Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Rep ; 28(12): 3092-3104.e5, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31533033

ABSTRACT

The immune system adapts to constitutive antigens to preserve self-tolerance, which is a major barrier for anti-tumor immunity. Antigen-specific reversal of tolerance constitutes a major goal to spur therapeutic applications. Here, we show that robust, iterative, systemic stimulation targeting tissue-specific antigens in the context of acute infections reverses established CD8+ T cell tolerance to self, including in T cells that survive negative selection. This strategy results in large numbers of circulating and resident memory self-specific CD8+ T cells that are widely distributed and can be co-opted to control established malignancies bearing self-antigen without concomitant autoimmunity. Targeted expansion of both self- and tumor neoantigen-specific T cells acts synergistically to boost anti-tumor immunity and elicits protection against aggressive melanoma. Our findings demonstrate that T cell tolerance can be re-adapted to responsiveness through robust antigenic exposure, generating self-specific CD8+ T cells that can be used for cancer treatment.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Immunity, Cellular , Melanoma/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Transgenic
2.
J Immunol ; 202(10): 2945-2956, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30988115

ABSTRACT

Imprime PGG (Imprime) is an i.v. administered, yeast ß-1,3/1,6 glucan in clinical development with checkpoint inhibitors. Imprime-mediated innate immune activation requires immune complex formation with naturally occurring IgG anti-ß glucan Abs (ABA). We administered Imprime to healthy human volunteers to assess the necessity of ABA for Imprime-mediated immunopharmacodynamic (IPD) changes. Imprime (4 mg/kg) was administered i.v. in single and multiple infusions. Subsets of subjects were premedicated with antihistamine and corticosteroid. Peripheral blood was measured before, during and after Imprime administration for IPD changes (e.g., ABA, circulating immune complexes, complement activation, complete blood counts, cytokine/chemokine, and gene expression changes). IPD changes were analyzed based on pretreatment serum ABA levels: low-ABA (<20 µg/ml), mid-ABA (≥20-50 µg/ml), and high-ABA (≥50 µg/ml). At the end of infusion, free serum ABA levels decreased, circulating immune complex levels increased, and complement activation was observed. At ∼1-4 h after end of infusion, increased expression of cytokines/chemokines, a 1.5-4-fold increase in neutrophil and monocyte counts and a broad activation of innate immune genes were observed. Low-ABA subjects typically showed minimal IPD changes except when ABA levels rose above 20 µg/ml after repeated Imprime dosing. Mild-to-moderate infusion-related reactions occurred in subjects with ABA ≥20 µg/ml. Premedications alleviated some of the infusion-related reactions, but also inhibited cytokine responses. In conclusion, ABA levels, being critical for Imprime-mediated immune activation may provide a plausible, mechanism-based biomarker to identify patients most likely to respond to Imprime-based anticancer immunotherapy.


Subject(s)
Adjuvants, Immunologic , Fungal Polysaccharides , Immunotherapy , Neoplasms , Saccharomyces cerevisiae/chemistry , beta-Glucans , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Antibodies, Fungal/blood , Antibodies, Fungal/immunology , Chemokines/blood , Chemokines/immunology , Female , Fungal Polysaccharides/administration & dosage , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/pharmacokinetics , Humans , Male , Neoplasms/blood , Neoplasms/immunology , Neoplasms/therapy , beta-Glucans/administration & dosage , beta-Glucans/chemistry , beta-Glucans/pharmacokinetics
3.
J Exp Med ; 216(5): 1214-1229, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30923043

ABSTRACT

This study examines the extent to which memory CD4+ T cells share immunosurveillance strategies with CD8+ resident memory T cells (TRM). After acute viral infection, memory CD4+ T cells predominantly used residence to survey nonlymphoid tissues, albeit not as stringently as observed for CD8+ T cells. In contrast, memory CD4+ T cells were more likely to be resident within lymphoid organs than CD8+ T cells. Migration properties of memory-phenotype CD4+ T cells in non-SPF parabionts were similar, generalizing these results to diverse infections and conditions. CD4+ and CD8+ TRM shared overlapping transcriptional signatures and location-specific features, such as granzyme B expression in the small intestine, revealing tissue-specific and migration property-specific, in addition to lineage-specific, differentiation programs. Functionally, mucosal CD4+ TRM reactivation locally triggered both chemokine expression and broad immune cell activation. Thus, residence provides a dominant mechanism for regionalizing CD4+ T cell immunity, and location enforces shared transcriptional, phenotypic, and functional properties with CD8+ T cells.


Subject(s)
Arenaviridae Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Immunologic Surveillance/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Arenaviridae Infections/virology , Cell Movement/immunology , Chimera/immunology , Female , Granzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , RNA-Seq , Transcriptome
4.
Nature ; 532(7600): 512-6, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-27096360

ABSTRACT

Our current understanding of immunology was largely defined in laboratory mice, partly because they are inbred and genetically homogeneous, can be genetically manipulated, allow kinetic tissue analyses to be carried out from the onset of disease, and permit the use of tractable disease models. Comparably reductionist experiments are neither technically nor ethically possible in humans. However, there is growing concern that laboratory mice do not reflect relevant aspects of the human immune system, which may account for failures to translate disease treatments from bench to bedside. Laboratory mice live in abnormally hygienic specific pathogen free (SPF) barrier facilities. Here we show that standard laboratory mouse husbandry has profound effects on the immune system and that environmental changes produce mice with immune systems closer to those of adult humans. Laboratory mice--like newborn, but not adult, humans--lack effector-differentiated and mucosally distributed memory T cells. These cell populations were present in free-living barn populations of feral mice and pet store mice with diverse microbial experience, and were induced in laboratory mice after co-housing with pet store mice, suggesting that the environment is involved in the induction of these cells. Altering the living conditions of mice profoundly affected the cellular composition of the innate and adaptive immune systems, resulted in global changes in blood cell gene expression to patterns that more closely reflected the immune signatures of adult humans rather than neonates, altered resistance to infection, and influenced T-cell differentiation in response to a de novo viral infection. These data highlight the effects of environment on the basal immune state and response to infection and suggest that restoring physiological microbial exposure in laboratory mice could provide a relevant tool for modelling immunological events in free-living organisms, including humans.


Subject(s)
Animal Husbandry/methods , Animals, Laboratory/immunology , Animals, Wild/immunology , Environment , Immune System/immunology , Immunity/immunology , Models, Animal , Adult , Animals , Cell Differentiation , Environmental Exposure , Female , Humans , Immunity, Innate/immunology , Immunologic Memory , Infant, Newborn , Male , Mice , Phenotype , Specific Pathogen-Free Organisms , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Virus Diseases/immunology , Virus Diseases/virology
5.
J Immunol ; 196(9): 3920-6, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27001957

ABSTRACT

IL-15 regulates central and effector memory CD8 T cell (TCM and TEM, respectively) homeostatic proliferation, maintenance, and longevity. Consequently, IL-15 availability hypothetically defines the carrying capacity for total memory CD8 T cells within the host. In conflict with this hypothesis, previous observations demonstrated that boosting generates preternaturally abundant TEM that increases the total quantity of memory CD8 T cells in mice. In this article, we provide a potential mechanistic explanation by reporting that boosted circulating TEM do not require IL-15 for maintenance. We also investigated tissue-resident memory CD8 T cells (TRM), which protect nonlymphoid tissues from reinfection. We observed up to a 50-fold increase in the total magnitude of TRM in mouse mucosal tissues after boosting, suggesting that the memory T cell capacity in tissues is flexible and that TRM may not be under the same homeostatic regulation as primary central memory CD8 T cells and TEM Further analysis identified distinct TRM populations that depended on IL-15 for homeostatic proliferation and survival, depended on IL-15 for homeostatic proliferation but not for survival, or did not depend on IL-15 for either process. These observations on the numerical regulation of T cell memory indicate that there may be significant heterogeneity among distinct TRM populations and also argue against the common perception that developing vaccines that confer protection by establishing abundant TEM and TRM will necessarily erode immunity to previously encountered pathogens as the result of competition for IL-15.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interleukin-15/metabolism , Mucous Membrane/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Proliferation , Cells, Cultured , Cytotoxicity, Immunologic , Homeostasis , Immunization, Secondary , Mice , Mice, Inbred C57BL , Viral Vaccines/immunology
6.
Immunity ; 44(1): 155-166, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26750312

ABSTRACT

Exposure to inhaled allergens generates T helper 2 (Th2) CD4(+) T cells that contribute to episodes of inflammation associated with asthma. Little is known about allergen-specific Th2 memory cells and their contribution to airway inflammation. We generated reagents to understand how endogenous CD4(+) T cells specific for a house dust mite (HDM) allergen form and function. After allergen exposure, HDM-specific memory cells persisted as central memory cells in the lymphoid organs and tissue-resident memory cells in the lung. Experimental blockade of lymphocyte migration demonstrated that lung-resident cells were sufficient to induce airway hyper-responsiveness, which depended upon CD4(+) T cells. Investigation into the differentiation of pathogenic Trm cells revealed that interleukin-2 (IL-2) signaling was required for residency and directed a program of tissue homing migrational cues. These studies thus identify IL-2-dependent resident Th2 memory cells as drivers of lung allergic responses.


Subject(s)
Asthma/immunology , Immunologic Memory/immunology , Interleukin-2/immunology , Lung/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Antigens, Dermatophagoides/immunology , Cell Differentiation/immunology , Cell Separation , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyroglyphidae/immunology
7.
Cell ; 161(4): 737-49, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957682

ABSTRACT

Memory CD8 T cells protect against intracellular pathogens by scanning host cell surfaces; thus, infection detection rates depend on memory cell number and distribution. Population analyses rely on cell isolation from whole organs, and interpretation is predicated on presumptions of near complete cell recovery. Paradigmatically, memory is parsed into central, effector, and resident subsets, ostensibly defined by immunosurveillance patterns but in practice identified by phenotypic markers. Because isolation methods ultimately inform models of memory T cell differentiation, protection, and vaccine translation, we tested their validity via parabiosis and quantitative immunofluorescence microscopy of a mouse memory CD8 T cell population. We report three major findings: lymphocyte isolation fails to recover most cells and biases against certain subsets, residents greatly outnumber recirculating cells within non-lymphoid tissues, and memory subset homing to inflammation does not conform to previously hypothesized migration patterns. These results indicate that most host cells are surveyed for reinfection by segregated residents rather than by recirculating cells that migrate throughout the blood and body.


Subject(s)
Arenaviridae Infections/immunology , Immunologic Memory , Lymphocytic choriomeningitis virus/physiology , Monitoring, Immunologic , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Inflammation/immunology , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL
8.
J Leukoc Biol ; 97(2): 217-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25395301

ABSTRACT

Vaccines are desired that maintain abundant memory T cells at nonlymphoid sites of microbial exposure, where they may be anatomically positioned for immediate pathogen interception. Here, we test the impact of antigen persistence on mouse CD8 and CD4 T cell distribution and differentiation by comparing responses to infections with different strains of LCMV that cause either acute or chronic infections. We used in vivo labeling techniques that discriminate between T cells present within tissues and abundant populations that fail to be removed from vascular compartments, despite perfusion. LCMV persistence caused up to ∼30-fold more virus-specific CD8 T cells to distribute to the lung compared with acute infection. Persistent infection also maintained mucosal-homing α4ß7 integrin expression, higher granzyme B expression, alterations in the expression of the TRM markers CD69 and CD103, and greater accumulation of virus-specific CD8 T cells in the large intestine, liver, kidney, and female reproductive tract. Persistent infection also increased LCMV-specific CD4 T cell quantity in mucosal tissues and induced maintenance of CXCR4, an HIV coreceptor. This study clarifies the relationship between viral persistence and CD4 and CD8 T cell distribution and mucosal phenotype, indicating that chronic LCMV infection magnifies T cell migration to nonlymphoid tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Mucosal , Immunologic Memory , Intestinal Mucosa/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Female , Gene Expression Regulation/immunology , Granzymes/immunology , Integrin alpha Chains/immunology , Integrin alpha4/immunology , Integrin beta Chains/immunology , Intestinal Mucosa/pathology , Lectins, C-Type/immunology , Lymphocytic Choriomeningitis/pathology , Mice , Organ Specificity/immunology , Receptors, CXCR4/immunology
9.
Science ; 346(6205): 98-101, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25170049

ABSTRACT

The pathogen recognition theory dictates that, upon viral infection, the innate immune system first detects microbial products and then responds by providing instructions to adaptive CD8 T cells. Here, we show in mice that tissue resident memory CD8 T cells (T(RM) cells), non-recirculating cells located at common sites of infection, can achieve near-sterilizing immunity against viral infections by reversing this flow of information. Upon antigen resensitization within the mouse female reproductive mucosae, CD8(+) T(RM) cells secrete cytokines that trigger rapid adaptive and innate immune responses, including local humoral responses, maturation of local dendritic cells, and activation of natural killer cells. This provided near-sterilizing immunity against an antigenically unrelated viral infection. Thus, CD8(+) T(RM) cells rapidly trigger an antiviral state by amplifying receptor-derived signals from previously encountered pathogens.


Subject(s)
Adaptive Immunity/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Innate/immunology , Immunologic Memory , Virus Diseases/immunology , Animals , Antigens, Viral/immunology , Female , Immunity, Humoral/immunology , Interferon-gamma/immunology , Mice , Mice, Inbred C57BL , Mucous Membrane/immunology , Mucous Membrane/virology , Vascular Cell Adhesion Molecule-1/immunology
10.
J Immunol ; 192(7): 2961-4, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24600038

ABSTRACT

Resident memory CD8 T cells (TRM) are a nonrecirculating subset positioned in nonlymphoid tissues to provide early responses to reinfection. Although TRM are associated with nonlymphoid tissues, we asked whether they populated secondary lymphoid organs (SLO). We show that a subset of virus-specific memory CD8 T cells in SLO exhibit phenotypic signatures associated with TRM, including CD69 expression. Parabiosis revealed that SLO CD69(+) memory CD8 T cells do not circulate, defining them as TRM. SLO TRM were overrepresented in IL-15-deficient mice, suggesting independent regulation compared with central memory CD8 T cells and effector memory CD8 T cells. These cells were positioned at SLO entry points for peripheral Ags: the splenic marginal zone, red pulp, and lymph node sinuses. Consistent with a potential role in guarding SLO pathogen entry points, SLO TRM did not vacate their position in response to peripheral alarm signals. These data extend the range of tissue resident memory to SLO.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphoid Tissue/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , CD8-Positive T-Lymphocytes/metabolism , Flow Cytometry , Host-Pathogen Interactions/immunology , Interleukin-15/deficiency , Interleukin-15/genetics , Interleukin-15/immunology , L-Selectin/immunology , L-Selectin/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymph Nodes/immunology , Lymph Nodes/virology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Spleen/immunology , Spleen/virology
11.
Immunity ; 39(1): 171-83, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23890070

ABSTRACT

Memory CD8+ T cell quantity and quality determine protective efficacy against reinfection. Heterologous prime boost vaccination minimizes contraction of anamnestic effectors and maximizes memory CD8+ T cell quantity but reportedly erodes proliferative potential and protective efficacy. This study exploited heterologous prime boost vaccination to discover parameters regulating effector CD8+ T cell contraction and memory differentiation. When abundant memory T cells were established, boosting induced only 5-8 cell divisions, unusually rapid memory T cell differentiation as measured by phenotype and mitochondrial bioenergetic function, long-lived survival of 50% of effector T cells, and preservation of proliferative potential. Conversely, boosting in situations of low memory CD8+ T cell frequencies induced many cell divisions, increased contraction of effector cells, and caused senescence, low mitochondrial membrane potential, and poorly protective memory. Thus, anamnestic memory T cell differentiation is flexible, and abundant quantity can be achieved while maximizing protective efficacy and preserving proliferative potential.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation , Immunization, Secondary/methods , Immunologic Memory/immunology , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Survival/immunology , Flow Cytometry , Immunophenotyping , Mice , Mice, Inbred C57BL , Mitochondria/immunology , Mitochondria/metabolism , Time Factors
12.
Nat Immunol ; 14(5): 509-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23542740

ABSTRACT

CD8(+) T cells eliminate intracellular infections through two contact-dependent effector functions: cytolysis and secretion of antiviral cytokines. Here we identify the following additional function for memory CD8(+) T cells that persist at front-line sites of microbial exposure: to serve as local sensors of previously encountered antigens that precipitate innate-like alarm signals and draw circulating memory CD8(+) T cells into the tissue. When memory CD8(+) T cells residing in the female mouse reproductive tract encountered cognate antigen, they expressed interferon-γ (IFN-γ), potentiated robust local expression of inflammatory chemokines and induced rapid recruitment of circulating memory CD8(+) T cells. Anamnestic responses in front-line tissues are thus an integrated collaboration between front-line and circulating populations of memory CD8(+) T cells, and vaccines should establish both populations to maximize rapid responses.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens, Viral/immunology , Cell Movement/immunology , Cells, Cultured , Female , Genitalia, Female/immunology , Host-Pathogen Interactions , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Transplantation Chimera
13.
J Immunol ; 188(10): 4866-75, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22504644

ABSTRACT

Differentiation and maintenance of recirculating effector memory CD8 T cells (T(EM)) depends on prolonged cognate Ag stimulation. Whether similar pathways of differentiation exist for recently identified tissue-resident effector memory T cells (T(RM)), which contribute to rapid local protection upon pathogen re-exposure, is unknown. Memory CD8αß(+) T cells within small intestine epithelium are well-characterized examples of T(RM), and they maintain a long-lived effector-like phenotype that is highly suggestive of persistent Ag stimulation. This study sought to define the sources and requirements for prolonged Ag stimulation in programming this differentiation state, including local stimulation via cognate or cross-reactive Ags derived from pathogens, microbial flora, or dietary proteins. Contrary to expectations, we found that prolonged cognate Ag stimulation was dispensable for intestinal T(RM) ontogeny. In fact, chronic antigenic stimulation skewed differentiation away from the canonical intestinal T cell phenotype. Resident memory signatures, CD69 and CD103, were expressed in many nonlymphoid tissues including intestine, stomach, kidney, reproductive tract, pancreas, brain, heart, and salivary gland and could be driven by cytokines. Moreover, TGF-ß-driven CD103 expression was required for T(RM) maintenance within intestinal epithelium in vivo. Thus, induction and maintenance of long-lived effector-like intestinal T(RM) differed from classic models of T(EM) ontogeny and were programmed through a novel location-dependent pathway that was required for the persistence of local immunological memory.


Subject(s)
Cell Differentiation/immunology , Epitopes, T-Lymphocyte/physiology , Immunologic Memory/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Line , Female , Immunophenotyping , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , Time Factors , Tissue Distribution/immunology
14.
J Exp Med ; 207(3): 553-64, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20156972

ABSTRACT

Migration to intestinal mucosa putatively depends on local activation because gastrointestinal lymphoid tissue induces expression of intestinal homing molecules, whereas skin-draining lymph nodes do not. This paradigm is difficult to reconcile with reports of intestinal T cell responses after alternative routes of immunization. We reconcile this discrepancy by demonstrating that activation within spleen results in intermediate induction of homing potential to the intestinal mucosa. We further demonstrate that memory T cells within small intestine epithelium do not routinely recirculate with memory T cells in other tissues, and we provide evidence that homing is similarly dynamic in humans after subcutaneous live yellow fever vaccine immunization. These data explain why systemic immunization routes induce local cell-mediated immunity within the intestine and indicate that this tissue must be seeded with memory T cell precursors shortly after activation.


Subject(s)
Immunologic Memory , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Yellow Fever Vaccine/therapeutic use , Animals , Antigens, CD34/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Humans , Immunization , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Receptors, Lymphocyte Homing/immunology , Spleen/immunology , T-Lymphocytes/physiology
15.
Vaccine ; 27(28): 3766-74, 2009 Jun 08.
Article in English | MEDLINE | ID: mdl-19464560

ABSTRACT

There is a substantial need to develop better influenza virus vaccines that can protect populations that are not adequately protected by the currently licensed vaccines. While live attenuated influenza virus vaccines induce superior immune responses compared to inactivated vaccines, the manufacturing process of both types of influenza virus vaccines is time consuming and may not be adequate during a pandemic. Adjuvants would be particularly useful if they could enhance the immune response to live attenuated influenza virus vaccines so that the amount of vaccine needed for a protective dose could be reduced. The glycolipid, alpha-galactosylceramide (alpha-GalCer), has recently been shown to have adjuvant activity for both inactivated and replicating recombinant vaccines. The goal of these experiments was to determine whether a derivative of alpha-GalCer, alpha-C-galactosylceramide (alpha-C-GalCer) can enhance the immune response elicited by a live attenuated influenza virus vaccine containing an NS1 protein truncation and reduce the amount of vaccine required to provide protection after challenge. Our results indicated that the adjuvant reduced both morbidity and mortality in BALB/c mice after challenge with wild type influenza virus. The adjuvant also increased the amount of influenza virus specific total IgG, IgG1, and IgG2a antibodies as well as IFN-gamma secreting CD8(+) T cells. By using knockout mice that are not able to generate NKT cells, we were able to demonstrate that the mechanism of adjuvant activity is dependent on NKT cells. Thus, our data indicate that stimulators of NKT cells represent a new avenue of adjuvants to pursue for live attenuated virus vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Galactosylceramides/pharmacology , Influenza Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/immunology , Chick Embryo , Female , Galactosylceramides/administration & dosage , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Severity of Illness Index , Survival Analysis , Vaccines, Attenuated/immunology
16.
J Virol ; 82(21): 10580-90, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18768976

ABSTRACT

Type B influenza viruses can cause substantial morbidity and mortality in the population, and vaccination remains by far the best means of protection against infections with these viruses. Here, we report the construction of mutant influenza B viruses for potential use as improved live-virus vaccine candidates. Employing reverse genetics, we altered the NS1 gene, which encodes a type I interferon (IFN) antagonist. The resulting NS1 mutant viruses induced IFN and, as a consequence, were found to be attenuated in vitro and in vivo. The absence of pathogenicity of the NS1 mutants in both BALB/c and C57BL/6 PKR(-/-) mice was confirmed. We also provide evidence that influenza B virus NS1 mutants induce a self-adjuvanted immune response and confer effective protection against challenge with both homologous and heterologous B virus strains in mice.


Subject(s)
Influenza B virus/genetics , Influenza B virus/immunology , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Viral Nonstructural Proteins/genetics , Animals , Antibodies, Viral/blood , Body Weight , Cell Line , Chlorocebus aethiops , Dogs , Female , Immunoglobulin A/analysis , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Lung/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/immunology , Orthomyxoviridae Infections/prevention & control , Survival Analysis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Plaque Assay
17.
J Virol ; 81(10): 5091-101, 2007 May.
Article in English | MEDLINE | ID: mdl-17344289

ABSTRACT

During eukaryotic mRNA transcription, the synthetic activity and mRNA processing factor interactions of RNA polymerase II (RNAP II) are regulated by phosphorylation of its carboxyl-terminal domain (CTD), with modification occurring primarily on serines 2 and 5 of the CTD. We previously showed that herpes simplex virus type 1 (HSV-1) infection rapidly triggers the loss of RNAP II forms bearing serine 2 phosphorylation (Ser-2P RNAP II). Here we show that the HSV-1 immediate-early (IE) protein ICP22 is responsible for this effect during the IE phase of infection. This activity does not require the viral UL13 protein kinase, which is required for several other regulatory functions of ICP22. Additionally, we show that transient expression of ICP22 can trigger the loss of Ser-2P RNAP II in transfected cells. Thus, the ability of ICP22 to cause the loss of Ser-2 RNAP II does not require other viral factors or the context of the infected cell. Expression of the HSV-1 ICP22-related protein US1.5, which corresponds to residues 147 to 420 of ICP22, also triggers a loss of Ser-2P RNAP II in transfected cells, whereas expression of the varicella-zoster virus ICP22 homolog, ORF63, does not. Our study also provides evidence for a second, viral late gene-dependent pathway that triggers loss of Ser-2P RNAP II in infected cells, consistent with the recent work of Dai-Ju et al. (J. Q. Dai-Ju, L. Li, L. A. Johnson, and R. M. Sandri-Goldin, J. Virol. 80:3567-3581, 2006). Therefore, it appears that HSV-1 has evolved redundant mechanisms for triggering the loss of a specific phosphorylated form of RNAP II.


Subject(s)
Herpesvirus 1, Human/physiology , Immediate-Early Proteins/physiology , RNA Polymerase II/metabolism , Animals , Chlorocebus aethiops , Immunoblotting , Microscopy, Fluorescence , Phosphorylation , Protein Kinases/physiology , Serine/metabolism , Vero Cells , Viral Envelope Proteins/physiology , Viral Regulatory and Accessory Proteins
18.
J Virol ; 79(17): 11323-34, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103184

ABSTRACT

Previous studies have shown that herpes simplex virus type 1 (HSV-1) infection alters the phosphorylation of the carboxyl-terminal domain (CTD) of RNA polymerase II (RNAP II), creating a new form of the enzyme known as RNAP II(I). However, the specific phosphorylation changes induced by HSV-1 have not been characterized. In this study, we used phospho-specific anti-CTD antibodies to probe the structure of the postinfection RNAP II. We find that RNAP II(I) is phosphorylated on serine-5 (Ser-5) of the CTD consensus repeat but generally lacks phosphorylation on serine-2 (Ser-2). Since Ser-2 phosphorylation is normally associated with efficient transcriptional elongation and the recruitment of pre-mRNA processing factors, our results suggest that RNAP II(I) may have altered elongation properties and decreased interactions with the mRNA processing machinery. The viral factors responsible for the reduction in Ser-2 CTD phosphorylation were studied. We found that viral immediate-early (IE) gene expression is required and sufficient, in the context of infection, for loss of Ser-2 phosphorylation. However, studies with viral mutants failed to implicate a single IE protein (among ICP0, ICP4, ICP22, and ICP27) in this process. Although most Ser-2-phosphorylated RNAP II is lost after infection, our immunofluorescence analyses identified a small subfraction that escapes loss and relocalizes to splicing antigen-rich nuclear speckles. A similar phenomenon is seen in uninfected cells after various treatments that inhibit RNAP II transcription. We hypothesize that the HSV-1-induced relocalization of residual Ser-2-phosphorylated RNAP II to nuclear speckles reflects a host response to the inhibition of cellular gene transcription.


Subject(s)
Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , RNA Polymerase II/metabolism , Animals , Cell Line , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Phosphorylation , Protein Structure, Tertiary , Serine , Transcription, Genetic , Virus Replication
19.
J Biol Chem ; 278(10): 8401-6, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12477720

ABSTRACT

Perilipin A coats the lipid storage droplets in adipocytes and is polyphosphorylated by protein kinase A (PKA); the fact that PKA activates lipolysis in adipocytes suggests a role for perilipins in this process. To assess whether perilipins participate directly in PKA-mediated lipolysis, we have expressed constructs coding for native and mutated forms of the two major splice variants of the perilipin gene, perilipins A and B, in Chinese hamster ovary fibroblasts. Perilipins localize to lipid droplet surfaces and displace the adipose differentiation-related protein that normally coats the droplets in these cells. Perilipin A inhibits triacylglycerol hydrolysis by 87% when PKA is quiescent, but activation of PKA and phosphorylation of perilipin A engenders a 7-fold lipolytic activation. Mutation of PKA sites within the N-terminal region of perilipin abrogates the PKA-mediated lipolytic response. In contrast, perilipin B exerts only minimal protection against lipolysis and is unresponsive to PKA activation. Since Chinese hamster ovary cells contain no PKA-activated lipase, we conclude that the expression of perilipin A alone is sufficient to confer PKA-mediated lipolysis in these cells. Moreover, the data indicate that the unique C-terminal portion of perilipin A is responsible for its protection against lipolysis and that phosphorylation at the N-terminal PKA sites attenuates this protective effect.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphoproteins/physiology , Triglycerides/metabolism , Animals , CHO Cells , Carrier Proteins , Cricetinae , Cricetulus , Enzyme Activation , Fluorescent Antibody Technique , Mice , Mutagenesis , Perilipin-1 , Phosphoproteins/genetics , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...