Network


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

Hotspot


Dive into the research topics where Thomas J. Gardner is active.

Publication


Featured researches published by Thomas J. Gardner.


Science Translational Medicine | 2013

Neutralizing antibodies against previously encountered influenza virus strains increase over time: a longitudinal analysis.

Matthew S. Miller; Thomas J. Gardner; Florian Krammer; Lauren C. Aguado; Domenico Tortorella; Christopher F. Basler; Peter Palese

Antigenic variation promotes neutralizing antibodies to both seasonal and pandemic influenza A strains in humans. Only a Matter of Time In the fast-paced world of scientific research, most conclusions are drawn on few—or single—time points or with animal models that may accelerate—but not always accurately reproduce—disease progression. Longitudinal analyses are rare. Indeed, this is the case in influenza virus research, where it is thought that previous infections influence the outcome of subsequent infections, but little is actually known about how sequential exposures to antigenically diverse viruses affect the antibody response to influenza A viruses. Now, Miller et al. use samples obtained over a 20-year period from 40 individuals involved in the Framingham Heart Study to look at antibody titers to seasonal and pandemic influenza strains over time. The authors find longitudinal increases in neutralizing antibodies to previously encountered seasonal and pandemic flu strains. This increase was not age-dependent because it was observed against strains encountered later in life as well as in earlier exposures. Titers to the more conserved hemagglutinin stalk domain increased modestly as well, but no neutralizing antibody titer increase was observed against a more antigenically stable virus (human cytomegalovirus). These results suggest that antigenic variation may drive the hierarchical humoral immune response to influenza strains. The contribution of antigenic variation to antibody titers to the conserved stalk region supports the pursuit of vaccine strategies that increase exposure to antigenically diverse strains of influenza. Antigenic diversity shapes immunity in distinct and unexpected ways. This is particularly true of the humoral response generated against influenza A viruses. Although it is known that immunological memory developed against previously encountered influenza A virus strains affects the outcome of subsequent infections, exactly how sequential exposures to antigenically variant viruses shape the humoral immune response in humans remains poorly understood. To address this important question, we performed a longitudinal analysis of antibody titers against various pandemic and seasonal strains of influenza virus spanning a 20-year period (1987 to 2008) with samples from 40 individuals (birth dates, 1917 to 1952) obtained from the Framingham Heart Study. Longitudinal increases in neutralizing antibody titers were observed against previously encountered pandemic H2N2, H3N2, and H1N1 influenza A virus strains. Antibody titers against seasonal strains encountered later in life also increased longitudinally at a rate similar to that against their pandemic predecessors. Titers of cross-reactive antibodies specific to the hemagglutinin stalk domain were also investigated because they are influenced by exposure to antigenically diverse influenza A viruses. These titers rose modestly over time, even in the absence of major antigenic shifts. No sustained increase in neutralizing antibody titers against an antigenically more stable virus (human cytomegalovirus) was observed. The results herein describe a role for antigenic variation in shaping the humoral immune compartment and provide a rational basis for the hierarchical nature of antibody titers against influenza A viruses in humans.


Immunologic Research | 2012

Diverse immune evasion strategies by human cytomegalovirus

Vanessa M. Noriega; Veronika Redmann; Thomas J. Gardner; Domenico Tortorella

Members of the Herpesviridae family have the capacity to undergo both lytic and latent infection to establish a lifelong relationship with their host. Following primary infection, human cytomegalovirus (HCMV) can persist as a subclinical, recurrent infection for the lifetime of an individual. This quiescent portion of its life cycle is termed latency and is associated with periodic bouts of reactivation during times of immunosuppression, inflammation, or stress. In order to exist indefinitely and establish infection, HCMV encodes a multitude of immune modulatory mechanisms devoted to escaping the host antiviral response. HCMV has become a paradigm for studies of viral immune evasion of antigen presentation by both major histocompatibility complex (MHC) class I and II molecules. By restricting the presentation of viral antigens during both productive and latent infection, HCMV limits elimination by the human immune system. This review will focus on understanding how the virus manipulates the pathways of antigen presentation in order to modulate the host response to infection.


Nature Communications | 2016

ISG15 deficiency and increased viral resistance in humans but not mice

Scott D. Speer; Zhi Li; Sofija Buta; Béatrice Payelle-Brogard; Li Qian; Frederic Vigant; Erminia Rubino; Thomas J. Gardner; Tim Wedeking; Mark Hermann; James Duehr; Ozden Sanal; Ilhan Tezcan; Nahal Mansouri; Payam Tabarsi; Davood Mansouri; Véronique Francois-Newton; Coralie F. Daussy; Marisela R. Rodriguez; Deborah J. Lenschow; Alexander N. Freiberg; Domenico Tortorella; Jacob Piehler; Benhur Lee; Adolfo García-Sastre; Sandra Pellegrini; Dusan Bogunovic

ISG15 is an interferon (IFN)-α/β-induced ubiquitin-like protein. It exists as a free molecule, intracellularly and extracellularly, and conjugated to target proteins. Studies in mice have demonstrated a role for Isg15 in antiviral immunity. By contrast, human ISG15 was shown to have critical immune functions, but not in antiviral immunity. Namely, free extracellular ISG15 is crucial in IFN-γ-dependent antimycobacterial immunity, while free intracellular ISG15 is crucial for USP18-mediated downregulation of IFN-α/β signalling. Here we describe ISG15-deficient patients who display no enhanced susceptibility to viruses in vivo, in stark contrast to Isg15-deficient mice. Furthermore, fibroblasts derived from ISG15-deficient patients display enhanced antiviral protection, and expression of ISG15 attenuates viral resistance to WT control levels. The species-specific gain-of-function in antiviral immunity observed in ISG15 deficiency is explained by the requirement of ISG15 to sustain USP18 levels in humans, a mechanism not operating in mice.


Molecular Immunology | 2012

Human cytomegalovirus US3 modulates destruction of MHC class I molecules.

Vanessa M. Noriega; Julia Hesse; Thomas J. Gardner; Katrin Besold; Bodo Plachter; Domenico Tortorella

Human cytomegalovirus (HCMV), a member of the Herpesviridae family, is proficient at establishing lifelong persistence within the host in part due to immune modulating genes that limit immune recognition. HCMV encodes at least five glycoproteins within its unique short (US) genomic region that interfere with MHC class I antigen presentation, thus hindering viral clearance by cytotoxic T lymphocytes (CTL). Specifically, US3 retains class I within the endoplasmic reticulum (ER), while US2 and US11 induce class I heavy chain destruction. A cooperative effect on class I down-regulation during stable expression of HCMV US2 and US3 has been established. To address the impact of US3 on US11-mediated MHC class I down-regulation, the fate of class I molecules was examined in US3/US11-expressing cells and virus infection studies. Co-expression of US3 and US11 resulted in a decrease of surface expression of class I molecules. However, the class I molecules in US3/US11 cells were mostly retained in the ER with an attenuated rate of proteasome destruction. Analysis of class I levels from virus-infected cells using HCMV variants either expressing US3 or US11 revealed efficient surface class I down-regulation upon expression of both viral proteins. Cells infected with both US3 and US11 expressing viruses demonstrate enhanced retention of MHC class I complexes within the ER. Collectively, the data suggests a paradigm where HCMV-induced surface class I down-regulation occurs by diverse mechanisms dependent on the expression of specific US genes. These results validate the commitment of HCMV to limiting the surface expression of class I levels during infection.


Viruses | 2014

Human Cytomegalovirus US28 Facilitates Cell-to-Cell Viral Dissemination

Vanessa M. Noriega; Thomas J. Gardner; Veronika Redmann; Gerold Bongers; Sergio A. Lira; Domenico Tortorella

Human cytomegalovirus (HCMV) encodes a number of viral proteins with homology to cellular G protein-coupled receptors (GPCRs). These viral GPCRs, including US27, US28, UL33, and UL78, have been ascribed numerous functions during infection, including activating diverse cellular pathways, binding to immunomodulatory chemokines, and impacting virus dissemination. To investigate the role of US28 during virus infection, two variants of the clinical isolate TB40/E were generated: TB40/E-US28YFP expressing a C-terminal yellow fluorescent protein tag, and TB40/E-FLAGYFP in which a FLAG-YFP cassette replaces the US28 coding region. The TB40/E-US28YFP protein localized as large perinuclear fluorescent structures at late times post-infection in fibroblasts, endothelial, and epithelial cells. Interestingly, US28YFP is a non-glycosylated membrane protein throughout the course of infection. US28 appears to impact cell-to-cell spread of virus, as the ΔUS28 virus (TB40/E-FLAGYFP) generated a log-greater yield of extracellular progeny whose spread could be significantly neutralized in fibroblasts. Most strikingly, in epithelial cells, where dissemination of virus occurs exclusively by the cell-to-cell route, TB40/E-FLAGYFP (ΔUS28) displayed a significant growth defect. The data demonstrates that HCMV US28 may contribute at a late stage of the viral life cycle to cell-to-cell dissemination of virus.


Microbiology and Molecular Biology Reviews | 2016

Virion Glycoprotein-Mediated Immune Evasion by Human Cytomegalovirus: a Sticky Virus Makes a Slick Getaway

Thomas J. Gardner; Domenico Tortorella

SUMMARY The prototypic herpesvirus human cytomegalovirus (CMV) exhibits the extraordinary ability to establish latency and maintain a chronic infection throughout the life of its human host. This is even more remarkable considering the robust adaptive immune response elicited by infection and reactivation from latency. In addition to the ability of CMV to exist in a quiescent latent state, its persistence is enabled by a large repertoire of viral proteins that subvert immune defense mechanisms, such as NK cell activation and major histocompatibility complex antigen presentation, within the cell. However, dissemination outside the cell presents a unique existential challenge to the CMV virion, which is studded with antigenic glycoprotein complexes targeted by a potent neutralizing antibody response. The CMV virion envelope proteins, which are critical mediators of cell attachment and entry, possess various characteristics that can mitigate the humoral immune response and prevent viral clearance. Here we review the CMV glycoprotein complexes crucial for cell attachment and entry and propose inherent properties of these proteins involved in evading the CMV humoral immune response. These include viral glycoprotein polymorphism, epitope competition, Fc receptor-mediated endocytosis, glycan shielding, and cell-to-cell spread. The consequences of CMV virion glycoprotein-mediated immune evasion have a major impact on persistence of the virus in the population, and a comprehensive understanding of these evasion strategies will assist in designing effective CMV biologics and vaccines to limit CMV-associated disease.


Clinical and Vaccine Immunology | 2013

Development of a high-throughput assay to measure the neutralization capability of anti-cytomegalovirus antibodies.

Thomas J. Gardner; Cynthia Bolovan-Fritts; Melissa W. Teng; Veronika Redmann; Thomas Kraus; Rhoda Sperling; Thomas M. Moran; William J. Britt; Leor S. Weinberger; Domenico Tortorella

ABSTRACT Infection by human cytomegalovirus (CMV) elicits a strong humoral immune response and robust anti-CMV antibody production. Diagnosis of virus infection can be carried out by using a variety of serological assays; however, quantification of serum antibodies against CMV may not present an accurate measure of a patients ability to control a virus infection. CMV strains that express green fluorescent protein (GFP) fusion proteins can be used as screening tools for evaluating characteristics of CMV infection in vitro. In this study, we employed a CMV virus strain, AD169, that ectopically expresses a yellow fluorescent protein (YFP) fused to the immediate-early 2 (IE2) protein product (AD169IE2-YFP) to quantify a CMV infection in human cells. We created a high-throughput cell-based assay that requires minimal amounts of material and provides a platform for rapid analysis of the initial phase of virus infection, including virus attachment, fusion, and immediate-early viral gene expression. The AD169IE2-YFP cell infection system was utilized to develop a neutralization assay with a monoclonal antibody against the viral surface glycoprotein gH. The high-throughput assay was extended to measure the neutralization capacity of serum from CMV-positive subjects. These findings describe a sensitive and specific assay for the quantification of a key immunological response that plays a role in limiting CMV dissemination and transmission. Collectively, we have demonstrated that a robust high-throughput infection assay can analyze the early steps of the CMV life cycle and quantify the potency of biological reagents to attenuate a virus infection.


Toxins | 2013

Novel Class of Potential Therapeutics that Target Ricin Retrograde Translocation

Veronika Redmann; Thomas J. Gardner; Zerlina Lau; Keita Morohashi; Dan P. Felsenfeld; Domenico Tortorella

Ricin toxin, an A-B toxin from Ricinus communis, induces cell death through the inhibition of protein synthesis. The toxin binds to the cell surface via its B chain (RTB) followed by its retrograde trafficking through intracellular compartments to the ER where the A chain (RTA) is transported across the membrane and into the cytosol. Ricin A chain is transported across the ER membrane utilizing cellular proteins involved in the disposal of aberrant ER proteins by a process referred to as retrograde translocation. Given the current lack of therapeutics against ricin intoxication, we developed a high-content screen using an enzymatically attenuated RTA chimera engineered with a carboxy-terminal enhanced green fluorescent protein (RTAE177Qegfp) to identify compounds that target RTA retrograde translocation. Stabilizing RTAE177Qegfp through the inclusion of proteasome inhibitor produced fluorescent peri-nuclear granules. Quantitative analysis of the fluorescent granules provided the basis to discover compounds from a small chemical library (2080 compounds) with known bioactive properties. Strikingly, the screen found compounds that stabilized RTA molecules within the cell and several compounds limited the ability of wild type RTA to suppress protein synthesis. Collectively, a robust high-content screen was developed to discover novel compounds that stabilize intracellular ricin and limit ricin intoxication.


Scientific Reports | 2016

Human cytomegalovirus gH stability and trafficking are regulated by ER-associated degradation and transmembrane architecture

Thomas J. Gardner; Rosmel Hernandez; Vanessa M. Noriega; Domenico Tortorella

The prototypic betaherpesvirus human cytomegalovirus (CMV) establishes life-long persistence within its human host. While benign in healthy individuals, CMV poses a significant threat to the immune compromised, including transplant recipients and neonates. The CMV glycoprotein complex gH/gL/gO mediates infection of fibroblasts, and together with the gH/gL/UL128/130/131 a pentameric complex permits infection of epithelial, endothethial, and myeloid cells. Given the central role of the gH/gL complex during infection, we were interested in studying cellular trafficking of the gH/gL complex through generation of human cells that stably express gH and gL. When expressed alone, CMV gH and gL were degraded through the ER-associated degradation (ERAD) pathway. However, co-expression of these proteins stabilized the polypeptides and enhanced their cell-surface expression. To further define regulatory factors involved in gH/gL trafficking, a CMV gH chimera in which the gH transmembrane and cytoplasmic tail were replaced with that of human CD4 protein permitted cell surface gH expression in absence of gL. We thus demonstrate the ability of distinct cellular processes to regulate the trafficking of viral glycoproteins. Collectively, the data provide insight into the processing and trafficking requirements of CMV envelope protein complexes and provide an example of the co-opting of cellular processes by CMV.


Viruses | 2016

The Microtubule Inhibitor Podofilox Inhibits an Early Entry Step of Human Cytomegalovirus.

Tobias Cohen; Toni M. Schwarz; Frederic Vigant; Thomas J. Gardner; Rosmel Hernandez; Benhur Lee; Domenico Tortorella

Human cytomegalovirus is a ubiquitous β-herpesvirus that infects many different cell types through an initial binding to cell surface receptors followed by a fusion event at the cell membrane or endocytic vesicle. A recent high-throughput screen to identify compounds that block a step prior to viral gene expression identified podofilox as a potent and nontoxic inhibitor. Time-of-addition studies in combination with quantitative-PCR analysis demonstrated that podofilox limits an early step of virus entry at the cell surface. Podofilox was also able to drastically reduce infection by herpes simplex 1, an α-herpesvirus with a very similar entry process to CMV. Podofilox caused a reduced maximal plateau inhibition of infection by viruses with single step binding processes prior to fusion-like Newcastle disease virus, Sendai virus, and influenza A virus or viruses that enter via endocytosis like vesicular stomatitis virus and a clinical-like strain of CMV. These results indicate that microtubules appear to be participating in the post-binding step of virus entry including the pre- and post-penetration events. Modulation of the plasma membrane is required to promote virus entry for herpesviruses, and that podofilox, unlike colchicine or nocodazole, is able to preferentially target microtubule networks at the plasma membrane.

Collaboration


Dive into the Thomas J. Gardner's collaboration.

Top Co-Authors

Avatar

Domenico Tortorella

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Veronika Redmann

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Vanessa M. Noriega

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Adolfo García-Sastre

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Peter Palese

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Thomas M. Moran

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ana Fernandez-Sesma

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Benhur Lee

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Benjamin D. Greenbaum

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Camilla Melegari

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge