Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell ; 181(7): 1502-1517.e23, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32559462

ABSTRACT

RNA viruses are a major human health threat. The life cycles of many highly pathogenic RNA viruses like influenza A virus (IAV) and Lassa virus depends on host mRNA, because viral polymerases cleave 5'-m7G-capped host transcripts to prime viral mRNA synthesis ("cap-snatching"). We hypothesized that start codons within cap-snatched host transcripts could generate chimeric human-viral mRNAs with coding potential. We report the existence of this mechanism of gene origination, which we named "start-snatching." Depending on the reading frame, start-snatching allows the translation of host and viral "untranslated regions" (UTRs) to create N-terminally extended viral proteins or entirely novel polypeptides by genetic overprinting. We show that both types of chimeric proteins are made in IAV-infected cells, generate T cell responses, and contribute to virulence. Our results indicate that during infection with IAV, and likely a multitude of other human, animal and plant viruses, a host-dependent mechanism allows the genesis of hybrid genes.


Subject(s)
RNA Caps/genetics , RNA Virus Infections/genetics , Recombinant Fusion Proteins/genetics , 5' Untranslated Regions/genetics , Animals , Cattle , Cell Line , Cricetinae , Dogs , Humans , Influenza A virus/metabolism , Mice , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Open Reading Frames/genetics , RNA Caps/metabolism , RNA Virus Infections/metabolism , RNA Viruses/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Fusion Proteins/metabolism , Transcription, Genetic/genetics , Viral Proteins/metabolism , Virus Replication/genetics
2.
Nature ; 619(7969): 338-347, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380775

ABSTRACT

Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.


Subject(s)
Birds , Host Microbial Interactions , Influenza A virus , Influenza in Birds , Influenza, Human , Viral Zoonoses , Animals , Humans , Birds/virology , Influenza A virus/classification , Influenza A virus/genetics , Influenza A virus/growth & development , Influenza A virus/isolation & purification , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/prevention & control , Influenza, Human/transmission , Influenza, Human/virology , Primates , Respiratory System/metabolism , Respiratory System/virology , Risk Assessment , Viral Zoonoses/prevention & control , Viral Zoonoses/transmission , Viral Zoonoses/virology , Virus Replication
3.
Immunity ; 50(2): 378-389.e5, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30784579

ABSTRACT

Currently, we lack an understanding of the individual and combinatorial roles for chemokine receptors in the inflammatory process. We report studies on mice with a compound deletion of Ccr1, Ccr2, Ccr3, and Ccr5, which together control monocytic and eosinophilic recruitment to resting and inflamed sites. Analysis of resting tissues from these mice, and mice deficient in each individual receptor, provides clear evidence for redundant use of these receptors in establishing tissue-resident monocytic cell populations. In contrast, analysis of cellular recruitment to inflamed sites provides evidence of specificity of receptor use for distinct leukocyte subtypes and no indication of comprehensive redundancy. We find no evidence of involvement of any of these receptors in the recruitment of neutrophils or lymphocytes to resting or acutely inflamed tissues. Our data shed important light on combinatorial inflammatory chemokine receptor function and highlight Ccr2 as the primary driver of myelomonocytic cell recruitment in acutely inflamed contexts.


Subject(s)
Eosinophils/immunology , Inflammation/immunology , Monocytes/immunology , Receptors, CCR/immunology , Animals , Chemokines/immunology , Chemokines/metabolism , Eosinophils/metabolism , Gene Expression Profiling/methods , Inflammation/genetics , Inflammation/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Receptors, CCR/genetics , Receptors, CCR/metabolism , Receptors, CCR1/immunology , Receptors, CCR1/metabolism , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Receptors, CCR3/immunology , Receptors, CCR3/metabolism , Receptors, CCR5/immunology , Receptors, CCR5/metabolism
4.
Eur J Immunol ; 53(11): e2350559, 2023 11.
Article in English | MEDLINE | ID: mdl-37490492

ABSTRACT

Cytokine production by memory T cells is a key mechanism of T cell mediated protection. However, we have limited understanding of the persistence of cytokine producing T cells during memory cell maintenance and secondary responses. We interrogated antigen-specific CD4 T cells using a mouse influenza A virus infection model. Although CD4 T cells detected using MHCII tetramers declined in lymphoid and non-lymphoid organs, we found similar numbers of cytokine+ CD4 T cells at days 9 and 30 in the lymphoid organs. CD4 T cells with the capacity to produce cytokines expressed higher levels of pro-survival molecules, CD127 and Bcl2, than non-cytokine+ cells. Transcriptomic analysis revealed a heterogeneous population of memory CD4 T cells with three clusters of cytokine+ cells. These clusters match flow cytometry data and reveal an enhanced survival signature in cells capable of producing multiple cytokines. Following re-infection, multifunctional T cells expressed low levels of the proliferation marker, Ki67, whereas cells that only produce the anti-viral cytokine, interferon-γ, were more likely to be Ki67+ . Despite this, multifunctional memory T cells formed a substantial fraction of the secondary memory pool. Together these data indicate that survival rather than proliferation may dictate which populations persist within the memory pool.


Subject(s)
CD4-Positive T-Lymphocytes , Influenza A virus , CD4-Positive T-Lymphocytes/metabolism , Ki-67 Antigen , Cytokines/metabolism , Interferon-gamma/metabolism , Immunologic Memory
5.
Immunology ; 163(3): 239-249, 2021 07.
Article in English | MEDLINE | ID: mdl-33556186

ABSTRACT

Communication between stromal and immune cells is essential to maintain tissue homeostasis, mount an effective immune response and promote tissue repair. This 'crosstalk' occurs in both the steady state and following a variety of insults, for example, in response to local injury, at sites of infection or cancer. What do we mean by crosstalk between cells? Reciprocal activation and/or regulation occurs between immune and stromal cells, by direct cell contact and indirect mechanisms, including the release of soluble cytokines. Moving beyond cell-to-cell contact, this review investigates the complexity of 'cross-space' cellular communication. We highlight different examples of cellular communication by a variety of lung stromal and immune cells following tissue insults. This review examines how the 'geography of the lung microenvironment' is altered in various disease states; more specifically, we investigate how this influences lung epithelial cells and fibroblasts via their communication with immune cells and each other.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Fibroblasts/immunology , Lung/pathology , Stromal Cells/immunology , Animals , Cell Communication , Cellular Microenvironment , Humans , Immunity, Cellular
6.
Immunology ; 162(1): 68-83, 2021 01.
Article in English | MEDLINE | ID: mdl-32931017

ABSTRACT

Memory T cells respond rapidly in part because they are less reliant on a heightened levels of costimulatory molecules. This enables rapid control of secondary infecting pathogens but presents challenges to efforts to control or silence memory CD4 T cells, for example in antigen-specific tolerance strategies for autoimmunity. We have examined the transcriptional and functional consequences of reactivating memory CD4 T cells in the absence of an adjuvant. We find that memory CD4 T cells generated by infection or immunisation survive secondary activation with antigen delivered without adjuvant, regardless of their location in secondary lymphoid organs or peripheral tissues. These cells were, however, functionally altered following a tertiary immunisation with antigen and adjuvant, proliferating poorly but maintaining their ability to produce inflammatory cytokines. Transcriptional and cell cycle analysis of these memory CD4 T cells suggests they are unable to commit fully to cell division potentially because of low expression of DNA repair enzymes. In contrast, these memory CD4 T cells could proliferate following tertiary reactivation by viral re-infection. These data indicate that antigen-specific tolerogenic strategies must examine multiple parameters of Tcell function, and provide insight into the molecular mechanisms that may lead to deletional tolerance of memory CD4 T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Immunologic Memory/immunology , Animals , Antigens/immunology , Autoimmunity/immunology , Cell Cycle/immunology , Cell Proliferation/physiology , Cytokines/immunology , DNA Repair/immunology , Female , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Transcription, Genetic/immunology
7.
Eur J Immunol ; 50(5): 666-675, 2020 05.
Article in English | MEDLINE | ID: mdl-32114694

ABSTRACT

Analysis of chemokine receptor, and atypical chemokine receptor, expression is frequently hampered by the lack of availability of high-quality antibodies and the species specificity of those that are available. We have previously described methodology utilizing Alexa-Fluor-labeled chemokine ligands as versatile reagents to detect receptor expression. Previously this has been limited to hematopoietic cells and methodology for assessing expression of receptors on stromal cells has been lacking. Among chemokine receptors, the ones most frequently expressed on stromal cells belong to the atypical chemokine receptor subfamily. These receptors do not signal in the classic sense in response to ligand but scavenge their ligands and degrade them and thus sculpt in vivo chemokine gradients. Here, we demonstrate the ability to use either intratracheal or intravenous, Alexa-Fluor-labeled chemokine administration to detect stromal cell populations expressing the atypical chemokine receptor ACKR2. Using this methodology, we demonstrate, for the first time, expression of ACKR2 on blood endothelial cells. This observation sets the lung aside from other tissues in which ACKR2 is exclusively expressed on lymphatic endothelial cells and suggest unique roles for ACKR2 in the pulmonary environment.


Subject(s)
Endothelial Cells/immunology , Lung/immunology , Receptors, Chemokine/immunology , Stromal Cells/immunology , Animals , Carbocyanines/chemistry , Endothelial Cells/cytology , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/immunology , Flow Cytometry , Fluorescent Dyes/chemistry , Gene Expression , Lung/blood supply , Lung/cytology , Mice , Mice, Knockout , Receptors, Chemokine/genetics , Staining and Labeling/methods , Stromal Cells/cytology
8.
Eur J Immunol ; 49(11): 2019-2029, 2019 11.
Article in English | MEDLINE | ID: mdl-31177549

ABSTRACT

T cell protective immunity is associated with multifunctional memory cells that produce several different cytokines. Currently, our understanding of when and how these cells are generated is limited. We have used an influenza virus mouse infection model to investigate whether the cytokine profile of memory T cells is reflective of primary responding cells or skewed toward a distinct profile. We found that, in comparison to primary cells, memory T cells tended to make multiple cytokines simultaneously. Analysis of the timings of release of cytokine by influenza virus-specific T cells, demonstrated that primary responding CD4 T cells from lymphoid organs were unable to produce a sustained cytokine response. In contrast CD8 T cells, memory CD4 T cells, and primary responding CD4 T cells from the lung produced a sustained cytokine response throughout the restimulation period. Moreover, memory CD4 T cells were more resistant than primary responding CD4 T cells to inhibitors that suppress T cell receptor signaling. Together, these data suggest that memory CD4 T cells display superior cytokine responses compared to primary responding cells. These data are key to our ability to identify the cues that drive the generation of protective memory CD4 T cells following infection.


Subject(s)
Antigens, Viral/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Gene Expression Regulation/immunology , Immunity, Cellular/drug effects , Immunologic Memory , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Immunity, Cellular/genetics , Immunophenotyping , Influenza A virus/chemistry , Influenza A virus/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Lung/cytology , Lung/drug effects , Lung/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Organ Specificity , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Nat Immunol ; 9(12): 1356-63, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18931680

ABSTRACT

Nedd4 and Itch are E3 ubiquitin ligases that ubiquitinate similar targets in vitro and thus are thought to function similarly. T cells lacking Itch show spontaneous activation and T helper type 2 polarization. To test whether loss of Nedd4 affects T cells in the same way, we generated Nedd4(+/+) and Nedd4(-/-) fetal liver chimeras. Nedd4(-/-) T cells developed normally but proliferated less, produced less interleukin 2 and provided inadequate help to B cells. Nedd4(-/-) T cells contained more of the E3 ubiquitin ligase Cbl-b, and Nedd4 was required for polyubiquitination of Cbl-b induced by CD28 costimulation. Our data demonstrate that Nedd4 promotes the conversion of naive T cells into activated T cells. We propose that Nedd4 and Itch ubiquitinate distinct target proteins in vivo.


Subject(s)
Lymphocyte Activation/immunology , Proto-Oncogene Proteins c-cbl/metabolism , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/immunology , Ubiquitination/immunology , Animals , CD28 Antigens/immunology , CD28 Antigens/metabolism , Endosomal Sorting Complexes Required for Transport , Flow Cytometry , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Nedd4 Ubiquitin Protein Ligases , Proto-Oncogene Proteins c-cbl/immunology , T-Lymphocytes/metabolism , Transplantation Chimera , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
J Immunol ; 201(8): 2510-2519, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30158126

ABSTRACT

Chemokines have been shown to be essential players in a range of cancer contexts. In this study, we demonstrate that mice deficient in the atypical chemokine receptor Ackr2 display impaired development of metastasis in vivo in both cell line and spontaneous models. Further analysis reveals that this relates to increased expression of the chemokine receptor CCR2, specifically by KLRG1+ NK cells from the Ackr2-/- mice. This leads to increased recruitment of KLRG1+ NK cells to CCL2-expressing tumors and enhanced tumor killing. Together, these data indicate that Ackr2 limits the expression of CCR2 on NK cells and restricts their tumoricidal activity. Our data have important implications for our understanding of the roles for chemokines in the metastatic process and highlight Ackr2 and CCR2 as potentially manipulable therapeutic targets in metastasis.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Receptors, Chemokine/metabolism , Animals , Carcinoma, Lewis Lung , Cell Movement , Chemokine CCL2/metabolism , Cytotoxicity, Immunologic , Lectins, C-Type , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Immunologic/metabolism
11.
Immunology ; 2018 Mar 23.
Article in English | MEDLINE | ID: mdl-29570776

ABSTRACT

Immunological memory provides rapid protection to pathogens previously encountered through infection or vaccination. CD4 T-cells play a central role in all adaptive immune responses. Vaccines must, therefore, activate CD4 T-cells if they are to generate protective immunity. For many diseases, we do not have effective vaccines. These include human immunodeficiency virus (HIV), tuberculosis and malaria, which are responsible for many millions of deaths each year across the globe. CD4 T-cells play many different roles during the immune response coordinating the actions of many other cells. In order to harness the diverse protective effects of memory CD4 T-cells, we need to understand how memory CD4 T-cells are generated and how they protect the host. Here we review recent findings on the location of different subsets of memory CD4 T-cells that are found in peripheral tissues (tissue resident memory T-cells) and in the circulation (central and effector memory T-cells). We discuss the generation of these cells, and the evidence that demonstrates how they provide immune protection in animal and human challenge models.

12.
Proc Natl Acad Sci U S A ; 111(21): 7735-40, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821788

ABSTRACT

A major goal for immunotherapy is to tolerize the immune cells that coordinate tissue damage in autoimmune and alloantigen responses. CD4 T cells play a central role in many of these conditions and improved antigen-specific regulation or removal of these cells could revolutionize current treatments. A confounding factor is that little is known about whether and how tolerance is induced in memory CD4 T cells. We used MHC class II tetramers to track and analyze a population of endogenous antigen-specific memory CD4 T cells exposed to soluble peptide in the absence of adjuvant. We found that such memory T cells proliferated and reentered the memory pool apparently unperturbed by the incomplete activation signals provided by the peptide. Upon further restimulation in vivo, CD4 memory T cells that had been previously exposed to peptide proliferated, provided help to primary responding B cells, and migrated to inflamed sites. However, these reactivated memory cells failed to survive. The reduction in T-cell number was marked by low expression of the antiapoptotic molecule B cell lymphoma 2 (Bcl2) and increased expression of activated caspase molecules. Consequently, these cells failed to sustain a delayed-type hypersensitivity response. Moreover, following two separate exposures to soluble antigen, no T-cell recall response and no helper activity for B cells could be detected. These results suggest that the induction of tolerance in memory CD4 T cells is possible but that deletion and permanent removal of the antigen-specific T cells requires reactivation following exposure to the tolerogenic antigen.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Immunologic Memory/immunology , Immunotherapy/methods , Lymphocyte Activation/immunology , Analysis of Variance , Animals , Antigens/immunology , Caspases/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Genes, MHC Class II/immunology , Mice , Mice, Inbred C57BL
13.
J Immunol ; 190(3): 913-21, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275606

ABSTRACT

We re-examined the observation that γδ T cells, when transferred from mice tolerized to an inhaled conventional Ag, suppress the allergic IgE response to this Ag specifically. Using OVA and hen egg lysozyme in crisscross fashion, we confirmed the Ag-specific IgE-regulatory effect of the γδ T cells. Although only Vγ4(+) γδ T cells are regulators, the Ag specificity does not stem from specificity of their γδ TCRs. Instead, the Vγ4(+) γδ T cells failed to respond to either Ag, but rapidly acquired Ag-specific regulatory function in vivo following i.v. injection of non-T cells derived from the spleen of Ag-tolerized mice. This correlated with their in vivo Ag acquisition from i.v. injected Ag-loaded splenic non-T cells, and in vivo transfer of membrane label provided evidence for direct contact between the injected splenic non-T cells and the Vγ4(+) γδ T cells. Together, our data suggest that Ag itself, when acquired by γδ T cells, directs the specificity of their IgE suppression.


Subject(s)
Antigens/immunology , Asthma/immunology , Immunoglobulin E/immunology , Muramidase/immunology , Ovalbumin/immunology , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/immunology , Administration, Inhalation , Adoptive Transfer , Aerosols , Animals , Antigens/administration & dosage , Antigens/toxicity , Asthma/etiology , Cell Separation , Female , Humans , Immune Tolerance , Immunological Synapses , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Muramidase/administration & dosage , Muramidase/toxicity , Ovalbumin/administration & dosage , Ovalbumin/toxicity , Spleen/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/transplantation
14.
J Immunol ; 189(9): 4275-83, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23008448

ABSTRACT

Autoreactive anergic B lymphocytes are considered to be dangerous because of their potential for activation and recruitment into autoimmune responses. However, they persist for days and constitute ∼5% of the B cell pool. We assessed their functional potential in the Ars/A1 transgene model, where anergic B cells express a dual-reactive Ag receptor that binds, in addition to a self-Ag, the hapten p-azophenylarsonate (Ars). When Ars/A1 B cells were transferred into adoptive recipients that were immunized with foreign proteins covalently conjugated with Ars, endogenous IgG immune responses to both were selectively and severely diminished, and the development of T helper cells was impaired. Approximately 95% inhibition of the anti-Ars response was attained with ∼4000 transferred Ars/A1 B cells through redundant mechanisms, one of which depended on their expression of MHC class II but not upon secretion of IL-10 or IgM. This Ag-specific suppressive activity implicates the autoreactive anergic B cell as an enforcer of immunological tolerance to self-Ags.


Subject(s)
Antibody Formation , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Clonal Anergy/immunology , Epitopes, B-Lymphocyte/immunology , Immunosuppression Therapy/methods , Adoptive Transfer , Animals , Autoantigens/biosynthesis , Autoantigens/metabolism , B-Lymphocyte Subsets/transplantation , Cells, Cultured , Epitopes, B-Lymphocyte/metabolism , Immunoglobulin G/biosynthesis , Mice , Mice, 129 Strain , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Self Tolerance/genetics , Self Tolerance/immunology , Spleen/immunology , Spleen/metabolism , Spleen/transplantation , p-Azobenzenearsonate/biosynthesis , p-Azobenzenearsonate/metabolism
15.
Proc Natl Acad Sci U S A ; 108(19): 7914-9, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518876

ABSTRACT

Vaccines can greatly reduce the spread of and deaths from many infectious diseases. However, many infections have no successful vaccines. Better understanding of the generation of protective CD8 memory T cells by vaccination is essential for the rational design of new vaccines that aim to prime cellular immune responses. Here we demonstrate that the combination of two adjuvants that are currently licensed for use in humans can be used to prime long-lived memory CD8 T cells that protect mice from viral challenge. The universally used adjuvant, aluminum salts, primed long-lived memory CD8 T cells; however, effective cytotoxic T-cell differentiation occurred only in the presence of an additional adjuvant, monophosphoryl lipid A (MPL). MPL-induced IL-6 was required for cytotoxic differentiation. The IL-6 acted by inducing granzyme B production and reducing expression of inhibitory molecule PD1 on the surface of the primed CD8 T cells. CD8 memory T cells generated by antigen delivered with both aluminum salts and MPL provided significant protection from influenza A challenge. These adjuvants could be used in human vaccines to prime protective memory CD8 T cells.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Lipid A/analogs & derivatives , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/administration & dosage , Animals , Antigen Presentation , Antigens, Surface/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , Cattle , Cytokines/biosynthesis , Female , Humans , Immunologic Memory , Influenza A virus/immunology , Interleukin-6/biosynthesis , Interleukin-6/deficiency , Interleukin-6/genetics , Lipid A/administration & dosage , Mice , Mice, Knockout , Mice, Transgenic , Nucleocapsid Proteins , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Ovalbumin/administration & dosage , Ovalbumin/immunology , Programmed Cell Death 1 Receptor , RNA-Binding Proteins/administration & dosage , RNA-Binding Proteins/immunology , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/immunology , Viral Core Proteins/administration & dosage , Viral Core Proteins/immunology
16.
Mucosal Immunol ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38851589

ABSTRACT

Influenza A virus (IAV) infection leads to the formation of mucosal memory CD4 T cells that can protect the host. An in-depth understanding of the signals that shape memory cell development is required for more effective vaccine design. We have examined the formation of memory CD4 T cells in the lung following IAV infection of mice, characterizing changes to the lung landscape and immune cell composition. IAV-specific CD4 T cells were found throughout the lung at both primary and memory time points. These cells were found near lung airways and in close contact with a range of immune cells including macrophages, dendritic cells, and B cells. Interactions between lung IAV-specific CD4 T cells and major histocompatibility complex (MHC)II+ cells during the primary immune response were important in shaping the subsequent memory pool. Treatment with an anti-MHCII blocking antibody increased the proportion of memory CD4 T cells found in lung airways but reduced interferon-γ expression by IAV-specific immunodominant memory CD4 T cells. The immunodominant CD4 T cells expressed higher levels of programmed death ligand 1 (PD1) than other IAV-specific CD4 T cells and PD1+ memory CD4 T cells were located further away from MHCII+ cells than their PD1-low counterparts. This distinction in location was lost in mice treated with anti-MHCII antibodies. These data suggest that sustained antigen presentation in the lung impacts the formation of memory CD4 T cells by regulating their cytokine production and location.

17.
J Immunol ; 186(5): 2889-96, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270407

ABSTRACT

CD4 T cell help for B cells is critical for effective Ab responses. Although many of the molecules involved in helper functions of naive CD4 T cells have been characterized, much less is known about the helper capabilities of memory CD4 T cells, an important consideration for the design of vaccines that aim to prime protective memory CD4 T cells. In this study, we demonstrate that memory CD4 T cells enable B cells to expand more rapidly and class switch earlier than do primary responding CD4 T cells. This accelerated response does not require large numbers of memory cells, and similar numbers of primary responding cells provide less effective help than do memory cells. However, only memory CD4 T cells that express the B cell follicle homing molecule, CXCR5, are able to accelerate the response, suggesting that the rapidity of the Ab response depends on the ability of CD4 memory T cells to migrate quickly toward B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Memory , Receptors, CXCR5/biosynthesis , Amino Acid Sequence , Animals , B-Lymphocyte Subsets/microbiology , B-Lymphocyte Subsets/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Movement/immunology , Female , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Receptors, CXCR5/physiology , Receptors, Lymphocyte Homing/administration & dosage , Receptors, Lymphocyte Homing/biosynthesis , Receptors, Lymphocyte Homing/physiology , Resting Phase, Cell Cycle/immunology
18.
Discov Immunol ; 2(1): kyad014, 2023.
Article in English | MEDLINE | ID: mdl-37842651

ABSTRACT

Interferon gamma (IFNγ) is a potent antiviral cytokine that can be produced by many innate and adaptive immune cells during infection. Currently, our understanding of which cells produce IFNγ and where they are located at different stages of an infection is limited. We have used reporter mice to investigate in vivo expression of Ifnγ mRNA in the lung and secondary lymphoid organs during and following influenza A virus (IAV) infection. We observed a triphasic production of Ifnγ expression. Unconventional T cells and innate lymphoid cells, particularly NK cells, were the dominant producers of early Ifnγ, while CD4 and CD8 T cells were the main producers by day 10 post-infection. Following viral clearance, some memory CD4 and CD8 T cells continued to express Ifnγ in the lungs and draining lymph node. Interestingly, Ifnγ production by lymph node natural killer (NK), NKT, and innate lymphoid type 1 cells also continued to be above naïve levels, suggesting memory-like phenotypes for these cells. Analysis of the localization of Ifnγ+ memory CD4 and CD8 T cells demonstrated that cytokine+ T cells were located near airways and in the lung parenchyma. Following a second IAV challenge, lung IAV-specific CD8 T cells rapidly increased their expression of Ifnγ while CD4 T cells in the draining lymph node increased their Ifnγ response. Together, these data suggest that Ifnγ production fluctuates based on cellular source and location, both of which could impact subsequent immune responses.

19.
J Exp Med ; 203(4): 897-906, 2006 Apr 17.
Article in English | MEDLINE | ID: mdl-16549596

ABSTRACT

Secondary T cell responses are enhanced because of an expansion in numbers of antigen-specific (memory) cells. Using major histocompatibility complex class II tetramers we have tracked peptide-specific endogenous (non-T cell receptor transgenic) CD4 memory T cells in normal and in costimulation-deficient mice. CD4 memory T cells were detectable after immunization for more than 200 days, although decay was apparent. Memory cells generated in CD40 knockout mice by immunization with peptide-pulsed wild-type dendritic cells survived in the absence of CD40 and proliferated when boosted with peptide (plus adjuvant) in a CD40-independent fashion. However, differentiation of the memory cells into cytokine-producing effector cells did not occur in the absence of CD40. The data indicate that memory cells can be generated without passing through the effector cell stage.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/physiology , Cell Differentiation , Immunologic Memory , Amino Acid Sequence , Animals , CD40 Antigens/genetics , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/immunology , Cells, Cultured , Dendritic Cells/immunology , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data
20.
Blood ; 116(24): 5191-9, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20876456

ABSTRACT

It has been recognized for nearly 80 years that insoluble aluminum salts are good immunologic adjuvants and that they form long-lived nodules in vivo. Nodule formation has long been presumed to be central for adjuvant activity by providing an antigen depot, but the composition and function of these nodules is poorly understood. We show here that aluminum salt nodules formed within hours of injection and contained the clotting protein fibrinogen. Fibrinogen was critical for nodule formation and required processing to insoluble fibrin by thrombin. DNase treatment partially disrupted the nodules, and the nodules contained histone H3 and citrullinated H3, features consistent with extracellular traps. Although neutrophils were not essential for nodule formation, CD11b(+) cells were implicated. Vaccination of fibrinogen-deficient mice resulted in normal CD4 T-cell and antibody responses and enhanced CD8 T-cell responses, indicating that nodules are not required for aluminum's adjuvant effect. Moreover, the ability of aluminum salts to retain antigen in the body, the well-known depot effect, was unaffected by the absence of nodules. We conclude that aluminum adjuvants form fibrin-dependent nodules in vivo, that these nodules have properties of extracellular traps, and the nodules are not required for aluminum salts to act as adjuvants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum/administration & dosage , Adjuvants, Immunologic/pharmacology , Aluminum/pharmacology , Animals , Fibrin/metabolism , Fibrinogen/metabolism , Histones/metabolism , Mice , Mice, Knockout , Salts , T-Lymphocytes/immunology , Thrombin/metabolism , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL