Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(4): 112410, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37071533

ABSTRACT

Genital herpes is characterized by recurrent episodes of epithelial blistering. The mechanisms causing this pathology are ill defined. Using a mouse model of vaginal herpes simplex virus 2 (HSV-2) infection, we show that interleukin-18 (IL-18) acts upon natural killer (NK) cells to promote accumulation of the serine protease granzyme B in the vagina, coinciding with vaginal epithelial ulceration. Genetic loss of granzyme B or therapeutic inhibition by a specific protease inhibitor reduces disease and restores epithelial integrity without altering viral control. Distinct effects of granzyme B and perforin deficiency on pathology indicates that granzyme B acts independent of its classic cytotoxic role. IL-18 and granzyme B are markedly elevated in human herpetic ulcers compared with non-herpetic ulcers, suggesting engagement of these pathways in HSV-infected patients. Our study reveals a role for granzyme B in destructing mucosal epithelium during HSV-2 infection, identifying a therapeutic target to augment treatment of genital herpes.


Subject(s)
Herpes Genitalis , Herpes Simplex , Female , Humans , Granzymes/metabolism , Herpesvirus 2, Human/metabolism , Interleukin-18 , Killer Cells, Natural/metabolism , Ulcer , Vagina
2.
Mucosal Immunol ; 16(1): 3-4, 2023 02.
Article in English | MEDLINE | ID: mdl-36640865

ABSTRACT

Tissue-resident memory T (TRM) cells exert antiviral effects in situ using a variety of cell-intrinsic mechanisms. In this issue of Mucosal Immunology, Rosato and colleagues demonstrate that mucosal CD8 TRM cells can also facilitate the extravasation of antibodies with neutralizing capacities by modulating transcytotic receptors and vascular permeability, thus revealing a new mechanism by which TRM cells modulate humoral responses and protect the host from incoming pathogens.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Memory T Cells , Immunity, Cellular , Mucous Membrane
3.
Elife ; 102021 05 28.
Article in English | MEDLINE | ID: mdl-34047696

ABSTRACT

Neutrophil responses against pathogens must be balanced between protection and immunopathology. Factors that determine these outcomes are not well-understood. In a mouse model of genital herpes simplex virus-2 (HSV-2) infection, which results in severe genital inflammation, antibody-mediated neutrophil depletion reduced disease. Comparative single-cell RNA-sequencing analysis of vaginal cells against a model of genital HSV-1 infection, which results in mild inflammation, demonstrated sustained expression of interferon-stimulated genes (ISGs) only after HSV-2 infection primarily within the neutrophil population. Both therapeutic blockade of IFNα/ß receptor 1 (IFNAR1) and genetic deletion of IFNAR1 in neutrophils concomitantly decreased HSV-2 genital disease severity and vaginal IL-18 levels. Therapeutic neutralization of IL-18 also diminished genital inflammation, indicating an important role for this cytokine in promoting neutrophil-dependent immunopathology. Our study reveals that sustained type I interferon (IFN) signaling is a driver of pathogenic neutrophil responses and identifies IL-18 as a novel component of disease during genital HSV-2 infection.


Herpes simplex virus (HSV) is a human pathogen that causes genital herpes, an incurable disease that results in recurrent sores and inflammation. Infection with HSV induces a strong antiviral immune response, which results in large numbers of immune cells arriving at these lesions. But while some of these cells help to control viral replication, others might contribute to the inflammation that drives the disease. One of the first immune cells to respond to infection are neutrophils. Although neutrophils are generally protective, especially against bacteria and fungi, they have also been implicated in tissue damage and severe inflammation during viral infections. But what determines whether a neutrophil will help to fight off an infection or increase disease severity is still an open question. To investigate this, Lebratti, Lim et al. studied mice that had been infected with the genital herpes virus HSV-2, which is known to cause significant amounts of inflammation in mice. The experiments revealed that a signaling molecule called type I interferon, which is thought to be antiviral, causes neutrophils at the site of the infection to produce proteins, such as IL-18, which trigger an inflammatory reaction. Lebratti, Lim et al. found that type I interferon and IL-18 had shifting roles during the course of infection. In the early stages, both molecules had a protective effect, confirming results from previous studies. However, as the infection progressed, sustained levels of type I interferon signaling in neutrophils led to excess amounts of IL-18. Lebratti, Lim et al. discovered that blocking interferon signaling or decreasing the levels of IL-18 later during infection unexpectedly reduced the severity of the disease and resulted in less genital tissue damage. Further experiments also showed that mice infected with another genital herpes virus called HSV-1 did not experience sustained levels of type I interferon. This may explain why this virus causes less severe disease in mice. Understanding how the immune system reacts to viruses could reveal new targets for treatments of genital herpes. At the moment, there is little information about IL-18 production during genital herpes in humans. So, the next step is to see whether neutrophils behave in the same way and whether IL-18 can be detected during human disease. It is possible that the same immune components could promote disease in other infections too. If so, this work may help uncover new drug targets for other viral diseases.


Subject(s)
Herpes Genitalis/virology , Herpesvirus 2, Human/pathogenicity , Immunity, Mucosal , Interferon Type I/metabolism , Interleukin-18/metabolism , Mucous Membrane/virology , Neutrophil Activation , Neutrophils/virology , Vagina/virology , Animals , Antibodies/pharmacology , Chlorocebus aethiops , Disease Models, Animal , Female , Herpes Genitalis/immunology , Herpes Genitalis/metabolism , Herpes Genitalis/prevention & control , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/immunology , Host-Pathogen Interactions , Immunity, Mucosal/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/drug effects , Mucous Membrane/innervation , Mucous Membrane/metabolism , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Vagina/drug effects , Vagina/immunology , Vagina/metabolism , Vero Cells
4.
mSphere ; 5(5)2020 09 02.
Article in English | MEDLINE | ID: mdl-32878932

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected millions within just a few months, causing severe respiratory disease and mortality. Assays to monitor SARS-CoV-2 growth in vitro depend on time-consuming and costly RNA extraction steps, hampering progress in basic research and drug development efforts. Here, we developed a simplified quantitative real-time PCR assay that bypasses viral RNA extraction steps and can monitor SARS-CoV-2 growth from a small amount of cell culture supernatants. In addition, we show that this approach is easily adaptable to numerous other RNA and DNA viruses. Using this assay, we screened the activities of a number of compounds that were predicted to alter SARS-CoV-2 entry and replication as well as HIV-1-specific drugs in a proof-of-concept study. We found that E64D (inhibitor of endosomal proteases cathepsin B and L) and apilimod (endosomal trafficking inhibitor) potently decreased the amount of SARS-CoV-2 RNA in cell culture supernatants with minimal cytotoxicity. Surprisingly, we found that the macropinocytosis inhibitor ethylisopropylamiloride (EIPA) similarly decreased SARS-CoV-2 RNA levels in supernatants, suggesting that entry may additionally be mediated by an alternative pathway. HIV-1-specific inhibitors nevirapine (a nonnucleoside reverse transcriptase inhibitor [NNRTI]), amprenavir (a protease inhibitor), and allosteric integrase inhibitor 2 (ALLINI-2) modestly inhibited SARS-CoV-2 replication, albeit the 50% inhibitory concentration (IC50) values were much higher than that required for HIV-1. Taking the data together, this simplified assay will expedite basic SARS-CoV-2 research, be amenable to mid-throughput screening assays (i.e., drug, CRISPR, small interfering RNA [siRNA], etc.), and be applicable to a broad number of RNA and DNA viruses.IMPORTANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of the coronavirus disease 2019 (COVID-19) pandemic, is continuing to cause immense respiratory disease and social and economic disruptions. Conventional assays that monitor SARS-CoV-2 growth in cell culture rely on costly and time-consuming RNA extraction procedures, hampering progress in basic SARS-CoV-2 research and development of effective therapeutics. Here, we developed a simple quantitative real-time PCR assay to monitor SARS-CoV-2 growth in cell culture supernatants that does not necessitate RNA extraction and that is as accurate and sensitive as existing methods. In a proof-of-concept screen, we found that E64D, apilimod, EIPA, and remdesivir can substantially impede SARS-Cov-2 replication, providing novel insight into viral entry and replication mechanisms. In addition, we show that this approach is easily adaptable to numerous other RNA and DNA viruses. This simplified assay will undoubtedly expedite basic SARS-CoV-2 and virology research and be amenable to use in drug screening platforms to identify therapeutics against SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Betacoronavirus/growth & development , Cell Culture Techniques/methods , Coronavirus Infections/virology , Pneumonia, Viral/virology , Real-Time Polymerase Chain Reaction/methods , Betacoronavirus/genetics , Betacoronavirus/pathogenicity , COVID-19 , Pandemics , RNA, Viral/analysis , RNA, Viral/isolation & purification , SARS-CoV-2 , Virus Replication/drug effects
5.
JCI Insight ; 5(5)2020 03 12.
Article in English | MEDLINE | ID: mdl-32161194

ABSTRACT

Herpes simplex virus-2 (HSV-2) and HSV-1 both can cause genital herpes, a chronic infection that establishes a latent reservoir in the nervous system. Clinically, the recurrence frequency of HSV-1 genital herpes is considerably less than HSV-2 genital herpes, which correlates with reduced neuronal infection. The factors dictating the disparate outcomes of HSV-1 and HSV-2 genital herpes are unclear. In this study, we show that vaginal infection of mice with HSV-1 leads to the rapid appearance of mature DCs in the draining lymph node, which is dependent on an early burst of NK cell-mediated IFN-γ production in the vagina that occurs after HSV-1 infection but not HSV-2 infection. Rapid DC maturation after HSV-1 infection, but not HSV-2 infection, correlates with the accelerated generation of a neuroprotective T cell response and early accumulation of IFN-γ-producing T cells at the site of infection. Depletion of T cells or loss of IFN-γ receptor (IFN-γR) expression in sensory neurons both lead to a marked loss of neuroprotection only during HSV-1, recapitulating a prominent feature of HSV-2 infection. Our experiments reveal key differences in host control of neuronal HSV-1 and HSV-2 infection after genital exposure of mice, and they define parameters of a successful immune response against genital herpes.


Subject(s)
Herpes Simplex/immunology , Nervous System Diseases/immunology , Nervous System Diseases/virology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Female , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/pathogenicity , Host-Pathogen Interactions , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL
6.
J Virol ; 93(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31462571

ABSTRACT

Type III interferon (IFN), or IFN lambda (IFN-λ), is an essential component of the innate immune response to mucosal viral infections. In both the intestine and the lung, signaling via the IFN-λ receptor (IFNLR) controls clinically important viral pathogens, including influenza virus, norovirus, and rotavirus. While it is thought that IFN-λ cytokines are the exclusive ligands for signaling through IFNLR, it is not known whether genetic ablation of these cytokines phenotypically recapitulates disruption of the receptor. Here, we report the serendipitous establishment of Ifnl2-/- Ifnl3-/- mice, which lack all known functional murine IFN-λ cytokines. We demonstrate that, like Ifnlr1-/- mice lacking IFNLR signaling, these mice display defective control of murine norovirus, reovirus, and influenza virus and therefore genocopy Ifnlr1-/- mice. Thus, for regulation of viral infections at mucosal sites of both the intestine and lung, signaling via IFNLR can be fully explained by the activity of known cytokines IFN-λ2 and IFN-λ3. Our results confirm the current understanding of ligand-receptor interactions for type III IFN signaling and highlight the importance of this pathway in regulation of mucosal viral pathogens.IMPORTANCE Type III interferons are potent antiviral cytokines important for regulation of viruses that infect at mucosal surfaces. Studies using mice lacking the Ifnlr1 gene encoding the type III interferon receptor have demonstrated that signaling through this receptor is critical for protection against influenza virus, norovirus, and reovirus. Using a genetic approach to disrupt murine type III interferon cytokine genes Ifnl2 and Ifnl3, we found that mice lacking these cytokines fully recapitulate the impaired control of viruses observed in mice lacking Ifnlr1 Our results support the idea of an exclusive role for known type III interferon cytokines in signaling via IFNLR to mediate antiviral effects at mucosal surfaces. These findings emphasize the importance of type III interferons in regulation of a variety of viral pathogens and provide important genetic evidence to support our understanding of the ligand-receptor interactions in this pathway.


Subject(s)
Cytokines/genetics , Interferons/genetics , Interleukins/genetics , Animals , Cell Line , Cytokines/metabolism , Female , Immunity, Innate , Interferons/metabolism , Interleukins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Virus Diseases/metabolism , Interferon Lambda
7.
PLoS One ; 14(7): e0219941, 2019.
Article in English | MEDLINE | ID: mdl-31329630

ABSTRACT

Escherichia coli infection of the female reproductive tract is a significant cause of disease in humans and animals, but simple animal models are lacking. Here we report that vaginal inoculation of uropathogenic E. coli strains UTI89 and CFT073 in non-pregnant, estrogen-treated mice resulted in robust colonization of the vagina and uterine horns, whereas titers of the lab strain MG1655 were significantly lower. Non-estrogenized mice also became colonized, but there was more variation in titers. A dose of 104 colony-forming units (CFU) UTI89 was sufficient to result in colonization in all estrogenized mice, and we also observed bacterial transfer between inoculated and uninoculated estrogenized cage mates. UTI89 infection led to inflammation and leukocyte infiltration into the uterine horns as evidenced by tissue histology. Flow cytometry experiments revealed that neutrophil, monocyte and eosinophil populations were significantly increased in infected uterine horns. This model is a simple way to study host-pathogen interactions in E. coli vaginal colonization and uterine infection. There are immediate implications for investigators studying urinary tract infection using mouse models, as few E. coli are required to achieve reproductive colonization, resulting in an additional, underappreciated mucosal reservoir.


Subject(s)
Escherichia coli Infections/microbiology , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Uterine Diseases/microbiology , Animals , Colony Count, Microbial , Escherichia coli Infections/pathology , Female , Mice , Mice, Inbred C57BL , Uterine Diseases/pathology , Uterus/microbiology , Uterus/pathology
8.
J Virol ; 92(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29343577

ABSTRACT

Zika virus (ZIKV), which can cause devastating disease in fetuses of infected pregnant women, can be transmitted by mosquito inoculation and sexual routes. Little is known about immune protection against sexually transmitted ZIKV. In this study, we show that previous infection through intravaginal or subcutaneous routes with a contemporary Brazilian strain of ZIKV can protect against subsequent intravaginal challenge with a homologous strain. Both routes of inoculation induced high titers of ZIKV-specific and neutralizing antibody in serum and the vaginal lumen. Virus-specific T cells were recruited to and retained in the female reproductive tract after intravaginal and subcutaneous ZIKV infection. Studies in mice with genetic or acquired deficiencies in B and/or T cells demonstrated that both lymphocyte populations redundantly protect against intravaginal challenge in ZIKV-immune animals. Passive transfer of ZIKV-immune IgG or T cells significantly limited intravaginal infection of naive mice, although antibody more effectively prevented dissemination throughout the reproductive tract. Collectively, our experiments begin to establish the immune correlates of protection against intravaginal ZIKV infection, which should inform vaccination strategies in nonpregnant and pregnant women.IMPORTANCE The recent ZIKV epidemic resulted in devastating outcomes in fetuses and may affect reproductive health. Unlike other flaviviruses, ZIKV can be spread by sexual contact as well as a mosquito vector. While previous studies have identified correlates of protection for mosquito-mediated infection, few have focused on immunity against sexual transmission. As exposure to ZIKV via mosquito bite has likely occurred to many living in areas where ZIKV is endemic, our study addresses whether this route of infection can protect against subsequent sexual exposure. We demonstrate that subcutaneous ZIKV infection can protect against subsequent vaginal infection by generating both local antiviral T cell and antibody responses. Our research begins to define the immune correlates of protection for ZIKV infection in the vagina and provides a foundation for testing ZIKV vaccines against sexual transmission.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/immunology , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Vagina/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Cells, Cultured , Female , Immunity, Humoral , Mice , Mice, Inbred C57BL , Vagina/drug effects , Vagina/virology , Zika Virus Infection/immunology , Zika Virus Infection/virology
9.
Curr Opin Virol ; 28: 61-67, 2018 02.
Article in English | MEDLINE | ID: mdl-29175730

ABSTRACT

Memory T cells are an important component of the adaptive immune response. Tissue-resident memory T cells (TRM) are a recently described subset of memory T cells that reside in peripheral tissues and are maintained independently of circulating subsets of memory T cells. Importantly, TRM are frequently found in barrier tissues that commonly serve as entry portals for pathogens such as viruses. Mounting evidence shows that TRM are superior to their circulating counterparts in conferring protective immunity against a wide range of viruses. In this review, we will discuss the role of TRM in controlling viral infection with a focus on CD8+ TRM, the factors that regulate differentiation and a potential role for TRM in future vaccine development.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Virus Diseases/immunology , Adaptive Immunity , Animals , Cell Differentiation/immunology , Humans , Mice , Viruses
10.
Nat Commun ; 7: 13346, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27827367

ABSTRACT

Tissue-resident memory CD8+ T (CD8 TRM) cells are an essential component of protective immune responses at barrier tissues, including the female genital tract. However, the mechanisms that lead to the initiation of CD8 TRM-mediated protective immunity after viral infection are unclear. Here we report that CD8 TRM cells established by 'prime and pull' method confer protection against genital HSV-2 infection, and that IFN-γ produced by CD8 TRM cells is required for this protection. Furthermore, we find that CD8 TRM-cell restimulation depends on a population of CD301b+ antigen-presenting cells (APC) in the lamina propria. Elimination of MHC class I on CD301b+ dendritic cells abrogates protective immunity, suggesting the requirement for cognate antigen presentation to CD8 TRM cells by CD301b+ dendritic cells. These results define the requirements for CD8 TRM cells in protection against genital HSV-2 infection and identify the population of APC that are responsible for activating these cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Herpes Genitalis/immunology , Herpesvirus 2, Human/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/transplantation , Dendritic Cells/metabolism , Female , Herpes Genitalis/therapy , Herpes Genitalis/virology , Herpesvirus 2, Human/physiology , Immunologic Memory/immunology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Vagina/immunology , Vagina/virology
11.
Immunity ; 40(5): 801-13, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24837104

ABSTRACT

Epidemiological evidence suggests that chronic infections impair immune responses to unrelated pathogens and vaccines. The underlying mechanisms, however, are unclear and distinguishing effects on priming versus development of immunological memory has been challenging. We investigated whether bystander chronic infections impact differentiation of memory CD8(+) T cells, the hallmark of protective immunity against intracellular pathogens. Chronic bystander infections impaired development of memory CD8(+) T cells in several mouse models and humans. These effects were independent of initial priming and were associated with chronic inflammatory signatures. Chronic inflammation negatively impacted the number of bystander CD8(+) T cells and their memory development. Distinct underlying mechanisms of altered survival and differentiation were revealed with the latter regulated by the transcription factors T-bet and Blimp-1. Thus, exposure to prolonged bystander inflammation impairs the effector to memory transition. These data have relevance for immunity and vaccination during persisting infections and chronic inflammation.


Subject(s)
Bacterial Infections/immunology , Bystander Effect/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Virus Diseases/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Chronic Disease , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , T-Box Domain Proteins/immunology , Transcription Factors/immunology
12.
Trends Immunol ; 34(10): 487-94, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24012144

ABSTRACT

Genital herpes is an incurable, chronic disease that affects millions of people worldwide. Not only does genital herpes cause painful, recurrent symptoms, it is also a significant risk factor for the acquisition of other sexually transmitted infections such as HIV-1. Antiviral drugs are used to treat herpes simplex virus (HSV) infection, but they cannot stop viral shedding and transmission. Thus, developing a vaccine that can prevent or clear infection will be crucial in limiting the spread of disease. In this review we outline recent studies that improve our understanding of host responses against HSV infection, discuss past clinical vaccine trials, and highlight new strategies for vaccine design against genital herpes.


Subject(s)
Herpes Genitalis/immunology , Herpes Genitalis/prevention & control , Viral Vaccines/immunology , Animals , Humans
13.
Immunol Rev ; 255(1): 165-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947354

ABSTRACT

Tissues such as the genital tract, skin, and lung act as barriers against invading pathogens. To protect the host, incoming microbes must be quickly and efficiently controlled by the immune system at the portal of entry. Memory is a hallmark of the adaptive immune system, which confers long-term protection and is the basis for efficacious vaccines. While the majority of existing vaccines rely on circulating antibody for protection, struggles to develop antibody-based vaccines against infections such as herpes simplex virus (HSV) and human immunodeficiency virus (HIV) have underscored the need to generate memory T cells for robust antiviral control. The circulating memory T-cell population is generally divided into two subsets: effector memory (TEM ) and central memory (TCM ). These two subsets can be distinguished by their localization, as TCM home to secondary lymphoid organs and TEM circulate through non-lymphoid tissues. More recently, studies have identified a third subset, called tissue-resident memory (TRM ) cells, based on its migratory properties. This subset is found in peripheral tissues that require expression of specific chemoattractants and homing receptors for T-cell recruitment and retention, including barrier sites such as the skin and genital tract. In this review, we categorize different tissues in the body based on patterns of memory T-cell migration and tissue residency. This review also describes the rules for TRM generation and the properties that distinguish them from circulating TEM and TCM cells. Finally, based on the failure of recent T-cell-based vaccines to provide optimal protection, we also discuss the potential role of TRM cells in vaccine design against microbes that invade through the peripheral tissues and highlight new vaccination strategies that take advantage of this newly described memory T-cell subset.


Subject(s)
Immunologic Memory , T-Lymphocyte Subsets/immunology , Animals , Cell Movement/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Lymphocyte Activation/immunology , Phenotype , T-Lymphocyte Subsets/metabolism , Vaccines/immunology , Virus Diseases/immunology , Virus Diseases/metabolism , Viruses/immunology
14.
Nature ; 491(7424): 463-7, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23075848

ABSTRACT

Most successful existing vaccines rely on neutralizing antibodies, which may not require specific anatomical localization of B cells. However, efficacious vaccines that rely on T cells for protection have been difficult to develop, as robust systemic memory T-cell responses do not necessarily correlate with host protection. In peripheral sites, tissue-resident memory T cells provide superior protection compared to circulating memory T cells. Here we describe a simple and non-inflammatory vaccine strategy that enables the establishment of a protective memory T-cell pool within peripheral tissue. The female genital tract, which is a portal of entry for sexually transmitted infections, is an immunologically restrictive tissue that prevents entry of activated T cells in the absence of inflammation or infection. To overcome this obstacle, we developed a vaccine strategy that we term 'prime and pull' to establish local tissue-resident memory T cells at a site of potential viral exposure. This approach relies on two steps: conventional parenteral vaccination to elicit systemic T-cell responses (prime), followed by recruitment of activated T cells by means of topical chemokine application to the restrictive genital tract (pull), where such T cells establish a long-term niche and mediate protective immunity. In mice, prime and pull protocol reduces the spread of infectious herpes simplex virus 2 into the sensory neurons and prevents development of clinical disease. These results reveal a promising vaccination strategy against herpes simplex virus 2, and potentially against other sexually transmitted infections such as human immunodeficiency virus.


Subject(s)
Herpes Genitalis/immunology , Herpes Genitalis/prevention & control , Immunologic Memory/immunology , Vaccination , Viral Vaccines/immunology , Administration, Topical , Animals , Antibodies, Viral/analysis , Cell Count , Chemokine CXCL10/administration & dosage , Chemokine CXCL9/administration & dosage , Female , Mice , Mice, Inbred C57BL , Survival Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Vagina/immunology , Viral Load
15.
Cell Res ; 22(9): 1325-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22565287

ABSTRACT

Tissue-resident memory T cells (TRM) are a new subset of memory cells that have been associated with enhanced protective immunity for their tissue of residence. A recent study by Jiang et al. sheds light on the migration behavior of TRM in both infected and unifected skin, and their ability to provide protection against re-infection even within a previously uninfected skin tissue.


Subject(s)
Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , E-Selectin/metabolism , Mice , P-Selectin/metabolism , Skin/metabolism , T-Lymphocytes, Regulatory/metabolism , Vaccinia virus/pathogenicity
16.
Blood ; 116(25): 5560-70, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-20884806

ABSTRACT

The requirements for tonic T-cell receptor (TCR) signaling in CD8(+) memory T-cell generation and homeostasis are poorly defined. The SRC homology 2 (SH2)-domain-containing leukocyte protein of 76 kDa (SLP-76) is critical for proximal TCR-generated signaling. We used temporally mediated deletion of SLP-76 to interrupt tonic and activating TCR signals after clearance of the lymphocytic choriomeningitis virus (LCMV). SLP-76-dependent signals are required during the contraction phase of the immune response for the normal generation of CD8 memory precursor cells. Conversely, LCMV-specific memory CD8 T cells generated in the presence of SLP-76 and then acutely deprived of TCR-mediated signals persist in vivo in normal numbers for more than 40 weeks. Tonic TCR signals are not required for the transition of the memory pool toward a central memory phenotype, but the absence of SLP-76 during memory homeostasis substantially alters the kinetics. Our data are consistent with a model in which tonic TCR signals are required at multiple stages of differentiation, but are dispensable for memory CD8 T-cell persistence.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Immunologic Memory , Phosphoproteins/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Female , Flow Cytometry , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
17.
Nat Med ; 16(10): 1147-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20890291

ABSTRACT

CD8(+) T cells in chronic viral infections such as HIV develop functional defects including loss of interleukin-2 (IL-2) secretion and decreased proliferative potential that are collectively termed 'exhaustion'. Exhausted T cells express increased amounts of multiple inhibitory receptors, such as programmed death-1 (PD-1), that contribute to impaired virus-specific T cell function. Although reversing PD-1 inhibition is therefore an attractive therapeutic strategy, the cellular mechanisms by which PD-1 ligation results in T cell inhibition are not fully understood. PD-1 is thought to limit T cell activation by attenuating T cell receptor (TCR) signaling. It is not known whether PD-1 also acts by upregulating genes in exhausted T cells that impair their function. Here we analyzed gene expression profiles from HIV-specific CD8(+) T cells in individuals with HIV and show that PD-1 coordinately upregulates a program of genes in exhausted CD8(+) T cells from humans and mice. This program includes upregulation of basic leucine transcription factor, ATF-like (BATF), a transcription factor in the AP-1 family. Enforced expression of BATF was sufficient to impair T cell proliferation and cytokine secretion, whereas BATF knockdown reduced PD-1 inhibition. Silencing BATF in T cells from individuals with chronic viremia rescued HIV-specific T cell function. Thus, inhibitory receptors can cause T cell exhaustion by upregulating genes--such as BATF--that inhibit T cell function. Such genes may provide new therapeutic opportunities to improve T cell immunity to HIV.


Subject(s)
Antigens, CD/physiology , Apoptosis Regulatory Proteins/physiology , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Profiling , HIV/immunology , T-Lymphocytes/physiology , Animals , Gene Expression Regulation , Humans , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lymphocytic Choriomeningitis/metabolism , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor
18.
J Virol ; 84(4): 2078-89, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19955307

ABSTRACT

The PD-1/PD-L pathway plays a major role in regulating T-cell exhaustion during chronic viral infections in animal models, as well as in humans, and blockade of this pathway can revive exhausted CD8(+) T cells. We examined the expression of PD-1 and its ligands, PD-L1 and PD-L2, in multiple tissues during the course of chronic viral infection and determined how the amount of PD-1 expressed, as well as the anatomical location, influenced the function of exhausted CD8 T cells. The amount of PD-1 on exhausted CD8 T cells from different anatomical locations did not always correlate with infectious virus but did reflect viral antigen in some tissues. Moreover, lower expression of PD-L1 in some locations, such as the bone marrow, favored the survival of PD-1(Hi) exhausted CD8 T cells, suggesting that some anatomical sites might provide a survival niche for subpopulations of exhausted CD8 T cells. Tissue-specific differences in the function of exhausted CD8 T cells were also observed. However, while cytokine production did not strictly correlate with the amount of PD-1 expressed by exhausted CD8 T cells from different tissues, the ability to degranulate and kill were tightly linked to PD-1 expression regardless of the anatomical location. These observations have implications for human chronic infections and for therapeutic interventions based on blockade of the PD-1 pathway.


Subject(s)
Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/metabolism , B7-1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Membrane Glycoproteins/metabolism , Peptides/metabolism , Animals , B7-H1 Antigen , Bone Marrow/immunology , Bone Marrow/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Survival , Chronic Disease , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Humans , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Organ Specificity , Programmed Cell Death 1 Receptor , Signal Transduction/immunology , Spleen/immunology , Spleen/metabolism , Time Factors , Virus Replication
19.
Immunity ; 31(2): 309-20, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19664943

ABSTRACT

T cell exhaustion is common during chronic infections and can prevent optimal immunity. Although recent studies have demonstrated the importance of inhibitory receptors and other pathways in T cell exhaustion, the underlying transcriptional mechanisms are unknown. Here, we define a role for the transcription factor Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection. Blimp-1 repressed key aspects of normal memory CD8(+) T cell differentiation and promoted high expression of inhibitory receptors during chronic infection. These cardinal features of CD8(+) T cell exhaustion were corrected by conditionally deleting Blimp-1. Although high expression of Blimp-1 fostered aspects of CD8(+) T cell exhaustion, haploinsufficiency indicated that moderate Blimp-1 expression sustained some effector function during chronic viral infection. Thus, we identify Blimp-1 as a transcriptional regulator of CD8(+) T cell exhaustion during chronic viral infection and propose that Blimp-1 acts as a transcriptional rheostat balancing effector function and T cell exhaustion.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Transcription Factors/metabolism , Virus Diseases/immunology , Acute Disease , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Surface/immunology , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Chronic Disease , Cytotoxicity, Immunologic/immunology , GPI-Linked Proteins , Granzymes/immunology , Granzymes/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1 , Programmed Cell Death 1 Receptor , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family , Transcription Factors/genetics , Virus Diseases/genetics , Lymphocyte Activation Gene 3 Protein
20.
Nat Immunol ; 10(1): 29-37, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19043418

ABSTRACT

T cell exhaustion often occurs during chronic infection and prevents optimal viral control. The molecular pathways involved in T cell exhaustion remain poorly understood. Here we show that exhausted CD8+ T cells are subject to complex layers of negative regulation resulting from the coexpression of multiple inhibitory receptors. Exhausted CD8+ T cells expressed up to seven inhibitory receptors. Coexpression of multiple distinct inhibitory receptors was associated with greater T cell exhaustion and more severe infection. Regulation of T cell exhaustion by various inhibitory pathways was nonredundant, as blockade of the T cell inhibitory receptors PD-1 and LAG-3 simultaneously and synergistically improved T cell responses and diminished viral load in vivo. Thus, CD8+ T cell responses during chronic viral infections are regulated by complex patterns of coexpressed inhibitory receptors.


Subject(s)
Antigens, CD/metabolism , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Receptors, Immunologic/metabolism , Animals , CD8-Positive T-Lymphocytes/virology , Chronic Disease , Disease Models, Animal , Down-Regulation , Immunologic Memory , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Lymphocyte Activation Gene 3 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...