Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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.

9.
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
10.
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
11.
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.

12.
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
13.
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.

14.
Int J Biochem Cell Biol ; 153: 106331, 2022 12.
Article in English | MEDLINE | ID: mdl-36368596

ABSTRACT

The lung is frequently and repeatedly exposed to invading pathogens and thus requires constant immunosurveillance. Professional antigen presenting cells (APCs), including dendritic cells, engulf invading pathogens and present their peptides via major histocompatibility complexes (MHC) I and II, to CD8 or CD4 T cells. Epithelial cells and stromal cells (including fibroblasts) provide more than structural support, they are increasingly recognised as key players in the immune response, acting as non-professional APCs through interactions with antigen experienced T cells that migrate to the lung. The importance of the contributions of non-professional and professional APCs to T cell function in vivo, is currently unclear. This review summarises the roles of professional and non-professional APCs in lung immunity, at the steady state and following viral insult, with particular emphasis on their ability to interact with and influence T cells.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells , Humans , CD4-Positive T-Lymphocytes , Lung , Major Histocompatibility Complex
15.
Int J Radiat Oncol Biol Phys ; 112(1): 197-211, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34478832

ABSTRACT

PURPOSE: Low-dose whole lung radiation therapy (LDLR) has been proposed as a treatment for patients with acute respiratory distress syndrome associated with SARS-CoV-2 infection, and clinical trials are underway. There is an urgent need for preclinical evidence to justify this approach and inform dose, scheduling, and mechanisms of action. METHODS AND MATERIALS: Female C57BL/6 mice were treated with intranasal bleomycin sulfate (7.5 or 11.25 units/kg, day 0) and then exposed to whole lung radiation therapy (0.5, 1.0, or 1.5 Gy, or sham; day 3). Bodyweight was measured daily, and lung tissue was harvested for histology and flow cytometry on day 10. Computed tomography lung imaging was performed before radiation (day 3) and pre-endpoint (day 10). RESULTS: Bleomycin caused pneumonitis of variable severity, which correlated with weight loss. LDLR at 1.0 Gy was associated with a significant increase in the proportion of mice recovering to 98% of initial bodyweight, and a proportion of these mice exhibited less severe histopathologic lung changes. Mice experiencing moderate initial weight loss were more likely to respond to LDLR than those experiencing severe initial weight loss. In addition, LDLR (1.0 Gy) significantly reduced bleomycin-induced increases in interstitial macrophages, CD103+ dendritic cells (DCs), and neutrophil-DC hybrids. Overall, bleomycin-treated mice exhibited significantly higher percentages of nonaerated lung in left than right lungs, and LDLR (1.0 Gy) limited further reductions in aerated lung volume in right but not left lungs. LDLR at 0.5 and 1.5 Gy did not improve bodyweight, flow cytometric, or radiologic readouts of bleomycin-induced pneumonitis. CONCLUSIONS: Our data support the concept that LDLR can ameliorate acute inflammatory lung injury, identify 1.0 Gy as the most effective dose, and provide evidence that it is more effective in the context of moderate than severe pneumonitis. Mechanistically, LDLR at 1.0 Gy significantly suppressed bleomycin-induced accumulation of pulmonary interstitial macrophages, CD103+ DCs, and neutrophil-DC hybrids.


Subject(s)
Pneumonia , Radiotherapy , Animals , Bleomycin , COVID-19/radiotherapy , Disease Models, Animal , Female , Humans , Lung/diagnostic imaging , Mice , Mice, Inbred C57BL , Pneumonia/chemically induced , Weight Loss
16.
J Immunol ; 183(7): 4403-14, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734227

ABSTRACT

To understand more about how the body recognizes alum we characterized the early innate and adaptive responses in mice injected with the adjuvant. Within hours of exposure, alum induces a type 2 innate response characterized by an influx of eosinophils, monocytes, neutrophils, DCs, NK cells and NKT cells. In addition, at least 13 cytokines and chemokines are produced within 4 h of injection including IL-1beta and IL-5. Optimal production of some of these, including IL-1beta, depends upon both macrophages and mast cells, whereas production of others, such as IL-5, depends on mast cells only, suggesting that both of these cell types can detect alum. Alum induces eosinophil accumulation partly through the production of mast cell derived IL-5 and histamine. Alum greatly enhances priming of endogenous CD4 and CD8 T cells independently of mast cells, macrophages, and of eosinophils. In addition, Ab levels and Th2 bias was similar in the absence of these cells. We found that the inflammation induced by alum was unchanged in caspase-1-deficient mice, which cannot produce IL-1beta. Furthermore, endogenous CD4 and CD8 T cell responses, Ab responses and the Th2 bias were also not impacted by the absence of caspase-1 or NLRP3. These data suggest that activation of the inflammasome and the type 2 innate response orchestrated by macrophages and mast cells in vivo are not required for adjuvant effect of alum on endogenous T and B cell responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Immunity, Cellular/drug effects , Immunity, Innate/drug effects , Inflammation Mediators/pharmacology , Macrophages, Peritoneal/immunology , Mast Cells/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Amino Acid Sequence , Animals , Biosensing Techniques , Carrier Proteins/physiology , Caspase 1/physiology , Cell Movement/drug effects , Cell Movement/immunology , Inflammation Mediators/administration & dosage , Inflammation Mediators/classification , Injections, Intraperitoneal , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , NLR Family, Pyrin Domain-Containing 3 Protein
17.
Proc Natl Acad Sci U S A ; 105(38): 14521-6, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18787120

ABSTRACT

Immunological memory is a hallmark of adaptive immunity, and understanding T cell memory will be central to the development of effective cell-mediated vaccines. The characteristics and functions of CD4 memory cells have not been well defined. Here we demonstrate that the increased size of the secondary response is solely a consequence of the increased antigen-specific precursor frequency within the memory pool. Memory cells proliferated less than primary responding cells, even within the same host. By analyzing the entry of primary and memory cells into the cell cycle, we found that the two populations proliferated similarly until day 5; after this time, fewer of the reactivated memory cells proliferated. At this time, fewer of the reactivated memory cells made IL-2 than primary responding cells, but more made IFNgamma. Both these factors affected the low proliferation of the memory cells, because either exogenous IL-2 or inhibition of IFNgamma increased the proliferation of the memory cells.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Immunologic Memory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Division/immunology , Cell Movement , Female , Interferon-gamma/immunology , Interleukin-2/immunology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Time Factors
18.
BMC Biol ; 8: 37, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20385031

ABSTRACT

For many diseases vaccines are lacking or only partly effective. Research on protective immunity and adjuvants that generate vigorous immune responses may help generate effective vaccines against such pathogens.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibody Formation/immunology , Immunity, Cellular/immunology , Signal Transduction/immunology , Vaccination/methods , CD4-Positive T-Lymphocytes/immunology , Humans , Immunologic Memory/immunology
19.
Immunology ; 130(1): 10-5, 2010 May.
Article in English | MEDLINE | ID: mdl-20331469

ABSTRACT

Immunological memory is one of the features that define the adaptive immune response: by generating specific memory cells after infection or vaccination, the host provides itself with a set of cells and molecules that can prevent future infections and disease. Despite the obvious importance of memory cells, memory CD4 T cells are incompletely understood. Here we discuss recent progress in understanding which activated T cells surmount the barrier to enter into the memory pool and, once generated, what signals are important for memory cell survival. There is still, however, little understanding of how (or even whether) memory CD4 T cells are useful once they have been created; a surprising thought considering the critical role CD4 T cells play in all adaptive primary immune responses. In light of this, we will discuss how CD4 T memory T cells respond to reactivation in vivo and whether they are malleable to a re-assignment of their effector response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , Humans
20.
Mucosal Immunol ; 13(6): 946-957, 2020 11.
Article in English | MEDLINE | ID: mdl-32457450

ABSTRACT

FoxP3+ regulatory T cells (Tregs) control inflammation and maintain mucosal homeostasis, but their functions during infection are poorly understood. Th1, Th2, and Th17 cells can be identified by master transcription factors (TFs) T-bet, GATA3, and RORγT; Tregs also express these TFs. While T-bet+ Tregs can selectively suppress Th1 cells, it is unclear whether distinct Treg populations can alter Th bias. To address this, we used Salmonella enterica serotype Typhimurium to induce nonlethal colitis. Following infection, we observed an early colonic Th17 response within total CD4 T cells, followed by a Th1 bias. The early Th17 response, which contains both Salmonella-specific and non-Salmonella-specific cells, parallels an increase in T-bet+ Tregs. Later, Th1 cells and RORγT+ Tregs dominate. This reciprocal dynamic may indicate that Tregs selectively suppress Th cells, shaping the immune response. Treg depletion 1-2 days post-infection shifted the early Th17 response to a Th1 bias; however, Treg depletion 6-7 days post-infection abrogated the Th1 bias. Thus, Tregs are necessary for the early Th17 response, and for a maximal Th1 response later. These data show that Tregs shape the overall tissue CD4 T cell response and highlight the potential for subpopulations of Tregs to be used in targeted therapeutic approaches.


Subject(s)
Salmonella Infections/immunology , Salmonella/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cellular Microenvironment , Forkhead Transcription Factors/genetics , Lymphocyte Activation , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL