Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Mol Ther ; 29(3): 1186-1198, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33278563

ABSTRACT

Historically poor clinical results of tumor vaccines have been attributed to weakly immunogenic antigen targets, limited specificity, and vaccine platforms that fail to induce high-quality polyfunctional T cells, central to mediating cellular immunity. We show here that the combination of antigen selection, construct design, and a robust vaccine platform based on the Synthetically Modified Alpha Replicon RNA Technology (SMARRT), a self-replicating RNA, leads to control of tumor growth in mice. Therapeutic immunization with SMARRT replicon-based vaccines expressing tumor-specific neoantigens or tumor-associated antigen were able to generate polyfunctional CD4+ and CD8+ T cell responses in mice. Additionally, checkpoint inhibitors, or co-administration of cytokine also expressed from the SMARRT platform, synergized to enhance responses further. Lastly, SMARRT-based immunization of non-human primates was able to elicit high-quality T cell responses, demonstrating translatability and clinical feasibility of synthetic replicon technology for therapeutic oncology vaccines.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Colonic Neoplasms/therapy , Immunity, Cellular/immunology , Replicon , Animals , Cancer Vaccines/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Primates , Tumor Cells, Cultured , Vaccination
2.
Sci Adv ; 6(45)2020 11.
Article in English | MEDLINE | ID: mdl-33148638

ABSTRACT

Zika virus (ZIKV) is associated with congenital malformations in infants born to infected mothers, and with Guillain-Barré syndrome in infected adults. Development of ZIKV vaccines has focused predominantly on the induction of neutralizing antibodies, although a suboptimal antibody response may theoretically enhance disease severity through antibody-dependent enhancement (ADE). Here, we report induction of a protective anti-ZIKV CD8+ T cell response in the HLA-B*0702 Ifnar1-/- transgenic mice using an alphavirus-based replicon RNA vaccine expressing ZIKV nonstructural protein NS3, a potent T cell antigen. The NS3 vaccine did not induce a neutralizing antibody response but elicited polyfunctional CD8+ T cells that were necessary and sufficient for preventing death in lethally infected adult mice and fetal growth restriction in infected pregnant mice. These data identify CD8+ T cells as the major mediators of ZIKV NS3 vaccine-induced protection and suggest a new strategy to develop safe and effective anti-flavivirus vaccines.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Antibodies, Neutralizing , CD8-Positive T-Lymphocytes , Humans , Mice , Vaccines, Synthetic , mRNA Vaccines
3.
J Adolesc Adult Lit ; 64(1): 11-17, 2020.
Article in English | MEDLINE | ID: mdl-32834710

ABSTRACT

In this commentary, the authors move beyond digital literacy and take up the question of what digital citizenship means and looks like in the context of the COVID-19 pandemic. To engage with questions of ethical practice, the authors begin with the International Society for Technology in Education framework for digital citizenship. They expand on these standards to argue for an awareness of the ethical questions facing citizens online that are difficult to encompass as a set of skills or competencies. The authors then take these considerations into a set of practical steps for teachers to nurture participatory and social justice-oriented digital citizenship as part of the curriculum. The authors conclude by noting the digital divide and social inequities that have been highlighted by the current crisis.

4.
PLoS Genet ; 6(12): e1001258, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21203491

ABSTRACT

The transcription factor ATF2 has been shown to attenuate melanoma susceptibility to apoptosis and to promote its ability to form tumors in xenograft models. To directly assess ATF2's role in melanoma development, we crossed a mouse melanoma model (Nras(Q61K)::Ink4a⁻/⁻) with mice expressing a transcriptionally inactive form of ATF2 in melanocytes. In contrast to 7/21 of the Nras(Q61K)::Ink4a⁻/⁻ mice, only 1/21 mice expressing mutant ATF2 in melanocytes developed melanoma. Gene expression profiling identified higher MITF expression in primary melanocytes expressing transcriptionally inactive ATF2. MITF downregulation by ATF2 was confirmed in the skin of Atf2⁻/⁻ mice, in primary human melanocytes, and in 50% of human melanoma cell lines. Inhibition of MITF transcription by MITF was shown to be mediated by ATF2-JunB-dependent suppression of SOX10 transcription. Remarkably, oncogenic BRAF (V600E)-dependent focus formation of melanocytes on soft agar was inhibited by ATF2 knockdown and partially rescued upon shMITF co-expression. On melanoma tissue microarrays, a high nuclear ATF2 to MITF ratio in primary specimens was associated with metastatic disease and poor prognosis. Our findings establish the importance of transcriptionally active ATF2 in melanoma development through fine-tuning of MITF expression.


Subject(s)
Activating Transcription Factor 2/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Activating Transcription Factor 2/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Down-Regulation , Female , Humans , Male , Melanocytes/metabolism , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmia-Associated Transcription Factor/metabolism , Skin/metabolism , Skin/pathology
5.
PLoS Pathog ; 5(10): e1000613, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19798433

ABSTRACT

Natural killer (NK) cells are stimulated by ligands on virus-infected cells. We have recently demonstrated that NK cells respond to human immunodeficiency virus type-1 (HIV-1)-infected autologous T-cells, in part, through the recognition of ligands for the NK cell activating receptor NKG2D on the surface of the infected cells. Uninfected primary CD4(pos) T-cell blasts express little, if any, NKG2D ligands. In the present study we determined the mechanism through which ligands for NKG2D are induced on HIV-1-infected cells. Our studies reveal that expression of vpr is necessary and sufficient to elicit the expression of NKG2D ligands in the context of HIV-1 infection. Vpr specifically induces surface expression of the unique-long 16 binding proteins (ULBP)-1 and ULBP-2, but not ULBP-3, MHC class I-related chain molecules (MIC)-A or MIC-B. In these studies we also demonstrated that Vpr increases the level of ULBP-1 and ULBP-2 mRNA in primary CD4(pos) T-cell blasts. The presence of ULBP-1 and ULBP-2 on HIV-1 infected cells is dependent on the ability of Vpr to associate with a protein complex know as Cullin 4a (Cul4a)/damaged DNA binding protein 1 (DDB1) and Cul4a-associated factor-1(DCAF-1) E3 ubiquitin ligase (Cul4a(DCAF-1)). ULBP-1 and -2 expression by Vpr is also dependent on activation of the DNA damage sensor, ataxia telangiectasia and rad-3-related kinase (ATR). When T-cell blasts are infected with a vpr-deficient HIV-1, NK cells are impaired in killing the infected cells. Thus, HIV-1 Vpr actively triggers the expression of the ligands to the NK cell activation receptor.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Products, vpr/metabolism , HIV Infections/immunology , Killer Cells, Natural/immunology , Protein Serine-Threonine Kinases/metabolism , Ataxia Telangiectasia Mutated Proteins , CD4-Positive T-Lymphocytes/virology , Cullin Proteins/biosynthesis , DNA Damage , Flow Cytometry , GPI-Linked Proteins , Gene Expression , Gene Expression Regulation, Viral , Gene Products, vpr/genetics , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/immunology , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins , Membrane Proteins/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/virology , Ubiquitin-Protein Ligases/metabolism
6.
J Virol ; 82(18): 9265-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18596088

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Vif recruits a Cullin 5 ubiquitin ligase that targets APOBEC3 proteins for degradation. Recently, Vif has also been shown to induce cell cycle disturbance in G(2). We show that in contrast to the expression of Vpr, the expression of Vif does not preclude cell division, and therefore, Vif causes delay and not arrest in G(2). We also demonstrate that the interaction of Vif with the ubiquitin ligase is required for cell cycle disruption, as was previously shown for HIV-1 Vpr. The presence of APOBEC3 D/E, F, and G had no influence on Vif-induced alteration of the cell cycle. We conclude that cell cycle delay by Vif is a result of ubiquitination and degradation of a cellular protein that is different from the known APOBEC3 family members.


Subject(s)
Cytosine Deaminase/metabolism , G2 Phase/drug effects , Gene Products, vif/pharmacology , HIV-1/pathogenicity , Ubiquitin-Protein Ligases/metabolism , APOBEC Deaminases , Cell Cycle/drug effects , Cell Line , Cytidine Deaminase , Gene Products, vif/metabolism , HIV-1/genetics , HIV-1/metabolism , Humans , Mutation , Transfection
8.
Virol J ; 4: 57, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17559673

ABSTRACT

HIV-1 Vpr is a viral accessory protein that activates ATR through the induction of DNA replication stress. ATR activation results in cell cycle arrest in G2 and induction of apoptosis. In the present study, we investigate the role of the ubiquitin/proteasome system (UPS) in the above activity of Vpr. We report that the general function of the UPS is required for Vpr to induce G2 checkpoint activation, as incubation of Vpr-expressing cells with proteasome inhibitors abolishes this effect. We further investigated in detail the specific E3 ubiquitin ligase subunits that Vpr manipulates. We found that Vpr binds to the DCAF1 subunit of a cullin 4a/DDB1 E3 ubiquitin ligase. The carboxy-terminal domain Vpr(R80A) mutant, which is able to bind DCAF1, is inactive in checkpoint activation and has dominant-negative character. In contrast, the mutation Q65R, in the leucine-rich domain of Vpr that mediates DCAF1 binding, results in an inactive Vpr devoid of dominant negative behavior. Thus, the interaction of Vpr with DCAF1 is required, but not sufficient, for Vpr to cause G2 arrest. We propose that Vpr recruits, through its carboxy terminal domain, an unknown cellular factor that is required for G2-to-M transition. Recruitment of this factor leads to its ubiquitination and degradation, resulting in failure to enter mitosis.


Subject(s)
Gene Products, vpr/physiology , HIV-1/physiology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Cell Line , G2 Phase , Gene Products, vpr/isolation & purification , HIV-1/drug effects , HeLa Cells/cytology , HeLa Cells/physiology , HeLa Cells/virology , Humans , Kidney , Oligopeptides/pharmacology , RNA, Small Interfering/genetics , RNA, Viral/genetics , Transfection , Virus Replication , vpr Gene Products, Human Immunodeficiency Virus
9.
PLoS Pathog ; 2(12): e127, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17140287

ABSTRACT

The HIV-1 accessory protein viral protein R (Vpr) causes G2 arrest and apoptosis in infected cells. We previously identified the DNA damage-signaling protein ATR as the cellular factor that mediates Vpr-induced G2 arrest and apoptosis. Here, we examine the mechanism of induction of apoptosis by Vpr and how it relates to induction of G2 arrest. We find that entry into G2 is a requirement for Vpr to induce apoptosis. We investigated the role of the mitochondrial permeability transition pore by knockdown of its essential component, the adenine nucleotide translocator. We found that Vpr-induced apoptosis was unaffected by knockdown of ANT. Instead, apoptosis is triggered through a different mitochondrial pore protein, Bax. In support of the idea that checkpoint activation and apoptosis induction are functionally linked, we show that Bax activation by Vpr was ablated when ATR or GADD45alpha was knocked down. Certain mutants of Vpr, such as R77Q and I74A, identified in long-term nonprogressors, have been proposed to inefficiently induce apoptosis while activating the G2 checkpoint in a normal manner. We tested the in vitro phenotypes of these mutants and found that their abilities to induce apoptosis and G2 arrest are indistinguishable from those of HIV-1NL4-3 vpr, providing additional support to the idea that G2 arrest and apoptosis induction are mechanistically linked.


Subject(s)
Adenine Nucleotide Translocator 1/physiology , Apoptosis/physiology , Cell Cycle/physiology , G2 Phase/physiology , Gene Products, vpr/physiology , bcl-2-Associated X Protein/physiology , Adenine Nucleotide Translocator 1/genetics , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins , Ataxia Telangiectasia Mutated Proteins , CD4-Positive T-Lymphocytes/cytology , Caspases/physiology , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Line , Down-Regulation , Gene Expression Regulation, Viral , Gene Products, vpr/genetics , HIV-1/genetics , HIV-1/pathogenicity , HIV-1/physiology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Mutagens/pharmacology , Mutation/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , bcl-2-Associated X Protein/genetics , vpr Gene Products, Human Immunodeficiency Virus
10.
J Virol ; 80(8): 3694-700, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16571786

ABSTRACT

Cyclophilin A (CypA) is a member of a family of cellular proteins that share a peptidyl prolyl cis-trans isomerase (PPIase) activity. CypA was previously reported to be required for the biochemical stability and function (specifically, induction of G2 arrest) of the human immunodeficiency virus type 1 (HIV-1) protein R (Vpr). In the present study, we examine the role of the Vpr-CypA interaction on Vpr-induced G2 arrest. We find that Vpr coimmunoprecipitates with CypA and that this interaction is disrupted by substitution of proline-35 of Vpr as well as incubation with the CypA inhibitor cyclosporine A (CsA). Surprisingly, the presence of CypA or its binding to Vpr is dispensable for the ability of Vpr to induce G2 arrest. Vpr expression in CypA-/- cells leads to induction of G2 arrest in a manner that is indistinguishable from that in CypA+ cells. CsA abolished CypA-Vpr binding but had no effect on induction of G2 arrest or Vpr steady-state levels. In view of these results, we propose that the interaction with CypA is independent of the ability of Vpr to induce cell cycle arrest. The interaction between Vpr and CypA is intriguing, and further studies should examine its potential effects on other functions of Vpr.


Subject(s)
Cyclophilin A/physiology , G2 Phase , Gene Products, vpr/physiology , Cell Line , Cyclophilin A/antagonists & inhibitors , Humans , Phenotype
11.
13.
J Virol ; 79(3): 1389-96, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15650165

ABSTRACT

Integration into the host cell DNA is an essential part of the retroviral life cycle and is required for the productive replication of a retrovirus. Retroviral integration involves cleavage of the host DNA and insertion of the viral DNA, forming an integration intermediate that contains two gaps, each with a viral 5' flap. The flaps are then removed, and the gap is filled by as yet unidentified nuclease and polymerase activities. It is thought that repair of these gaps flanking the site of retroviral integration is achieved by host DNA repair machinery. The ATM and Rad3-related protein (ATR) is a member of the phosphatidylinositol 3 kinase-related family of protein kinases that play a major role in sensing and triggering repair of DNA lesions in mammalian cells. In an effort to examine the role of ATR in retroviral integration, we used RNA interference to selectively downregulate ATR and measured integration efficiency. In addition, we examined the possible role that Vpr may play in enhancing integration and, in particular, whether activation of ATR by Vpr (Roshal et al., J. Biol. Chem. 278:25879-25886, 2003) will favor human immunodeficiency virus type 1 integration. We conclude that cells in which ATR has been depleted are competent for retroviral integration. We also conclude that the presence of Vpr as a virion-bound protein does not enhance integration of a lentivirus vector in dividing cells.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Retroviridae/genetics , Virus Integration , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line , DNA, Viral/genetics , Down-Regulation , Gene Products, vpr/genetics , Gene Products, vpr/metabolism , Genetic Vectors , HIV-1/genetics , HeLa Cells , Humans , Mice , Protein Serine-Threonine Kinases/genetics , RNA Interference , Transduction, Genetic , vpr Gene Products, Human Immunodeficiency Virus
15.
Todays FDA ; 17(9): 24-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16639824
16.
Mol Cell Biol ; 24(21): 9286-94, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15485898

ABSTRACT

Eukaryotic cells have evolved a complex mechanism for sensing DNA damage during genome replication. Activation of this pathway prevents entry into mitosis to allow for either DNA repair or, in the event of irreparable damage, commitment to apoptosis. Under conditions of replication stress, the damage signal is initiated by the ataxia-telangiectasia-mutated and Rad3-related kinase ATR. We recently demonstrated that the human immunodeficiency virus type 1 (HIV-1) gene product viral protein R (Vpr) arrests infected cells in the G(2) phase via the activation of ATR. In the present study, we show that the activation of ATR by Vpr is analogous to activation by certain genotoxic agents, both mechanistically and in its downstream consequences. Specifically, we show a requirement for Rad17 and Hus1 to induce G(2) arrest as well as Vpr-induced phosphorylation of histone 2A variant X (H2AX) and formation of nuclear foci containing H2AX and breast cancer susceptibility protein 1. These results demonstrate that G(2) arrest mediated by the HIV-1 gene product Vpr utilizes the cellular signaling pathway whose physiological function is to recognize replication stress. These findings should contribute to a greater understanding of how HIV-1 manipulates the CD4(+)-lymphocyte cell cycle and apoptosis induction in the progressive CD4(+)-lymphocyte depletion characteristic of HIV-1 pathogenesis.


Subject(s)
BRCA1 Protein/metabolism , Cell Cycle Proteins/metabolism , G2 Phase , Gene Products, vpr/metabolism , HIV-1/physiology , Histones/metabolism , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein/genetics , CD4 Antigens/metabolism , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , DNA-Binding Proteins , Gene Expression Regulation , Gene Products, vpr/genetics , Histones/genetics , Humans , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Schizosaccharomyces pombe Proteins , Thymus Gland/metabolism , Thymus Gland/pathology , Tumor Suppressor Proteins , vpr Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL