Network


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

Hotspot


Dive into the research topics where Suzanne E. Queen is active.

Publication


Featured researches published by Suzanne E. Queen.


RNA Biology | 2013

Real-time quantitative PCR and droplet digital PCR for plant miRNAs in mammalian blood provide little evidence for general uptake of dietary miRNAs: Limited evidence for general uptake of dietary plant xenomiRs

Kenneth W. Witwer; Melissa A. McAlexander; Suzanne E. Queen; Robert J. Adams

Evidence that exogenous dietary miRNAs enter the bloodstream and tissues of ingesting animals has been accompanied by an indication that at least one plant miRNA, miR168, participates in “cross-kingdom” regulation of a mammalian transcript. If confirmed, these findings would support investigation of miRNA-based dietary interventions in disease. Here, blood was obtained pre- and post-prandially (1, 4, 12 h) from pigtailed macaques that received a miRNA-rich plant-based substance. Plant and endogenous miRNAs were measured by RT-qPCR. Although low-level amplification was observed for some plant miRNA assays, amplification was variable and possibly non-specific, as suggested by droplet digital PCR. A consistent response to dietary intake was not observed. While our results do not support general and consistent uptake of dietary plant miRNAs, additional studies are needed to establish whether or not plant or animal xenomiRs are transferred across the gut in sufficient quantity to regulate endogenous genes.


The Journal of Infectious Diseases | 2010

Simian Immunodeficiency Virus-Infected Macaques Treated with Highly Active Antiretroviral Therapy Have Reduced Central Nervous System Viral Replication and Inflammation but Persistence of Viral DNA

M. Christine Zink; Angela K. Brice; Kathleen Kelly; Suzanne E. Queen; Lucio Gama; Ming Li; Robert J. Adams; Christopher M. Bartizal; John Varrone; S. Alireza Rabi; David R. Graham; Patrick M. Tarwater; Joseph L. Mankowski; Janice E. Clements

BACKGROUND During the era of highly active antiretroviral therapy (HAART), the prevalence of HIV-associated central nervous system (CNS) disease has increased despite suppression of plasma viremia. METHODS In a simian immunodeficiency virus (SIV) model system in which all animals develop AIDS and 90% develop CNS disease by 3 months after inoculation, pigtailed macaques were treated with a regimen of tenofovir disoproxil fumarate, saquinavir, atazanavir, and an integrase inhibitor starting at 12 days after inoculation and were euthanized at approximately 175 days after inoculation. RESULTS Plasma and cerebrospinal fluid (CSF) viral loads declined rapidly after the initiation of HAART. Brain viral RNA was undetectable at necropsy, but viral DNA levels were not different from those in untreated SIV-infected macaques. CNS inflammation was significantly reduced, with decreased brain expression of major histocompatibility complex class II and glial fibrillary acidic protein and reduced levels of CSF CCL2 and interleukin 6. Brain from treated macaques had significantly lower levels of interferon beta, type 1 interferon-inducible gene myxovirus (influenza) resistance A, and indolamine 2,3-dioxygenase messenger RNA, suggesting that immune hyperactivation was suppressed, and fewer CD4(+) and CD8(+) T cells, suggesting that trafficking of T cells from peripheral blood was reduced. Brain levels of CD68 protein and tumor necrosis factor alpha and interferon gamma RNA were reduced but were not significantly lower, indicating continued CNS inflammation. CONCLUSIONS These data, generated in a rigorous, high-viral-load SIV-infected macaque model, showed that HAART provided benefits with respect to CNS viral replication and inflammation but that no change in the level of viral DNA and continued CNS inflammation occurred in some macaques.


PLOS ONE | 2009

Coordinated regulation of SIV replication and immune responses in the CNS.

Kenneth W. Witwer; Lucio Gama; Ming Li; Christopher M. Bartizal; Suzanne E. Queen; John Varrone; Angela K. Brice; David R. Graham; Patrick M. Tarwater; Joseph L. Mankowski; M. Christine Zink; Janice E. Clements

Central nervous system (CNS) invasion during acute-stage HIV-infection has been demonstrated in a small number of individuals, but there is no evidence of neurological impairment at this stage and virus infection in brain appears to be controlled until late-stage disease. Using our reproducible SIV macaque model to examine the earliest stages of infection in the CNS, we identified immune responses that differentially regulate inflammation and virus replication in the brain compared to the peripheral blood and lymphoid tissues. SIV replication in brain macrophages and in brain of SIV-infected macaques was detected at 4 days post-inoculation (p.i.). This was accompanied by upregulation of innate immune responses, including IFNβ, IFNβ-induced gene MxA mRNA, and TNFα. Additionally, IL-10, the chemokine CCL2, and activation markers in macrophages, endothelial cells, and astrocytes were all increased in the brain at four days p.i. We observed synchronous control of virus replication, cytokine mRNA levels and inflammatory markers (MHC Class II, CD68 and GFAP) by 14 days p.i.; however, control failure was followed by development of CNS lesions in the brain. SIV infection was accompanied by induction of the dominant-negative isoform of C/EBPβ, which regulates SIV, CCL2, and IL6 transcription, as well as inflammatory responses in macrophages and astrocytes. This synchronous response in the CNS is in part due to the effect of the C/EBPβ on virus replication and cytokine expression in macrophage-lineage cells in contrast to CD4+ lymphocytes in peripheral blood and lymphoid tissues. Thus, we have identified a crucial period in the brain when virus replication and inflammation are controlled. As in HIV-infected individuals, though, this control is not sustained in the brain. Our results suggest that intervention with antiretroviral drugs or anti-inflammatory therapeutics with CNS penetration would sustain early control. These studies further suggest that interventions should target HIV-infected individuals with increased CCL2 levels or HIV RNA in the CNS.


Journal of Leukocyte Biology | 2012

Expansion of a subset of CD14highCD16negCCR2low/neg monocytes functionally similar to myeloid‐derived suppressor cells during SIV and HIV infection

Lucio Gama; Erin N. Shirk; Julia N. Russell; Karina I. Carvalho; Ming Li; Suzanne E. Queen; Jorge Kalil; M. Christine Zink; Janice E. Clements; Esper G. Kallas

Monocytes have been categorized in three main subpopulations based on CD14 and CD16 surface expression. Classical monocytes express the CD14++CD16−CCR2+ phenotype and migrate to inflammatory sites by quickly responding to CCL2 signaling. Here, we identified and characterized the expansion of a novel monocyte subset during HIV and SIV infection, which were undistinguishable from classical monocytes, based on CD14 and CD16 expression, but expressed significantly lower surface CCR2. Transcriptome analysis of sorted cells demonstrated that the CCR2low/neg cells are a distinct subpopulation and express lower levels of inflammatory cytokines and activation markers than their CCR2high counterparts. They exhibited impaired phagocytosis and greatly diminished chemotaxis in response to CCL2 and CCL7. In addition, these monocytes are refractory to SIV infection and suppress CD8+ T cell proliferation in vitro. These cells express higher levels of STAT3 and NOS2, suggesting a phenotype similar to monocytic myeloid‐derived cells, which suppress expansion of CD8+ T cells in vivo. They may reflect an antiproliferative response against the extreme immune activation observed during HIV and SIV infections. In addition, they may suppress antiviral responses and thus, have a role in AIDS pathogenesis. Antiretroviral therapy in infected macaque and human subjects caused this population to decline, suggesting that this atypical phenotype is linked to viral replication.


Journal of Virology | 2016

Quantitation of Productively Infected Monocytes and Macrophages of Simian Immunodeficiency Virus-Infected Macaques.

Claudia R. Avalos; Sarah L. Price; Ellen R. Forsyth; Julia N. Pin; Erin N. Shirk; Brandon T. Bullock; Suzanne E. Queen; Ming Li; Dane D. Gellerup; Shelby L. O'Connor; M. Christine Zink; Joseph L. Mankowski; Lucio Gama; Janice E. Clements

ABSTRACT Despite the success of combined antiretroviral therapy (ART), human immunodeficiency virus (HIV) infection remains a lifelong infection because of latent viral reservoirs in infected patients. The contribution of CD4+ T cells to infection and disease progression has been extensively studied. However, during early HIV infection, macrophages in brain and other tissues are infected and contribute to tissue-specific diseases, such as encephalitis and dementia in brain and pneumonia in lung. The extent of infection of monocytes and macrophages has not been rigorously assessed with assays comparable to those used to study infection of CD4+ T cells and to evaluate the number of CD4+ T cells that harbor infectious viral genomes. To assess the contribution of productively infected monocytes and macrophages to HIV- and simian immunodeficiency virus (SIV)-infected cells in vivo, we developed a quantitative virus outgrowth assay (QVOA) based on similar assays used to quantitate CD4+ T cell latent reservoirs in HIV- and SIV-infected individuals in whom the infection is suppressed by ART. Myeloid cells expressing CD11b were serially diluted and cocultured with susceptible cells to amplify virus. T cell receptor β RNA was measured as a control to assess the potential contribution of CD4+ T cells in the assay. Virus production in the supernatant was quantitated by quantitative reverse transcription-PCR. Productively infected myeloid cells were detected in blood, bronchoalveolar lavage fluid, lungs, spleen, and brain, demonstrating that these cells persist throughout SIV infection and have the potential to contribute to the viral reservoir during ART. IMPORTANCE Infection of CD4+ T cells and their role as latent reservoirs have been rigorously assessed; however, the frequency of productively infected monocytes and macrophages in vivo has not been similarly studied. Myeloid cells, unlike lymphocytes, are resistant to the cytopathic effects of HIV. Moreover, tissue-resident macrophages have the ability to self-renew and persist in the body for months to years. Thus, tissue macrophages, once infected, have the characteristics of a potentially stable viral reservoir. A better understanding of the number of productively infected macrophages is crucial to further evaluate the role of infected myeloid cells as a potential viral reservoir. In the study described here we compared the frequency of productively infected CD4+ T cells and macrophages in an SIV-infected macaque model. We developed a critical assay that will allow us to quantitate myeloid cells containing viral genomes that lead to productive infection in SIV-infected macaques and assess the role of macrophages as potential reservoirs.


Journal of Neuropathology and Experimental Neurology | 2007

Pathogenesis of simian immunodeficiency virus-induced alterations in macaque trigeminal ganglia.

Victoria A. Laast; Carlos A. Pardo; Patrick M. Tarwater; Suzanne E. Queen; Todd A. Reinhart; Mimi Ghosh; Robert J. Adams; M. Christine Zink; Joseph L. Mankowski

Peripheral neuropathy is the most frequent neurologic complication associated with human immunodeficiency virus (HIV) infection, yet its pathogenesis remains poorly understood. To study the mechanisms causing HIV-induced peripheral nervous system disease, we examined trigeminal ganglia obtained from simian immunodeficiency virus (SIV)-inoculated macaques. SIV-infected macaques developed multifocal trigeminal ganglionitis of varying severity characterized by multifocal mononuclear infiltrates, neuronophagia, and neuronal loss resembling reports of HIV-associated changes present in dorsal root ganglia. Neuronal density, measured by calculating the fractional area of trigeminal ganglia occupied by neurons, was significantly lower in SIV-infected macaques versus uninfected macaques (p = 0.001). To characterize the inflammatory cell population and measure productive viral infection in ganglia, trigeminal ganglia from SIV-infected macaques were immunostained for macrophage or cytotoxic lymphocyte markers and for SIV gp41. The extent of macrophage infiltration in trigeminal ganglia was inversely correlated with neuronal loss (p = 0.001), whereas cytotoxic lymphocyte infiltration was not associated with neuronal loss. These studies demonstrate that alterations in the somatosensory ganglia of SIV-infected macaques closely parallel those observed in HIV-infected individuals and show that study of SIV-infected macaques may help elucidate the pathophysiology of HIV-induced peripheral neuropathy.


The Journal of Infectious Diseases | 2013

Platelet Activation and Platelet-Monocyte Aggregate Formation Contribute to Decreased Platelet Count During Acute Simian Immunodeficiency Virus Infection in Pig-tailed Macaques

Kelly A. Metcalf Pate; Claire E. Lyons; Jamie L. Dorsey; Erin N. Shirk; Suzanne E. Queen; Robert J. Adams; Lucio Gama; Craig N. Morrell; Joseph L. Mankowski

Platelets are key participants in innate immune responses to pathogens. As a decrease in circulating platelet count is one of the initial hematologic indicators of human immunodeficiency virus (HIV) infection, we sought to determine whether decline in platelet number during acute infection results from decreased production, increased antibody-mediated destruction, or increased platelet activation in a simian immunodeficiency virus (SIV)/macaque model. During acute SIV infection, circulating platelets were activated with increased surface expression of P-selection, CD40L and major histocompatibility complex class I. Platelet production was maintained and platelet autoantibodies were not detected during acute infection. Concurrent with a decrease in platelet numbers and an increase in circulating monocytes, platelets were found sequestered in platelet-monocyte aggregates, thereby contributing to the decline in platelet counts. Because the majority of circulating CD16(+) monocytes formed complexes with platelets during acute SIV infection, a decreased platelet count may represent platelet participation in the innate immune response to HIV.


Journal of NeuroVirology | 2003

Elevated Peripheral Benzodiazepine Receptor Expression in Simian Immunodeficiency Virus Encephalitis

Joseph L. Mankowski; Suzanne E. Queen; Patrick J. Tarwater; Robert J. Adams; Tomás R. Guilarte

Measurement of central nervous system (CNS) expression of the peripheral benzodiazepine receptor (PBR), a microglia and macrophage activation marker, by positron emission tomography (PET) would aid clinical management of human immunodeficiency virus (HIV)-infected patients. To evaluate the utility of examining PBR expression in the CNS as a cellular activation marker in HIV CNS disease, PBR levels were measured in frontal cortex of simian immunodeficiency virus (SIV)-infected macaques with encephalitis and uninfected animals via PK11195 ligand autoradiography. [3H]-(R)-PK11195 binding to both grey matter (P = .017) and white matter (P = .038) was significantly higher in animals with SIV encephalitis (n = 10) versus control animals (n = 3). When PK11195 binding was compared with other microglial/macrophage activation markers (obtained via quantitative image analysis), a strong, significant association was found for both HAM56 (P = .004) and KP-1 (anti-CD68; P = .006) immunostaining in white matter. In contrast, grey matter PK11195 binding did not correlate with HAM56 (P = .46), KP-1 (P = .06), or glial fibrillary acidic protein (GFAP) immunostaining for astrocytic activation (P = .09). The regional nature of these increases in activation within the brain illustrates the crucial need to focus functional neuroimaging analyses of HIV-infected individuals on subcortical white matter to assess activation of microglia and macrophages.


The Journal of Infectious Diseases | 2015

A Murine Viral Outgrowth Assay to Detect Residual HIV Type 1 in Patients With Undetectable Viral Loads

Kelly A. Metcalf Pate; Christopher W. Pohlmeyer; Victoria E. Walker-Sperling; Jeremy B. Foote; Kevin M. Najarro; Catherine G. Cryer; Maria Salgado; Lucio Gama; Elizabeth L. Engle; Erin N. Shirk; Suzanne E. Queen; Stanley Chioma; Meghan S. Vermillion; Brandon T. Bullock; Ming Li; Claire E. Lyons; Robert J. Adams; M. Christine Zink; Janice E. Clements; Joseph L. Mankowski; Joel N. Blankson

BACKGROUND Sensitive assays are needed for detection of residual human immunodeficiency virus (HIV) in patients with undetectable plasma viral loads to determine whether eradication strategies are effective. The gold standard quantitative viral outgrowth assay (QVOA) underestimates the magnitude of the viral reservoir. We sought to determine whether xenograft of leukocytes from HIV type 1 (HIV)-infected patients with undetectable plasma viral loads into immunocompromised mice would result in viral amplification. METHODS Peripheral blood mononuclear cells or purified CD4(+) T cells from HIV or simian immunodeficiency virus (SIV)-infected subjects with undetectable plasma viral loads were adoptively transferred into NOD.Cg-Prkdc(scid)Il2rg(tm1Wjl)/SzJ (NSG) mice. The mice were monitored for viremia following depletion of human CD8(+) T cells to minimize antiviral activity. In some cases, humanized mice were also treated with activating anti-CD3 antibody. RESULTS With this murine viral outgrowth assay (MVOA), we successfully amplified replication-competent HIV or SIV from all subjects tested, including 5 HIV-positive patients receiving suppressive antiretroviral therapy (ART) and 6 elite controllers or suppressors who were maintaining undetectable viral loads without ART, including an elite suppressor from whom we were unable to recover virus by QVOA. CONCLUSIONS Our results suggest that the MVOA has the potential to serve as a powerful tool to identify residual HIV in patients with undetectable viral loads.


Journal of NeuroVirology | 1999

Alterations in blood-brain barrier glucose transport in SIV-infected macaques

Joseph L. Mankowski; Suzanne E. Queen; Lynn M Kirstein; Jeffrey P. Spelman; John Laterra; Ian A. Simpson; Robert J. Adams; Janice E. Clements; M. Christine Zink

The neurological manifestations of HIV infection may be in part due to alterations in the blood-brain barrier. These may be caused by structural changes in the barrier or may consist of subtle metabolic or biochemical disturbances in barrier function. In the CNS, the family of glucose transporter proteins plays a key role in controlling movement of glucose across cell membranes. The 55 kDa isoform of glucose transporter 1 (GLUT1) regulates import of glucose from blood to brain across the endothelial cells of the blood-brain barrier (BBB), whereas the 45 kDa form of GLUT1 predominantly regulates nonvascular glial glucose uptake. In this study, expression of 55 and 45 kDa forms of GLUT1 in different regions of the brain from 18 SIV-infected macaques was measured by quantitative immunoblot and then compared with the severity of SIV encephalitis to determine whether neurologic disease is related to altered glucose metabolism at the BBB and in brain parenchyma. An inverse relationship was found between severity of SIV encephalitis and expression of the endothelial 55 kDa isoform of GLUT1 at the BBB in cortical grey matter, caudate nucleus, and cerebellum. A similar relationship also was found for the glial 45 kDa GLUT1 isoform in cortical grey matter. In addition, a significant increase in 55 kDa GLUT1 expression was found in caudate nucleus during the early stages of infection. In the brains of macaques with moderate to severe encephalitis, 55 kDa GLUT1 expression had declined to pre-infection levels. These GLUT1 alterations at the BBB and in glial cells may reflect severe disturbances in the CNS microenvironment that contribute to CNS dysfunction.

Collaboration


Dive into the Suzanne E. Queen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert J. Adams

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

M. Christine Zink

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lucio Gama

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Patrick M. Tarwater

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Janice E. Clements

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Erin N. Shirk

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kelly A. Metcalf Pate

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jamie L. Dorsey

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sarah E. Beck

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge