Network


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

Hotspot


Dive into the research topics where Dustin Edwards is active.

Publication


Featured researches published by Dustin Edwards.


Viruses | 2011

Orf-I and Orf-II-Encoded Proteins in HTLV-1 Infection and Persistence

Dustin Edwards; Claudio Fenizia; Heather Gold; Maria Fernanda de Castro-Amarante; Cody Buchmann; Cynthia A. Pise-Masison; Genoveffa Franchini

The 3′ end of the human T-cell leukemia/lymphoma virus type-1 (HTLV-1) genome contains four overlapping open reading frames (ORF) that encode regulatory proteins. Here, we review current knowledge of HTLV-1 orf-I and orf-II protein products. Singly spliced mRNA from orf-I encodes p12, which can be proteolytically cleaved to generate p8, while differential splicing of mRNA from orf-II results in production of p13 and p30. These proteins have been demonstrated to modulate transcription, apoptosis, host cell activation and proliferation, virus infectivity and transmission, and host immune responses. Though these proteins are not essential for virus replication in vitro, p8, p12, p13, and p30 have an important role in the establishment and maintenance of HTLV-1 infection in vivo.


PLOS Pathogens | 2014

Co-dependence of HTLV-1 p12 and p8 Functions in Virus Persistence

Cynthia A. Pise-Masison; Maria Fernanda de Castro-Amarante; Yoshimi Enose-Akahata; R. Cody Buchmann; Claudio Fenizia; Robyn Washington Parks; Dustin Edwards; Martina Fiocchi; Luiz Carlos Junior Alcantara; Izabela Bialuk; Jhanelle Graham; Jean-Claude Walser; Katherine McKinnon; Bernardo Galvão-Castro; Antoine Gessain; David Venzon; Steven Jacobson; Genoveffa Franchini

HTLV-1 orf-I is linked to immune evasion, viral replication and persistence. Examining the orf-I sequence of 160 HTLV-1-infected individuals; we found polymorphism of orf-I that alters the relative amounts of p12 and its cleavage product p8. Three groups were identified on the basis of p12 and p8 expression: predominantly p12, predominantly p8 and balanced expression of p12 and p8. We found a significant association between balanced expression of p12 and p8 with high viral DNA loads, a correlate of disease development. To determine the individual roles of p12 and p8 in viral persistence, we constructed infectious molecular clones expressing p12 and p8 (D26), predominantly p12 (G29S) or predominantly p8 (N26). As we previously showed, cells expressing N26 had a higher level of virus transmission in vitro. However, when inoculated into Rhesus macaques, cells producing N26 virus caused only a partial seroconversion in 3 of 4 animals and only 1 of those animals was HTLV-1 DNA positive by PCR. None of the animals exposed to G29S virus seroconverted or had detectable viral DNA. In contrast, 3 of 4 animals exposed to D26 virus seroconverted and were HTLV-1 positive by PCR. In vitro studies in THP-1 cells suggested that expression of p8 was sufficient for productive infection of monocytes. Since orf-I plays a role in T-cell activation and recognition; we compared the CTL response elicited by CD4+ T-cells infected with the different HTLV-1 clones. Although supernatant p19 levels and viral DNA loads for all four infected lines were similar, a significant difference in Tax-specific HLA.A2-restricted killing was observed. Cells infected with Orf-I-knockout virus (12KO), G29S or N26 were killed by CTLs, whereas cells infected with D26 virus were resistant to CTL killing. These results indicate that efficient viral persistence and spread require the combined functions of p12 and p8.


Journal of Virology | 2014

Human T-Cell Leukemia/Lymphoma Virus Type 1 p30, but Not p12/p8, Counteracts Toll-Like Receptor 3 (TLR3) and TLR4 Signaling in Human Monocytes and Dendritic Cells

Claudio Fenizia; Martina Fiocchi; Kathryn S. Jones; Robyn Washington Parks; Michele Ceribelli; Sébastien Alain Chevalier; Dustin Edwards; Francis W. Ruscetti; Cynthia A. Pise-Masison; Genoveffa Franchini

ABSTRACT The human T-cell leukemia/lymphoma virus type 1 (HTLV-1) p30 protein, essential for virus infectivity in vivo, is required for efficient infection of human dendritic cells (DCs) but not B and T cells in vitro. We used a human monocytic cell line, THP-1, and dendritic cells to study the mechanism of p30 and p12/p8 requirements in these cell types. p30 inhibited the expression of interferon (IFN)-responsive genes (ISG) following stimulation by lipopolysaccharide (LPS) of Toll-like receptor 4 (TLR4) and by poly(I·C) of TLR3 but not of TLR7/8 with imiquimod. Results with THP-1 mirrored those for ex vivo human primary monocytes and monocyte-derived dendritic cells (Mo-mDC). The effect of p30 on TLR signaling was also demonstrated by ablating its expression within a molecular clone of HTLV-1. HTLV-1 infection of monocytes inhibited TLR3- and TLR4-induced ISG expression by 50 to 90% depending on the genes, whereas the isogenic clone p30 knockout virus was less effective at inhibiting TLR3 and TRL4 signaling and displayed lower infectivity. Viral expression and inhibition of ISG transcription was, however, rescued by restoration of p30 expression. A chromatin immunoprecipitation assay demonstrated that p30 inhibits initiation and elongation of PU.1-dependent transcription of IFN-α1, IFN-β, and TLR4 genes upon TLR stimulation. In contrast, experiments conducted with p12/p8 did not demonstrate an effect on ISG expression. These results provide a mechanistic explanation of the requirement of p30 for HTLV-1 infectivity in vivo, suggest that dampening interferon responses in monocytes and DCs is specific for p30, and represent an essential early step for permissive HTLV-1 infection and persistence.


PLOS ONE | 2012

A Public HTLV-1 Molecular Epidemiology Database for Sequence Management and Data Mining

Thessika Hialla Almeida Araújo; Leandro Inacio Souza-Brito; Pieter Libin; Koen Deforche; Dustin Edwards; Antonio Eduardo de Albuquerque-Junior; Anne-Mieke Vandamme; Bernardo Galvão-Castro; Luiz Carlos Junior Alcantara

Background It is estimated that 15 to 20 million people are infected with the human T-cell lymphotropic virus type 1 (HTLV-1). At present, there are more than 2,000 unique HTLV-1 isolate sequences published. A central database to aggregate sequence information from a range of epidemiological aspects including HTLV-1 infections, pathogenesis, origins, and evolutionary dynamics would be useful to scientists and physicians worldwide. Described here, we have developed a database that collects and annotates sequence data and can be accessed through a user-friendly search interface. The HTLV-1 Molecular Epidemiology Database website is available at http://htlv1db.bahia.fiocruz.br/. Methodology/Principal Findings All data was obtained from publications available at GenBank or through contact with the authors. The database was developed using Apache Webserver 2.1.6 and SGBD MySQL. The webpage interfaces were developed in HTML and sever-side scripting written in PHP. The HTLV-1 Molecular Epidemiology Database is hosted on the Gonçalo Moniz/FIOCRUZ Research Center server. There are currently 2,457 registered sequences with 2,024 (82.37%) of those sequences representing unique isolates. Of these sequences, 803 (39.67%) contain information about clinical status (TSP/HAM, 17.19%; ATL, 7.41%; asymptomatic, 12.89%; other diseases, 2.17%; and no information, 60.32%). Further, 7.26% of sequences contain information on patient gender while 5.23% of sequences provide the age of the patient. Conclusions/Significance The HTLV-1 Molecular Epidemiology Database retrieves and stores annotated HTLV-1 proviral sequences from clinical, epidemiological, and geographical studies. The collected sequences and related information are now accessible on a publically available and user-friendly website. This open-access database will support clinical research and vaccine development related to viral genotype.


Journal of Virology | 2008

Human T-Cell Leukemia Virus Type 1 Tax Relieves Repression of Proliferating Cell Nuclear Antigen Gene Expression

Dustin Edwards; Susan J. Marriott

ABSTRACT Human T-cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia. The transforming ability of Tax, the viral oncoprotein, is believed to depend on interactions with cell cycle regulators and on transactivation of genes that control cellular proliferation, including proliferating cell nuclear antigen (PCNA), a cofactor associated with DNA replication and repair. Tax associates with cellular transcription factors to alter their affinity for cognate DNA elements, leading to increased or decreased transcription from that promoter. Although it has been demonstrated that Tax transactivates the PCNA promoter, the mechanism of transcriptional activation is unknown. Here we report a cellular complex that binds specifically to a novel site within the minimal Tax-responsive element of the TATAA-less PCNA promoter. Mutation at this binding site or Tax expression inhibited complex formation and increased promoter activity, suggesting that the complex is a transcriptional repressor. The activation of PCNA gene expression by Tax and consequential decrease in nucleotide excision repair mediated by PCNA overexpression could contribute to the reduced DNA repair capacity and genomic instability observed in HTLV-1-infected cells.


Journal of Virology | 2014

Palmitoylation and p8-Mediated Human T-Cell Leukemia Virus Type 1 Transmission

Dustin Edwards; Risaku Fukumoto; Maria Fernanda de Castro-Amarante; Luiz Carlos Junior Alcantara; Bernardo Galvão-Castro; Robyn Washington Parks; Cynthia A. Pise-Masison; Genoveffa Franchini

ABSTRACT The orf-I gene of human T-cell leukemia type 1 (HTLV-1) encodes p8 and p12 and has a conserved cysteine at position 39. p8 and p12 form disulfide-linked dimers, and only the monomeric forms of p8 and p12 are palmitoylated. Mutation of cysteine 39 to alanine (C39A) abrogated dimerization and palmitoylation of both proteins. However, the ability of p8 to localize to the cell surface and to increase cell adhesion and viral transmission was not affected by the C39A mutation.


Viruses | 2011

Inhibition of Geranylgeranyl Transferase-I Decreases Cell Viability of HTLV-1-Transformed Cells

Dustin Edwards; Katherine McKinnon; Claudio Fenizia; Kyung-Jin Jung; John N. Brady; Cynthia A. Pise-Masison

Human T-cell leukemia virus type-1 (HTLV-1) is the etiological agent of adult T-cell leukemia (ATL), an aggressive and highly chemoresistant malignancy. Rho family GTPases regulate multiple signaling pathways in tumorigenesis: cytoskeletal organization, transcription, cell cycle progression, and cell proliferation. Geranylgeranylation of Rho family GTPases is essential for cell membrane localization and activation of these proteins. It is currently unknown whether HTLV-1-transformed cells are preferentially sensitive to geranylgeranylation inhibitors, such as GGTI-298. In this report, we demonstrate that GGTI-298 decreased cell viability and induced G2/M phase accumulation of HTLV-1-transformed cells, independent of p53 reactivation. HTLV-1-LTR transcriptional activity was inhibited and Tax protein levels decreased following treatment with GGTI-298. Furthermore, GGTI-298 decreased activation of NF-κB, a downstream target of Rho family GTPases. These studies suggest that protein geranylgeranylation contributes to dysregulation of cell survival pathways in HTLV-1-transformed cells.


Retrovirology | 2014

Both orf-I isoforms, p12 and p8, are required for efficient HTLV-1 infection and persistence

Cynthia A. Pise-Masison; Maria Fernanda de Castro-Amarante; Cody Buchmann; Yoshimi Enoso-Akahata; Claudio Fenezia; Dustin Edwards; Luiz Carlos Junior Alcantara; Izabela Bialuk; Steven Jacobson; Bernardo Galvão-Castro; Antoine Gessain; Genoveffa Franchini

HTLV-1 orf-I is important for immune evasion, viral replication and persistence. Here, we analyzed proviral sequences isolated from ex vivo PBMC samples of 163 HTLV-1-infected patients (85 carriers, 78 HAM/TSP). Of the 5000 sequences analyzed, orf-I expression was consistently maintained. While the highest percentage of isolates express both p12 and p8 (69%), the mutants that express mainly p8 (7%) or mainly p12 (24%) were highly correlated with decreased proviral loads. To determine the individual role of p12 and p8 in viral persistence, we constructed infectious molecular clones expressing either p12(G29S) or p8(N26) and established producer cell lines used to infect macaques. Three of four macaques infected with WT-HTLV-1 tested positive by PCR and were seropositive. Only one of the macaques infected with N26-HTLV-1 tested positive by PCR and two macaques were seropositive. None of the 8 animals infected with G29S-HTLV-1 tested PCR positive or seroconverted. These results suggest that expression of both p12 and p8 are critical for viral persistence. Since orf-I plays a role in T-cell activation and recognition, we compared the CTL responses elicited by CD4+ T-cell infected with the different HTLV-1 clones. Although the supernatant p19 levels and proviral loads for all four infected lines were similar, a significant difference in Tax-specific CTL killing was observed. Cells infected with orf-I-knockout virus, p12-G29S or p8-N26 were killed whereas cells infected with WT virus were not killed by Tax-specific CTLs. These results indicate that efficient viral persistence and spread requires expression of both p12 and p8.


Retrovirology | 2011

Role of dimerization and palmitoylation on the function of HTLV-1 p12 and p8

Dustin Edwards; Risaku Fukumoto; Nancy Van Prooyen; Heather Gold; Maria Fernanda de Castro-Amarante; Luiz Carlos Junior Alcantara; Genoveffa Franchini

HTLV-1 p12 is an endoplasmic reticulum resident protein that is cleaved to generate p8, which traffics to the cell surface. p12 promotes T-cell activation by increasing NFAT activity while p8 induces T-cell anergy and enhances virus transmission. We have previously demonstrated that p12 and p8 coimmunoprecipitate. Though the dimerization domain of p12 and p8 is unknown, these proteins contain a single cysteine residue which may form intermolecular disulfide bonds. We have also determined that p8 localizes to membrane lipid rafts. Of importance, palmitoylation increases the hydrophobicity of proteins to target them to lipid rafts. As palmitoylation occurs on cysteine residues, we hypothesize that this post-translational modification regulates p12 and p8 localization, dimerization, and function. In this study, we have demonstrated that wildtype p12 and p8 formed hetero- and homodimers while a mutation at the cysteine residue (C39A) inhibited dimer formation and that monomeric wildtype p12 and p8, but not the C39A mutant, are pamitoylated. Immunofluorescence analysis showed that wildtype p8 localized at the cell surface while C39A p8 did not. This result suggests that p8 homodimerization or palmitoylation regulates p8 localization. We also analyzed ex vivo DNA samples from HTLV-1-infected individuals and found DNA polymorphisms at position 39. These naturally occurring polymorphisms affected dimerization and localization of p8. Currently, we are investigating whether mutation at C39 affects NFAT activation, virus transmission, or proviral load. Determining the mechanism by which p12 and p8 localization, dimerization, and functions are regulated will add to our understanding of HTLV-1 pathogenesis.


Journal of Virology | 2015

Discovery and Characterization of Auxiliary Proteins Encoded by Type 3 Simian T-Cell Lymphotropic Viruses

Jocelyn Turpin; Chloé Journo; Nga Ling Ko; Flore Sinet; Alexandre Carpentier; Amandine Galioot; Dustin Edwards; Anne-Mieke Vandamme; Louis Gazzolo; Madeleine Duc Dodon; Antoine Gessain; Fatah Kashanchi; Ivan Balansard; Romain Lacoste; Renaud Mahieux

ABSTRACT Human T-cell lymphotropic virus type 1 (HTLV-1) and HTLV-2 encode auxiliary proteins that play important roles in viral replication, viral latency, and immune escape. The presence of auxiliary protein-encoding open reading frames (ORFs) in HTLV-3, the latest HTLV to be discovered, is unknown. Simian T-cell lymphotropic virus type 3 (STLV-3) is almost identical to HTLV-3. Given the lack of HTLV-3-infected cell lines, we took advantage of STLV-3-infected cells and of an STLV-3 molecular clone to search for the presence of auxiliary transcripts. Using reverse transcriptase PCR (RT-PCR), we first uncovered the presence of three unknown viral mRNAs encoding putative proteins of 5, 8, and 9 kDa and confirmed the presence of the previously reported RorfII transcript. The existence of these viral mRNAs was confirmed by using splice site-specific RT-PCR with ex vivo samples. We showed that p5 is distributed throughout the cell and does not colocalize with a specific organelle. The p9 localization is similar to that of HTLV-1 p12 and induced a strong decrease in the calreticulin signal, similarly to HTLV-1 p12. Although p8, RorfII, and Rex-3 share an N-terminal sequence that is predicted to contain a nucleolar localization signal (NoLS), only p8 is found in the nucleolus. The p8 location in the nucleolus is linked to a bipartite NoLS. p8 and, to a lesser extent, p9 repressed viral expression but did not alter Rex-3-dependent mRNA export. Using a transformation assay, we finally showed that none of the STLV-3 auxiliary proteins had the ability to induce colony formation, while both Tax-3 and antisense protein of HTLV-3 (APH-3) promoted cellular transformation. Altogether, these results complete the characterization of the newly described primate T-lymphotropic virus type 3 (PTLV-3). IMPORTANCE Together with their simian counterparts, HTLVs form the primate T-lymphotropic viruses. HTLVs arose from interspecies transmission between nonhuman primates and humans. HTLV-1 and HTLV-2 encode auxiliary proteins that play important roles in viral replication, viral latency, and immune escape. The presence of ORFs encoding auxiliary proteins in HTLV-3 or STLV-3 genomes was unknown. Using in silico analyses, ex vivo samples, or in vitro experiments, we have uncovered the presence of 3 previously unknown viral mRNAs encoding putative proteins and confirmed the presence of a previously reported viral transcript. We characterized the intracellular localization of the four proteins. We showed that two of these proteins repress viral expression but that none of them have the ability to induce colony formation. However, both Tax and the antisense protein APH-3 promote cell transformation. Our results allowed us to characterize 4 new retroviral proteins for the first time.

Collaboration


Dive into the Dustin Edwards's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Genoveffa Franchini

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Claudio Fenizia

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cody Buchmann

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Heather Gold

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge