Network


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

Hotspot


Dive into the research topics where Joan M. Chapman is active.

Publication


Featured researches published by Joan M. Chapman.


Journal of Experimental Medicine | 2008

Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection

R. Brad Jones; Lishomwa C. Ndhlovu; Jason D. Barbour; Prameet M. Sheth; Aashish R. Jha; Brian R. Long; Jessica C. Wong; Malathy Satkunarajah; Marc Schweneker; Joan M. Chapman; Gabor Gyenes; Bahareh Vali; Martin D. Hyrcza; Feng Yun Yue; Colin Kovacs; Aref Sassi; Mona Loutfy; Roberta Halpenny; Desmond Persad; Gerald Spotts; Frederick Hecht; Tae-Wook Chun; Joseph M. McCune; Rupert Kaul; James M. Rini; Douglas F. Nixon; Mario A. Ostrowski

Progressive loss of T cell functionality is a hallmark of chronic infection with human immunodeficiency virus 1 (HIV-1). We have identified a novel population of dysfunctional T cells marked by surface expression of the glycoprotein Tim-3. The frequency of this population was increased in HIV-1–infected individuals to a mean of 49.4 ± SD 12.9% of CD8+ T cells expressing Tim-3 in HIV-1–infected chronic progressors versus 28.5 ± 6.8% in HIV-1–uninfected individuals. Levels of Tim-3 expression on T cells from HIV-1–infected inviduals correlated positively with HIV-1 viral load and CD38 expression and inversely with CD4+ T cell count. In progressive HIV-1 infection, Tim-3 expression was up-regulated on HIV-1–specific CD8+ T cells. Tim-3–expressing T cells failed to produce cytokine or proliferate in response to antigen and exhibited impaired Stat5, Erk1/2, and p38 signaling. Blocking the Tim-3 signaling pathway restored proliferation and enhanced cytokine production in HIV-1–specific T cells. Thus, Tim-3 represents a novel target for the therapeutic reversal of HIV-1–associated T cell dysfunction.


PLOS ONE | 2006

Strong HIV-1-specific T cell responses in HIV-1-exposed uninfected infants and neonates revealed after regulatory T cell removal.

Fatema A. Legrand; Douglas F. Nixon; Christopher P. Loo; Erika Ono; Joan M. Chapman; Maristela Miyamoto; Ricardo Sobhie Diaz; Amélia Miyashiro Nunes dos Santos; Regina Célia de Menezes Succi; Jacob Abadi; Michael G. Rosenberg; Maria Isabel de Moraes-Pinto; Esper G. Kallas

Background In utero transmission of HIV-1 occurs on average in only 3%–15% of HIV-1-exposed neonates born to mothers not on antiretroviral drug therapy. Thus, despite potential exposure, the majority of infants remain uninfected. Weak HIV-1-specific T-cell responses have been detected in children exposed to HIV-1, and potentially contribute to protection against infection. We, and others, have recently shown that the removal of CD4+CD25+ T-regulatory (Treg) cells can reveal strong HIV-1 specific T-cell responses in some HIV-1 infected adults. Here, we hypothesized that Treg cells could suppress HIV-1-specific immune responses in young children. Methodology/Principal Findings We studied two cohorts of children. The first group included HIV-1-exposed-uninfected (EU) as well as unexposed (UNEX) neonates. The second group comprised HIV-1-infected and HIV-1-EU children. We quantified the frequency of Treg cells, T-cell activation, and cell-mediated immune responses. We detected high levels of CD4+CD25+CD127− Treg cells and low levels of CD4+ and CD8+ T cell activation in the cord blood of the EU neonates. We observed HIV-1-specific T cell immune responses in all of the children exposed to the virus. These T-cell responses were not seen in the cord blood of control HIV-1 unexposed neonates. Moreover, the depletion of CD4+CD25+ Treg cells from the cord blood of EU newborns strikingly augmented both CD4+ and CD8+ HIV-1-specific immune responses. Conclusions/Significance This study provides new evidence that EU infants can mount strong HIV-1-specific T cell responses, and that in utero CD4+CD25+ T-regulatory cells may be contributing to the lack of vertical transmission by reducing T cell activation.


PLOS Pathogens | 2007

T Cell Responses to Human Endogenous Retroviruses in HIV-1 Infection

Keith E. Garrison; R. Brad Jones; Duncan A. Meiklejohn; Naveed Anwar; Lishomwa C. Ndhlovu; Joan M. Chapman; Ann L. Erickson; Ashish Agrawal; Gerald Spotts; Frederick Hecht; Seth Rakoff-Nahoum; Jack Lenz; Mario A. Ostrowski; Douglas F. Nixon

Human endogenous retroviruses (HERVs) are remnants of ancient infectious agents that have integrated into the human genome. Under normal circumstances, HERVs are functionally defective or controlled by host factors. In HIV-1-infected individuals, intracellular defense mechanisms are compromised. We hypothesized that HIV-1 infection would remove or alter controls on HERV activity. Expression of HERV could potentially stimulate a T cell response to HERV antigens, and in regions of HIV-1/HERV similarity, these T cells could be cross-reactive. We determined that the levels of HERV production in HIV-1-positive individuals exceed those of HIV-1-negative controls. To investigate the impact of HERV activity on specific immunity, we examined T cell responses to HERV peptides in 29 HIV-1-positive and 13 HIV-1-negative study participants. We report T cell responses to peptides derived from regions of HERV detected by ELISPOT analysis in the HIV-1-positive study participants. We show an inverse correlation between anti-HERV T cell responses and HIV-1 plasma viral load. In HIV-1-positive individuals, we demonstrate that HERV-specific T cells are capable of killing cells presenting their cognate peptide. These data indicate that HIV-1 infection leads to HERV expression and stimulation of a HERV-specific CD8+ T cell response. HERV-specific CD8+ T cells have characteristics consistent with an important role in the response to HIV-1 infection: a phenotype similar to that of T cells responding to an effectively controlled virus (cytomegalovirus), an inverse correlation with HIV-1 plasma viral load, and the ability to lyse cells presenting their target peptide. These characteristics suggest that elicitation of anti-HERV-specific immune responses is a novel approach to immunotherapeutic vaccination. As endogenous retroviral sequences are fixed in the human genome, they provide a stable target, and HERV-specific T cells could recognize a cell infected by any HIV-1 viral variant. HERV-specific immunity is an important new avenue for investigation in HIV-1 pathogenesis and vaccine design.


AIDS | 2008

Suppression of HIV-1 plasma viral load below detection preserves IL-17 producing T cells in HIV-1 infection

Lishomwa C. Ndhlovu; Joan M. Chapman; Aashish R. Jha; Jennifer E. Snyder-Cappione; Moraima Pagán; Fabio E. Leal; Brigid S Boland; Philip J. Norris; Michael G. Rosenberg; Douglas F. Nixon

IL-17 is proinflammatory cytokine secreted by a unique CD4+ T (Th17) cell subset and proposed to play a role in host defense. We hypothesized that Th17 cells are lost in HIV-1 infection. HIV-1-infected children with plasma viremia below 50 copies/ml had IL-17 production, whereas those with detectable viremia had minimal secretion. These results imply viral-mediated destruction or impairment of Th17 cells and argue for complete suppression of viremia for reconstitution of Th17 cells.


The Journal of Infectious Diseases | 2007

Individuals with Pulmonary Tuberculosis Have Lower Levels of Circulating CD1d-Restricted NKT Cells

Jennifer E. Snyder-Cappione; Douglas F. Nixon; Christopher P. Loo; Joan M. Chapman; Duncan A. Meiklejohn; Fernando F. Melo; Priscilla R. Costa; Johan K. Sandberg; Denise S. Rodrigues; Esper G. Kallas

Mycobacterium tuberculosis (MTB) is a leading cause of mortality worldwide from an infectious agent. Natural killer T (NKT) cells recognize mycobacterial antigens and contribute to anti-MTB immunity in mouse models. NKT cells were measured in subjects with pulmonary tuberculosis, MTB-exposed individuals, and healthy controls. NKT cell levels are selectively lower in peripheral blood mononuclear cells from individuals with pulmonary tuberculosis than in both MTB-exposed subjects and healthy control subjects. This apparent loss of NKT cells from the peripheral blood is sustained during the 6 months after the initiation of MTB treatment. These findings indicate that NKT cells may be an important component of antituberculosis immunity.


Clinical and Vaccine Immunology | 2008

Elevated Frequency of Gamma Interferon-Producing NK Cells in Healthy Adults Vaccinated against Influenza Virus

Brian R. Long; Jakob Michaëlsson; Christopher P. Loo; Wassim M. Ballan; Bien-Aimee N. Vu; Frederick Hecht; Lewis L. Lanier; Joan M. Chapman; Douglas F. Nixon

ABSTRACT Recent studies indicate that innate immunity in influenza virus infection is an area of substantial importance for our understanding of influenza virus pathogenesis, yet our knowledge of the mechanisms controlling innate immunity remains limited. Further delineation of the roles of NK cells and innate immunity in viral infection may have important implications for the development of improved influenza virus vaccines. In this study, we evaluated the phenotype and function of NK and T lymphocytes, as well as influenza virus-specific immunoglobulin G production, prior to and following vaccination with the routinely administered trivalent influenza virus vaccine. We demonstrate influenza virus antigen-specific innate and adaptive cellular responses and evaluate changes in NK cell receptor expression over time. Our results demonstrate increased innate and adaptive cellular immune responses and show that NK cells are a significant source of gamma interferon (IFN-γ) following influenza virus vaccination. An increase in the frequency of IFN-γ-producing NK cells was observed in many subjects postvaccination. The subset distribution with respect to CD56dim and CD56bright NK cell subsets remained stable, as did the NK cell phenotype with respect to expression of cell surface activating and inhibitory receptors. These results may form the basis for further investigations of the role of NK cells in immunity to influenza.


BMC Immunology | 2008

The frequency of CD127low expressing CD4+CD25high T regulatory cells is inversely correlated with human T lymphotrophic virus type-1 (HTLV-1) proviral load in HTLV-1-infection and HTLV-1-associated myelopathy/tropical spastic paraparesis

Jakob Michaëlsson; Hugo Marcelo R Barbosa; Kimberley A Jordan; Joan M. Chapman; Milena Kc Brunialti; Walter Kleine Neto; Youko Nukui; Ester C. Sabino; Marco A. Chieia; Acary Souza Bulle Oliveira; Douglas F. Nixon; Esper G. Kallas

BackgroundCD4+CD25high regulatory T (TReg) cells modulate antigen-specific T cell responses, and can suppress anti-viral immunity. In HTLV-1 infection, a selective decrease in the function of TReg cell mediated HTLV-1-tax inhibition of FOXP3 expression has been described. The purpose of this study was to assess the frequency and phenotype of TReg cells in HTLV-1 asymptomatic carriers and in HTLV-1-associated neurological disease (HAM/TSP) patients, and to correlate with measures of T cell activation.ResultsWe were able to confirm that HTLV-I drives activation, spontaneous IFNγ production, and proliferation of CD4+ T cells. We also observed a significantly lower proportion of CTLA-4+ TReg cells (CD4+CD25high T cells) in subjects with HAM/TSP patients compared to healthy controls. Ki-67 expression was negatively correlated to the frequency of CTLA-4+ TReg cells in HAM/TSP only, although Ki-67 expression was inversely correlated with the percentage of CD127low TReg cells in healthy control subjects. Finally, the proportion of CD127low TReg cells correlated inversely with HTLV-1 proviral load.ConclusionTaken together, the results suggest that TReg cells may be subverted in HAM/TSP patients, which could explain the marked cellular activation, spontaneous cytokine production, and proliferation of CD4+ T cells, in particular those expressing the CD25highCD127low phenotype. TReg cells represent a potential target for therapeutic intervention for patients with HTLV-1-related neurological diseases.


Immunology | 2010

Increased number and function of natural killer cells in human immunodeficiency virus 1‐positive subjects co‐infected with herpes simplex virus 2

Brian R. Long; Ann E. Erickson; Joan M. Chapman; Jason D. Barbour; Bien-Aimee N. Vu; Emily L. Ho; Lewis L. Lanier; Mariana M. Sauer; Karina I. Carvalho; Douglas F. Nixon; Esper G. Kallas

Natural killer (NK) cells bridge the interface between innate and adaptive immunity and are implicated in the control of herpes simplex virus 2 (HSV‐2) infection. In subjects infected with human immunodeficiency virus 1 (HIV‐1), the critical impact of the innate immune response on disease progression has recently come into focus. Higher numbers of NK cells are associated with lower HIV‐1 plasma viraemia. Individuals with the compound genotype of killer cell immunoglobulin‐like receptor (KIR) 3DS1 and human leucocyte antigen (HLA)‐Bw4‐80I, or who have alleles of KIR3DL1 that encode proteins highly expressed on the NK cell surface, have a significant delay in disease progression. We studied the effect of HSV‐2 co‐infection in HIV‐1‐infected subjects, and show that HSV‐2 co‐infection results in a pan‐lymphocytosis, with elevated absolute numbers of CD4+ and CD8+ T cells, and NK cells. The NK cells in HSV‐2 co‐infected subjects functioned more efficiently, with an increase in degranulation after in vitro stimulation. The number of NK cells expressing the activating receptors NKp30 and NKp46, and expressing KIR3DL1 or KIR3DS1, was inversely correlated with HIV‐1 plasma viral load in subjects mono‐infected with HIV‐1, but not in subjects co‐infected with HSV‐2. This suggests that HSV‐2 infection mediates changes within the NK cell population that may affect immunity in HIV‐1 infection.


Journal of Virology | 2007

Antiretroviral Drug Therapy Alters the Profile of Human Immunodeficiency Virus Type 1-Specific T-Cell Responses and Shifts the Immunodominant Cytotoxic T-Lymphocyte Response from Gag to Pol

Annika C. Karlsson; Joan M. Chapman; B. D. Heiken; Esper G. Kallas; Jeffery N Martin; Frederick Hecht; Steven G. Deeks; Douglas F. Nixon

ABSTRACT Antiretroviral drug therapy and cytotoxic T lymphocytes (CTL) both exert selective pressures on human immunodeficiency virus type 1, which influence viral evolution. Compared to chronically infected, antiretroviral-untreated patients, most chronically infected, treated patients with detectable viremia lack a cellular immune response against the Gag 77-85(SL9) epitope but show a new immunodominant response against an epitope in protease PR 76-84. Hence, mutations induced by antiretroviral therapy likely alter the profile of epitopes presented to T cells and thus the direction of the response. The consequences of dual pressures from treatment and CTL need to be considered in monitoring of drug therapy.


PLOS ONE | 2007

Sequential broadening of CTL responses in early HIV-1 infection is associated with viral escape.

Annika C. Karlsson; Astrid K. N. Iversen; Joan M. Chapman; Tulio de Oliveira; Gerald Spotts; Andrew J. McMichael; Miles P. Davenport; Frederick Hecht; Douglas F. Nixon

Collaboration


Dive into the Joan M. Chapman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian R. Long

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerald Spotts

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lishomwa C. Ndhlovu

University of Hawaii at Manoa

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge