Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
PLoS Pathog ; 11(10): e1005199, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26431533

ABSTRACT

Memory CD8 T cells provide protection to immune hosts by eliminating pathogen-infected cells during re-infection. While parameters influencing the generation of primary (1°) CD8 T cells are well established, the factors controlling the development of secondary (2°) CD8 T cell responses remain largely unknown. Here, we address the mechanisms involved in the generation and development of 2° memory (M) CD8 T cells. We observed that the time at which 1° M CD8 T cells enter into immune response impacts their fate and differentiation into 2° M CD8 T cells. Late-entry of 1° M CD8 T cells into an immune response (relative to the onset of infection) not only facilitated the expression of transcription factors associated with memory formation in 2° effector CD8 T cells, but also influenced the ability of 2° M CD8 T cells to localize within the lymph nodes, produce IL-2, and undergo Ag-driven proliferation. The timing of stimulation of 1° M CD8 T cells also impacted the duration of expression of the high-affinity IL-2 receptor (CD25) on 2° effector CD8 T cells and their sensitivity to IL-2 signaling. Importantly, by blocking or enhancing IL-2 signaling in developing 2° CD8 T cells, we provide direct evidence for the role of IL-2 in controlling the differentiation of Ag-driven 2° CD8 T cell responses. Thus, our data suggest that the process of 1° M to 2° M CD8 T cell differentiation is not fixed and can be manipulated, a notion with relevance for the design of future prime-boost vaccination approaches.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-2/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Flow Cytometry , Immunoblotting , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Time Factors
2.
J Immunol ; 195(1): 116-25, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25980007

ABSTRACT

Patients who survive sepsis display suppressed immune functions, often manifested as an increased susceptibility to secondary infections. Recently, using a cecal-ligation and puncture (CLP) model of sepsis, we showed that sepsis induces substantial and long-lasting changes in the available naive CD8(+) T cell repertoire affecting the capacity of the host to respond to newly encountered acute infections. However, the extent to which sepsis changes the host susceptibility to chronic infection and affects CD8(+) T cell responses is currently unknown. In this study, we demonstrate that inbred and outbred mice recovering from a septic event are more susceptible to lymphocytic choriomeningitis virus (LCMV) clone-13 infection exhibited by mortality and viral burden. Primary virus-specific CD8(+) T cells in LCMV clone-13-infected septic mice displayed exacerbated CD8(+) T cell exhaustion illustrated by increased inhibitory molecule expression (e.g., programmed cell death 1, lymphocyte-activation gene 3, and 2B4) and diminished Ag-driven cytokine production (e.g., IFN-γ, TNF-α) compared with similarly infected sham-treated mice. Importantly, therapeutic inhibitory molecule dual blockade (anti-PD-L1 and anti-lymphocyte-activation gene 3) increased the number of circulating LCMV-specific CD8(+) T cells, and improved CD8(+) T cell function and pathogen control in chronically infected septic mice. Together, these results illustrate that polymicrobial sepsis compromises the overall health of the host leading to increased vulnerability to chronic infection and exacerbated CD8(+) T cell exhaustion. Collectively, our findings suggest that septic survivors may be more susceptible and at greater risk for developing exhaustible CD8(+) T cells upon encountering a subsequent chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Pathogen Interactions/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Sepsis/immunology , Animals , Antibodies/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Convalescence , Disease Susceptibility , Gene Expression Regulation , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Count , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/mortality , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Sepsis/complications , Sepsis/mortality , Sepsis/pathology , Signal Transduction , Signaling Lymphocytic Activation Molecule Family , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Viral Load , Lymphocyte Activation Gene 3 Protein
3.
J Immunol ; 193(12): 5873-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25378592

ABSTRACT

The extent to which obesity compromises the differentiation and maintenance of protective memory CD8 T cell responses and renders obese individuals susceptible to infection remains unknown. In this study, we show that diet-induced obesity did not impact the maintenance of pre-existing memory CD8 T cells, including acquisition of a long-term memory phenotype (i.e., CD27(hi), CD62L(hi), KLRG1(lo)) and function (i.e., cytokine production, secondary expansion, and memory CD8 T cell-mediated protection). Additionally, obesity did not influence the differentiation and maintenance of newly evoked memory CD8 T cell responses in inbred and outbred hosts generated in response to different types of systemic (LCMV, L. monocytogenes) and/or localized (influenza virus) infections. Interestingly, the rate of naive-to-memory CD8 T cell differentiation after a peptide-coated dendritic cell immunization was similar in lean and obese hosts, suggesting that obesity-associated inflammation, unlike pathogen- or adjuvant-induced inflammation, did not influence the development of endogenous memory CD8 T cell responses. Therefore, our studies reveal that the obese environment does not influence the development or maintenance of memory CD8 T cell responses that are either primed before or after obesity is established, a surprising notion with important implications for future studies aiming to elucidate the role obesity plays in host susceptibility to infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diet/adverse effects , Immunologic Memory/immunology , Obesity/etiology , Animals , Antigens/immunology , Bacterial Infections/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Diet, High-Fat/adverse effects , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Immunophenotyping , Inflammation/immunology , Lymphocyte Activation/drug effects , Male , Mice , Obesity/immunology , Peptides/immunology , Phenotype , Virus Diseases/immunology
4.
Proc Natl Acad Sci U S A ; 107(36): 15904-9, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20798038

ABSTRACT

In nonneuronal cells, herpes simplex virus 1 overcomes host defenses, replicates, and ultimately kills the infected cell. Among the host defenses suppressed by the virus is a repressor complex whose key components are histone deacetylase (HDAC) 1 or 2, RE-1 silencing transcription factor (REST), corepressor of REST (CoREST), and lysine-specific demethylase (LSD) 1. In neurons innervating cells at the portal of entry into the body, the virus establishes a "latent" infection in which viral DNA is silenced with the exception of a family of genes. The question posed here is whether the virus hijacks this repressor complex to silence itself in neurons during the latent state. To test this hypothesis, we inserted into the wild-type virus genome a wild-type REST [recombinant (R) 111], a dominant-negative REST (dnREST) lacking the N- and C-terminal repressor domains (R112), or an insertion control consisting of tandem repeats of stop codons (R113). The recombinant virus R112 carrying the dnREST replicated better and was more virulent than the wild-type parent or the other recombinant viruses when administered by the corneal or i.p. routes. Moreover, in contrast to other recombinants, corneal route inoculation by R112 recombinant virus resulted in higher DNA copy numbers, higher levels of infectious virus in eye, trigeminal ganglion, or brain, and virtually complete destruction of trigeminal ganglia in mice that may ultimately succumb to infection. These results support an earlier conclusion that the HDAC/CoREST/REST/LSD1 repressor complex is a significant component of the host innate immunity and are consistent with the hypothesis that HSV-1 hijacks the repressor to silence itself during latent infection.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , RNA Interference , Repressor Proteins/antagonists & inhibitors , Simplexvirus/pathogenicity , Virulence
5.
Semin Immunopathol ; 37(3): 301-10, 2015 May.
Article in English | MEDLINE | ID: mdl-25860798

ABSTRACT

Memory CD8 T cells play a critical role in providing protection to immune hosts by orchestrating rapid elimination of pathogen-infected cells after re-infection. Systemic bacterial infection with Listeria monocytogenes has been a favored approach for researchers to characterize pathogen-specific CD8 T cell responses, and in-depth understanding of L. monocytogenes biology has provided invaluable experimental tools that have been used to increase our understanding of memory CD8 T cell differentiation. Here, we describe how the tools from this murine model system of infection have been utilized to characterize pathogen-specific CD8 T cells in inbred and genetically diverse outbred hosts as they undergo naïve-to-memory CD8 T cell differentiation in vivo. We also discuss how studying L. monocytogenes-evoked CD8 T cell responses have provided insight on the degree of diminished T cell immunity in clinically relevant conditions such as sepsis and obesity. Overall, this review will highlight how infection with the intracellular pathogen L. monocytogenes has enabled analysis of systemic CD8 T cell responses and greatly contributed to what is known about memory CD8 T cell generation and differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Host-Pathogen Interactions/immunology , Immunologic Memory , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Animals , CD8-Positive T-Lymphocytes/metabolism , Humans , Listeriosis/metabolism , Lymphocyte Activation/immunology , Species Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL