human epidemics of Rift Valley fever, often initiated by contact with infected animals, are characterized by a febrile disease that sometimes leads to encephalitis or hemorrhagic fever. The global burden of the pathogen is increasing because it has recently disseminated beyond Africa, which is of particular concern because the virus can be transmitted by widely distributed mosquito species. There are no FDA-licensed vaccines or antiviral agents with activity against RVFV, and details of its life cycle and interaction with host cells are not well characterized. We used the power of genetic screening in human cells and found that RVFV utilizes glycosaminoglycans to attach to host cells. This furthers our understanding of the virus and informs the development of antiviral therapeutics.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Rift Valley fever virus/physiology , Virus Attachment , Cell Line , Genetic Testing , Heparan Sulfate Proteoglycans/genetics , Humans , Mutagenesis
7.
Vaccine ; 33(39): 5140-7, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26277072

ABSTRACT

The glycan shield on the human immunodeficiency virus 1 (HIV-1) envelope (Env) glycoprotein has drawn attention as a target for HIV-1 vaccine design given that an increasing number of potent and broadly neutralizing antibodies (bNAbs) recognize epitopes entirely or partially comprised of high mannose type N-linked glycans. In an attempt to generate immunogens that target the glycan shield of HIV-1, we previously engineered a triple mutant (TM) strain of Saccharomyces cerevisiae that results in exclusive presentation of high mannose type N-glycans, and identified five TM yeast glycoproteins that support strong binding of 2G12, a bNAb that targets a cluster of high mannose glycans on the gp120 subunit of Env. Here, we further analyzed the antigenicity and immunogenicity of these proteins in inducing anti-HIV responses. Our study demonstrated that the 2G12-reactive TM yeast glycoproteins efficiently bound to recently identified bNAbs including PGT125-130 and PGT135 that recognize high mannose glycan-dependent epitopes. Immunization of rabbits with a single TM yeast glycoprotein (Gp38 or Pst1), when conjugated to a promiscuous T-cell epitope peptide and coadministered with a Toll-like receptor 2 agonist, induced glycan-specific HIV-1 Env cross-reactive antibodies. The immune sera bound to both synthetic mannose oligosaccharides and gp120 proteins from a broad range of HIV-1 strains. The purified antibodies recognized and captured virions that contain both complex- and high mannose-type of N-glycans, and potently neutralized virions from different HIV-1 clades but only when the virions were enforced to retain high mannose N-glycans. This study provides insights into the elicitation of anti-carbohydrate, HIV-1 Env-cross reactive antibodies with a heterologous glycoprotein and may have applications in the design and administration of immunogens that target the viral glycan shield for development of an effective HIV-1 vaccine.


Subject(s)
Antibodies, Fungal/immunology , Antibodies, Heterophile/immunology , Antibodies, Neutralizing/immunology , HIV-1/immunology , Polysaccharides/immunology , Saccharomyces cerevisiae/immunology , Animals , Humans , Neutralization Tests , Rabbits
8.
PLoS Pathog ; 10(2): e1003911, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24516383

ABSTRACT

The Bunyaviridae comprise a large family of RNA viruses with worldwide distribution and includes the pathogenic New World hantavirus, Andes virus (ANDV). Host factors needed for hantavirus entry remain largely enigmatic and therapeutics are unavailable. To identify cellular requirements for ANDV infection, we performed two parallel genetic screens. Analysis of a large library of insertionally mutagenized human haploid cells and a siRNA genomic screen converged on components (SREBP-2, SCAP, S1P and S2P) of the sterol regulatory pathway as critically important for infection by ANDV. The significance of this pathway was confirmed using functionally deficient cells, TALEN-mediated gene disruption, RNA interference and pharmacologic inhibition. Disruption of sterol regulatory complex function impaired ANDV internalization without affecting virus binding. Pharmacologic manipulation of cholesterol levels demonstrated that ANDV entry is sensitive to changes in cellular cholesterol and raises the possibility that clinically approved regulators of sterol synthesis may prove useful for combating ANDV infection.


Subject(s)
Cholesterol/metabolism , Hantavirus Infections/metabolism , Host-Parasite Interactions/physiology , Orthohantavirus/pathogenicity , Virus Internalization , Cell Line , Flow Cytometry , Humans , Microscopy, Confocal , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Signal Transduction/physiology , Sterols/metabolism , Transduction, Genetic , Virus Replication/physiology
9.
Blood ; 123(1): 61-9, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24162716

ABSTRACT

HIV-1 entry into CD4(+) T cells requires binding of the virus to CD4 followed by engagement of either the C-C chemokine receptor 5 (CCR5) or C-X-C chemokine receptor 4 (CXCR4) coreceptor. Pharmacologic blockade or genetic inactivation of either coreceptor protects cells from infection by viruses that exclusively use the targeted coreceptor. We have used zinc-finger nucleases to drive the simultaneous genetic modification of both ccr5 and cxcr4 in primary human CD4(+) T cells. These gene-modified cells proliferated normally and were resistant to both CCR5- and CXCR4-using HIV-1 in vitro. When introduced into a humanized mouse model of HIV-1 infection, these coreceptor negative cells engraft and traffic normally, and are protected from infection with CCR5- and CXCR4-using HIV-1 strains. These data suggest that simultaneous disruption of the HIV coreceptors may provide a useful approach for the long-term, drug-free treatment of established HIV-1 infections.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Endodeoxyribonucleases/metabolism , HIV Infections/immunology , Receptors, CCR5/genetics , Receptors, CXCR4/genetics , Zinc Fingers , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Female , HEK293 Cells , HIV Infections/prevention & control , HIV Infections/therapy , HIV-1 , Humans , Male , Mice , Receptors, Chemokine/metabolism
10.
Cell Host Microbe ; 13(5): 613-623, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23684312

ABSTRACT

Receptor engagement by HIV-1 during host cell entry activates signaling pathways that can reprogram the cell for optimal viral replication. To obtain a global view of the signaling events induced during HIV-1 entry, we conducted a quantitative phosphoproteomics screen of primary human CD4(+) T cells after infection with an HIV-1 strain that engages the receptors CD4 and CXCR4. We quantified 1,757 phosphorylation sites with high stringency. The abundance of 239 phosphorylation sites from 175 genes, including several proteins in pathways known to be impacted by HIV-receptor binding, changed significantly within a minute after HIV-1 exposure. Several previously uncharacterized HIV-1 host factors were also identified and confirmed through RNAi depletion studies. Surprisingly, five serine/arginine-rich (SR) proteins involved in messenger RNA splicing, including the splicing factor SRm300 (SRRM2), were differentially phosophorylated. Mechanistic studies with SRRM2 suggest that HIV-1 modulates host cell alternative splicing machinery during entry in order to facilitate virus replication and release.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV-1/physiology , Host-Pathogen Interactions , Phosphoproteins/analysis , Proteome/analysis , Virus Internalization , Cells, Cultured , Gene Expression Regulation , Humans , Proteomics/methods , RNA Splicing , Virus Release , Virus Replication
11.
Proc Natl Acad Sci U S A ; 110(17): 6626-33, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23542380

ABSTRACT

Defining the virus-host interactions responsible for HIV-1 transmission, including the phenotypic requirements of viruses capable of establishing de novo infections, could be important for AIDS vaccine development. Previous analyses have failed to identify phenotypic properties other than chemokine receptor 5 (CCR5) and CD4+ T-cell tropism that are preferentially associated with viral transmission. However, most of these studies were limited to examining envelope (Env) function in the context of pseudoviruses. Here, we generated infectious molecular clones of transmitted founder (TF; n = 27) and chronic control (CC; n = 14) viruses of subtypes B (n = 18) and C (n = 23) and compared their phenotypic properties in assays specifically designed to probe the earliest stages of HIV-1 infection. We found that TF virions were 1.7-fold more infectious (P = 0.049) and contained 1.9-fold more Env per particle (P = 0.048) compared with CC viruses. TF viruses were also captured by monocyte-derived dendritic cells 1.7-fold more efficiently (P = 0.035) and more readily transferred to CD4+ T cells (P = 0.025). In primary CD4+ T cells, TF and CC viruses replicated with comparable kinetics; however, when propagated in the presence of IFN-α, TF viruses replicated to higher titers than CC viruses. This difference was significant for subtype B (P = 0.000013) but not subtype C (P = 0.53) viruses, possibly reflecting demographic differences of the respective patient cohorts. Together, these data indicate that TF viruses are enriched for higher Env content, enhanced cell-free infectivity, improved dendritic cell interaction, and relative IFN-α resistance. These viral properties, which likely act in concert, should be considered in the development and testing of AIDS vaccines.


Subject(s)
Dendritic Cells/immunology , HIV-1/genetics , Phenotype , Viral Envelope Proteins/metabolism , Virion/pathogenicity , Base Sequence , CD4-Positive T-Lymphocytes/immunology , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , HIV Infections/immunology , HIV Infections/transmission , HIV-1/immunology , Humans , Linear Models , Molecular Sequence Data , Sequence Analysis, DNA
12.
J Virol ; 87(5): 2401-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269796

ABSTRACT

Infection by HIV-1 most often results from the successful transmission and propagation of a single virus variant, termed the transmitted/founder (T/F) virus. Here, we compared the attachment and entry properties of envelope (Env) glycoproteins from T/F and chronic control (CC) viruses. Using a panel of 40 T/F and 47 CC Envs, all derived by single genome amplification, we found that 52% of clade C and B CC Envs exhibited partial resistance to the CCR5 antagonist maraviroc (MVC) on cells expressing high levels of CCR5, while only 15% of T/F Envs exhibited this same property. Moreover, subtle differences in the magnitude with which MVC inhibited infection on cells expressing low levels of CCR5, including primary CD4(+) T cells, were highly predictive of MVC resistance when CCR5 expression levels were high. These results are consistent with previous observations showing a greater sensitivity of T/F Envs to MVC inhibition on cells expressing very high levels of CCR5 and indicate that CC Envs are often capable of recognizing MVC-bound CCR5, albeit inefficiently on cells expressing physiologic levels of CCR5. When CCR5 expression levels are high, this phenotype becomes readily detectable. The utilization of drug-bound CCR5 conformations by many CC Envs was seen with other CCR5 antagonists, with replication-competent viruses, and did not obviously correlate with other phenotypic traits. The striking ability of clade C and B CC Envs to use MVC-bound CCR5 relative to T/F Envs argues that the more promiscuous use of CCR5 by these Env proteins is selected against at the level of virus transmission and is selected for during chronic infection.


Subject(s)
Cyclohexanes/pharmacology , HIV-1/physiology , Receptors, CCR5/metabolism , Triazoles/pharmacology , env Gene Products, Human Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/metabolism , CCR5 Receptor Antagonists , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , HEK293 Cells , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/virology , Humans , Maraviroc , Virus Attachment , Virus Internalization
13.
J Immunol ; 189(8): 3815-21, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22988032

ABSTRACT

CCR5, a cell surface molecule critical for the transmission and spread of HIV-1, is dynamically regulated during T cell activation and differentiation. The molecular mechanism linking T cell activation to modulation of CCR5 expression remains undefined. Kruppel-like factor 2 (KLF2) is a transcription factor that promotes quiescence, survival, and in part by modulating chemokine receptor levels, induces homing to secondary lymphoid organs. Given the relationship between T cell activation and chemokine receptor expression, we tested whether the abundance of KLF2 after T cell activation regulates CCR5 expression and, thus, susceptibility of a T cell to CCR5-dependent HIV-1 strains (R5). We observed a strong correlation between T cell activation, expression of KLF2 and CCR5, and susceptibility to infection. To directly measure how KLF2 affects CCR5 regulation, we introduced small interfering RNA targeting KLF2 expression and demonstrated that reduced KLF2 expression also resulted in less CCR5. Chromatin immunoprecipitation assays identified KLF2 bound to the CCR5 promoter in resting but not CD3/28 activated T cells, suggesting that KLF2 directly regulates CCR5 expression. Introduction of KLF2 under control of a heterologous promoter could restore CCR5 expression and R5 susceptibility to CD3/28 costimulated T cells and some transformed cell lines. Thus, KLF2 is a host factor that modulates CCR5 expression in CD4 T cells and influences susceptibility to R5 infection.


Subject(s)
Genetic Predisposition to Disease , HIV Infections/immunology , HIV-1/immunology , Kruppel-Like Transcription Factors/physiology , Receptors, CCR5/biosynthesis , CCR5 Receptor Antagonists , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line, Transformed , Down-Regulation/genetics , Down-Regulation/immunology , Drug Delivery Systems/methods , Genetic Predisposition to Disease/etiology , HIV Infections/genetics , HIV Infections/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Primary Cell Culture , Protein Binding/genetics , Protein Binding/immunology , RNA, Small Interfering/pharmacology , Receptors, CCR5/genetics , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology
14.
Article in English | MEDLINE | ID: mdl-22908191

ABSTRACT

The first step of the human immunodeficiency virus (HIV) replication cycle-binding and entry into the host cell-plays a major role in determining viral tropism and the ability of HIV to degrade the human immune system. HIV uses a complex series of steps to deliver its genome into the host cell cytoplasm while simultaneously evading the host immune response. To infect cells, the HIV protein envelope (Env) binds to the primary cellular receptor CD4 and then to a cellular coreceptor. This sequential binding triggers fusion of the viral and host cell membranes, initiating infection. Revealing the mechanism of HIV entry has profound implications for viral tropism, transmission, pathogenesis, and therapeutic intervention. Here, we provide an overview into the mechanism of HIV entry, provide historical context to key discoveries, discuss recent advances, and speculate on future directions in the field.


Subject(s)
HIV Infections/immunology , HIV/pathogenicity , env Gene Products, Human Immunodeficiency Virus/metabolism , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Transformation, Viral/physiology , HIV/immunology , HIV Envelope Protein gp120/metabolism , Humans , Integrins/metabolism , Protein Binding , Receptors, CCR4/metabolism , Receptors, CCR5/metabolism , Signal Transduction/physiology , Virion/metabolism
15.
PLoS Pathog ; 8(5): e1002686, 2012.
Article in English | MEDLINE | ID: mdl-22693444

ABSTRACT

Sexual transmission of human immunodeficiency virus type 1 (HIV-1) most often results from productive infection by a single transmitted/founder (T/F) virus, indicating a stringent mucosal bottleneck. Understanding the viral traits that overcome this bottleneck could have important implications for HIV-1 vaccine design and other prevention strategies. Most T/F viruses use CCR5 to infect target cells and some encode envelope glycoproteins (Envs) that contain fewer potential N-linked glycosylation sites and shorter V1/V2 variable loops than Envs from chronic viruses. Moreover, it has been reported that the gp120 subunits of certain transmitted Envs bind to the gut-homing integrin α4ß7, possibly enhancing virus entry and cell-to-cell spread. Here we sought to determine whether subtype C T/F viruses, which are responsible for the majority of new HIV-1 infections worldwide, share biological properties that increase their transmission fitness, including preferential α4ß7 engagement. Using single genome amplification, we generated panels of both T/F (n = 20) and chronic (n = 20) Env constructs as well as full-length T/F (n = 6) and chronic (n = 4) infectious molecular clones (IMCs). We found that T/F and chronic control Envs were indistinguishable in the efficiency with which they used CD4 and CCR5. Both groups of Envs also exhibited the same CD4+ T cell subset tropism and showed similar sensitivity to neutralization by CD4 binding site (CD4bs) antibodies. Finally, saturating concentrations of anti-α4ß7 antibodies failed to inhibit infection and replication of T/F as well as chronic control viruses, although the growth of the tissue culture-adapted strain SF162 was modestly impaired. These results indicate that the population bottleneck associated with mucosal HIV-1 acquisition is not due to the selection of T/F viruses that use α4ß7, CD4 or CCR5 more efficiently.


Subject(s)
CD4 Antigens/metabolism , HIV Infections/transmission , HIV-1/pathogenicity , Integrins/metabolism , Receptors, CCR5/metabolism , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Cloning, Molecular , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/metabolism , HIV-1/immunology , HIV-1/metabolism , Host-Pathogen Interactions , Humans , Integrins/immunology , Mucous Membrane/virology , Neutralization Tests , Viral Tropism , Virus Internalization , Virus Replication
16.
Viruses ; 4(2): 309-24, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22470838

ABSTRACT

Human Immunodeficiency Virus (HIV) entry into target cells is a multi-step process involving binding of the viral glycoprotein, Env, to its receptor CD4 and a coreceptor-either CCR5 or CXCR4. Understanding the means by which HIV enters cells has led to the identification of genetic polymorphisms, such as the 32 base-pair deletion in the ccr5 gene (ccr5∆32) that confers resistance to infection in homozygous individuals, and has also resulted in the development of entry inhibitors-small molecule antagonists that block infection at the entry step. The recent demonstration of long-term control of HIV infection in a leukemic patient following a hematopoietic stem cell transplant using cells from a ccr5∆32 homozygous donor highlights the important role of the HIV entry in maintaining an established infection and has led to a number of attempts to treat HIV infection by genetically modifying the ccr5 gene. In this review, we describe the HIV entry process and provide an overview of the different classes of approved HIV entry inhibitors while highlighting novel genetic strategies aimed at blocking HIV infection at the level of entry.


Subject(s)
HIV Infections/therapy , HIV Infections/virology , HIV/pathogenicity , Virus Internalization , Drug Therapy/methods , Genetic Therapy/methods , HIV Infections/prevention & control , Humans , Immunotherapy/methods , Models, Biological
17.
Adv Exp Med Biol ; 726: 223-42, 2012.
Article in English | MEDLINE | ID: mdl-22297516

ABSTRACT

Human immunodeficiency virus (HIV) entry is a complex and intricate process that facilitates delivery of the viral genome to the host cell. The only viral surface protein, Envelope (Env), is composed of a trimer of gp120 and gp41 heterodimers. It is essentially a fusion machine cloaked in a shroud of carbohydrate structures and variable loops of amino acids that enable it to evade the humoral immune response. For entry to occur gp120 sequentially engages the host protein CD4 and then one of two chemokine coreceptors, either CCR5 or CXCR4. CD4 binding facilitates exposure and formation of the coreceptor-binding site, and coreceptor binding then triggers the membrane fusion machinery in the gp41 subunit. Our understanding of HIV entry has led to the development of successful small molecule inhibitors for the clinical treatment of HIV infection as well as insights into viral tropism and pathogenesis.


Subject(s)
HIV/metabolism , Virus Internalization , CD4 Antigens/metabolism , HIV Infections/virology , Humans , Models, Molecular , Protein Conformation , Receptors, CCR5/chemistry , Receptors, CCR5/metabolism , Receptors, CXCR4/chemistry , Receptors, CXCR4/metabolism , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism
18.
Virology ; 423(2): 134-42, 2012 Feb 20.
Article in English | MEDLINE | ID: mdl-22209230

ABSTRACT

Puumala (PUUV) and Hantaan (HTNV) viruses are hantaviruses within the family Bunyaviridae and associated with Hemorrhagic Fever with Renal Syndrome (HFRS) in humans. Little is known about how these viruses interact with host cells, though pathogenic hantaviruses interact with α(v)ß(3) integrin. To study host cell interactions and rapidly test the ability of antibodies to prevent infection, we produced HTNV and PUUV pseudovirions on a vesicular stomatitis virus (VSV) core. Similar to replication-competent hantaviruses, infection was low-pH-dependent. Despite broad cell tropism, several human T cell lines were poorly permissive to hantavirus pseudovirions, compared to VSV, indicating a relative block to infection at the level of entry. Stable expression of α(v)ß(3) integrin in SupT1 cells did not restore infectivity. Finally, the pseudovirion system provided a rapid, quantitative, and specific method to screen for neutralizing antibodies in immune sera.


Subject(s)
Hantaan virus/physiology , Hemorrhagic Fever with Renal Syndrome/virology , Puumala virus/physiology , Viral Tropism , Virus Cultivation/methods , Animals , Antibodies, Viral/immunology , Cell Line , Hantaan virus/genetics , Hantaan virus/immunology , Hemorrhagic Fever with Renal Syndrome/genetics , Hemorrhagic Fever with Renal Syndrome/immunology , Humans , Mice , Neutralization Tests , Puumala virus/genetics , Puumala virus/immunology
19.
PLoS One ; 6(9): e24918, 2011.
Article in English | MEDLINE | ID: mdl-21949786

ABSTRACT

CXCL12 (SDF-1) is a chemokine that binds to and signals through the seven transmembrane receptor CXCR4. The CXCL12/CXCR4 signaling axis has been implicated in both cancer metastases and human immunodeficiency virus type 1 (HIV-1) infection and a more complete understanding of CXCL12/CXCR4 signaling pathways may support efforts to develop therapeutics for these diseases. Mass spectrometry-based phosphoproteomics has emerged as an important tool in studying signaling networks in an unbiased fashion. We employed stable isotope labeling with amino acids in cell culture (SILAC) quantitative phosphoproteomics to examine the CXCL12/CXCR4 signaling axis in the human lymphoblastic CEM cell line. We quantified 4,074 unique SILAC pairs from 1,673 proteins and 89 phosphopeptides were deemed CXCL12-responsive in biological replicates. Several well established CXCL12-responsive phosphosites such as AKT (pS473) and ERK2 (pY204) were confirmed in our study. We also validated two novel CXCL12-responsive phosphosites, stathmin (pS16) and AKT1S1 (pT246) by Western blot. Pathway analysis and comparisons with other phosphoproteomic datasets revealed that genes from CXCL12-responsive phosphosites are enriched for cellular pathways such as T cell activation, epidermal growth factor and mammalian target of rapamycin (mTOR) signaling, pathways which have previously been linked to CXCL12/CXCR4 signaling. Several of the novel CXCL12-responsive phosphoproteins from our study have also been implicated with cellular migration and HIV-1 infection, thus providing an attractive list of potential targets for the development of cancer metastasis and HIV-1 therapeutics and for furthering our understanding of chemokine signaling regulation by reversible phosphorylation.


Subject(s)
Chemokine CXCL12/pharmacology , Phosphoproteins/metabolism , Proteomics/methods , Signal Transduction/drug effects , Amino Acid Sequence , Blotting, Western , Cell Line , GTP-Binding Proteins/metabolism , Humans , Isotope Labeling , Molecular Sequence Data , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphoproteins/chemistry , Proteome/chemistry , Proteome/metabolism , Reproducibility of Results
20.
J Virol ; 85(20): 10669-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21835785

ABSTRACT

The great majority of human immunodeficiency virus type 1 (HIV-1) strains enter CD4+ target cells by interacting with one of two coreceptors, CCR5 or CXCR4. Here we describe a transmitted/founder (T/F) virus (ZP6248) that was profoundly impaired in its ability to utilize CCR5 and CXCR4 coreceptors on multiple CD4+ cell lines as well as primary human CD4+ T cells and macrophages in vitro yet replicated to very high titers (>80 million RNA copies/ml) in an acutely infected individual. Interestingly, the envelope (Env) glycoprotein of this clade B virus had a rare GPEK sequence in the crown of its third variable loop (V3) rather than the consensus GPGR sequence. Extensive sequencing of sequential plasma samples showed that the GPEK sequence was present in virtually all Envs, including those from the earliest time points after infection. The molecularly cloned (single) T/F virus was able to replicate, albeit poorly, in cells obtained from ccr5Δ32 homozygous donors. The ZP6248 T/F virus could also infect cell lines overexpressing the alternative coreceptors GPR15, APJ, and FPRL-1. A single mutation in the V3 crown sequence (GPEK->GPGK) of ZP6248 restored its infectivity in CCR5+ cells but reduced its ability to replicate in GPR15+ cells, indicating that the V3 crown motif played an important role in usage of this alternative coreceptor. These results suggest that the ZP6248 T/F virus established an acute in vivo infection by using coreceptor(s) other than CCR5 or CXCR4 or that the CCR5 coreceptor existed in an unusual conformation in this individual.


Subject(s)
HIV-1/physiology , Receptors, HIV/metabolism , Viral Tropism , Amino Acid Motifs , Amino Acid Substitution/genetics , Apelin Receptors , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Gene Expression , Humans , Macrophages/virology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , env Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...