Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Publication year range
1.
Cell ; 185(9): 1588-1601.e14, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35413241

ABSTRACT

Immune memory is tailored by cues that lymphocytes perceive during priming. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic created a situation in which nascent memory could be tracked through additional antigen exposures. Both SARS-CoV-2 infection and vaccination induce multifaceted, functional immune memory, but together, they engender improved protection from disease, termed hybrid immunity. We therefore investigated how vaccine-induced memory is shaped by previous infection. We found that following vaccination, previously infected individuals generated more SARS-CoV-2 RBD-specific memory B cells and variant-neutralizing antibodies and a distinct population of IFN-γ and IL-10-expressing memory SARS-CoV-2 spike-specific CD4+ T cells than previously naive individuals. Although additional vaccination could increase humoral memory in previously naive individuals, it did not recapitulate the distinct CD4+ T cell cytokine profile observed in previously infected subjects. Thus, imprinted features of SARS-CoV-2-specific memory lymphocytes define hybrid immunity.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19 , SARS-CoV-2 , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/immunology , Humans , Immunity, Humoral , Spike Glycoprotein, Coronavirus , T-Lymphocytes
2.
Cell ; 184(1): 169-183.e17, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33296701

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus is causing a global pandemic, and cases continue to rise. Most infected individuals experience mildly symptomatic coronavirus disease 2019 (COVID-19), but it is unknown whether this can induce persistent immune memory that could contribute to immunity. We performed a longitudinal assessment of individuals recovered from mild COVID-19 to determine whether they develop and sustain multifaceted SARS-CoV-2-specific immunological memory. Recovered individuals developed SARS-CoV-2-specific immunoglobulin (IgG) antibodies, neutralizing plasma, and memory B and memory T cells that persisted for at least 3 months. Our data further reveal that SARS-CoV-2-specific IgG memory B cells increased over time. Additionally, SARS-CoV-2-specific memory lymphocytes exhibited characteristics associated with potent antiviral function: memory T cells secreted cytokines and expanded upon antigen re-encounter, whereas memory B cells expressed receptors capable of neutralizing virus when expressed as monoclonal antibodies. Therefore, mild COVID-19 elicits memory lymphocytes that persist and display functional hallmarks of antiviral immunity.


Subject(s)
COVID-19/immunology , COVID-19/physiopathology , Immunologic Memory , SARS-CoV-2/physiology , Adult , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , COVID-19/blood , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , SARS-CoV-2/chemistry , Severity of Illness Index , Spike Glycoprotein, Coronavirus/metabolism , T-Lymphocytes/immunology
3.
Cell ; 176(3): 610-624.e18, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612739

ABSTRACT

Plasma cells (PC) are found in the CNS of multiple sclerosis (MS) patients, yet their source and role in MS remains unclear. We find that some PC in the CNS of mice with experimental autoimmune encephalomyelitis (EAE) originate in the gut and produce immunoglobulin A (IgA). Moreover, we show that IgA+ PC are dramatically reduced in the gut during EAE, and likewise, a reduction in IgA-bound fecal bacteria is seen in MS patients during disease relapse. Removal of plasmablast (PB) plus PC resulted in exacerbated EAE that was normalized by the introduction of gut-derived IgA+ PC. Furthermore, mice with an over-abundance of IgA+ PB and/or PC were specifically resistant to the effector stage of EAE, and expression of interleukin (IL)-10 by PB plus PC was necessary and sufficient to confer resistance. Our data show that IgA+ PB and/or PC mobilized from the gut play an unexpected role in suppressing neuroinflammation.


Subject(s)
Immunoglobulin A/metabolism , Interleukin-10/metabolism , Intestines/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Neuroimmunomodulation/immunology , Plasma Cells/metabolism
4.
Immunity ; 55(1): 98-114.e5, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34932944

ABSTRACT

Elevated gene expression of the costimulatory receptor Icos is a hallmark of CD8+ tissue-resident memory (Trm) T cells. Here, we examined the contribution of ICOS in Trm cell differentiation. Upon transfer into WT mice, Icos-/- CD8+ T cells exhibited defective Trm generation but produced recirculating memory populations normally. ICOS deficiency or ICOS-L blockade compromised establishment of CD8+ Trm cells but not their maintenance. ICOS ligation during CD8+ T cell priming did not determine Trm induction; rather, effector CD8+ T cells showed reduced Trm differentiation after seeding into Icosl-/- mice. IcosYF/YF CD8+ T cells were compromised in Trm generation, indicating a critical role for PI3K signaling. Modest transcriptional changes in the few Icos-/- Trm cells suggest that ICOS-PI3K signaling primarily enhances the efficiency of CD8+ T cell tissue residency. Thus, local ICOS signaling promotes production of Trm cells, providing insight into the contribution of costimulatory signals in the generation of tissue-resident populations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Memory T Cells/immunology , Adoptive Transfer , Animals , Antibodies, Blocking/metabolism , Cell Differentiation , Cells, Cultured , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
5.
Nat Immunol ; 19(8): 838-848, 2018 08.
Article in English | MEDLINE | ID: mdl-29988091

ABSTRACT

Foxo transcription factors play an essential role in regulating specialized lymphocyte functions and in maintaining T cell quiescence. Here, we used a system in which Foxo1 transcription-factor activity, which is normally terminated upon cell activation, cannot be silenced, and we show that enforcing Foxo1 activity disrupts homeostasis of CD4 conventional and regulatory T cells. Despite limiting cell metabolism, continued Foxo1 activity is associated with increased activation of the kinase Akt and a cell-intrinsic proliferative advantage; however, survival and cell division are decreased in a competitive setting or growth-factor-limiting conditions. Via control of expression of the transcription factor Myc and the IL-2 receptor ß-chain, termination of Foxo1 signaling couples the increase in cellular cholesterol to biomass accumulation after activation, thereby facilitating immunological synapse formation and mTORC1 activity. These data reveal that Foxo1 regulates the integration of metabolic and mitogenic signals essential for T cell competitive fitness and the coordination of cell growth with cell division.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Forkhead Box Protein O1/metabolism , T-Lymphocytes, Regulatory/physiology , Animals , Cell Proliferation , Cells, Cultured , Cholesterol/metabolism , Forkhead Box Protein O1/genetics , Gene Expression Profiling , Homeostasis , Immunological Synapses/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
9.
J Immunol ; 208(7): 1519-1524, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35288472

ABSTRACT

Multiple sclerosis (MS) is a demyelinating inflammatory disease of the CNS treated by diverse disease-modifying therapies that suppress the immune system. Severe acute respiratory syndrome coronavirus 2 mRNA vaccines have been very effective in immunocompetent individuals, but whether MS patients treated with modifying therapies are afforded the same protection is not known. This study determined that dimethyl fumarate caused a momentary reduction in anti-Spike (S)-specific Abs and CD8 T cell response. MS patients treated with B cell-depleting (anti-CD20) or sphingosine 1-phosphate receptor agonist (fingolimod) therapies lack significant S-specific Ab response. Whereas S-specific CD4 and CD8 T cell responses were largely compromised by fingolimod treatment, T cell responses were robustly generated in anti-CD20-treated MS patients, but with a reduced proportion of CD4+CXCR5+ circulating follicular Th cells. These data provide novel information regarding vaccine immune response in patients with autoimmunity useful to help improve vaccine effectiveness in these populations.


Subject(s)
COVID-19 , Multiple Sclerosis , COVID-19 Vaccines , Humans , Immunologic Memory , Multiple Sclerosis/drug therapy , SARS-CoV-2
10.
Immunity ; 38(6): 1261-70, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23791647

ABSTRACT

Thymically derived Foxp3⁺ regulatory T (Treg) cells have a propensity to recognize self-peptide:MHC complexes, but their ability to respond to epitope-defined foreign antigens during infectious challenge has not been demonstrated. Here we show that pulmonary infection with Mycobacterium tuberculosis (Mtb), but not Listeria monocytogenes (Lm), induced robust lymph node expansion of a highly activated population of pathogen-specific Treg cells from the pre-existing pool of thymically derived Treg cells. These antigen-specific Treg cells peaked in numbers 3 weeks after infection but subsequently underwent selective elimination driven, in part, by interleukin-12-induced intrinsic expression of the Th1-cell-promoting transcription factor T-bet. Thus, the initial Mtb-induced inflammatory response promotes pathogen-specific Treg cell proliferation, but these cells are actively culled later, probably to prevent suppression during later stages of infection. These findings have important implications for the prevention and treatment of tuberculosis and other chronic diseases in which antigen-specific Treg cells restrict immunity.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-12/immunology , Mycobacterium tuberculosis/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Tuberculosis, Pulmonary/immunology , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Cell Proliferation , Cells, Cultured , Clonal Deletion , Epitopes, T-Lymphocyte/immunology , Forkhead Transcription Factors/genetics , Immune Evasion , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/immunology , T-Box Domain Proteins/genetics , T-Lymphocytes, Regulatory/microbiology , Thymus Gland/pathology
11.
J Immunol ; 205(10): 2667-2678, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33055282

ABSTRACT

IL-2 is a critical regulator of immune homeostasis through its impact on both regulatory T (Treg) and effector T cells. However, the precise role of IL-2 in the maintenance and function of Treg cells in the adult peripheral immune system remains unclear. In this study, we report that neutralization of IL-2 in mice abrogated all IL-2R signaling in Treg cells, but was well tolerated and only gradually impacted Treg cell function and immune homeostasis. By contrast, despite substantially reduced IL-2 sensitivity, Treg cells maintained selective IL-2 signaling and prevented immune dysregulation following treatment with the inhibitory anti-CD25 Ab PC61. Reduction of Treg cells with a depleting version of the same CD25 Ab permitted CD8+ effector T cell proliferation before progressing to more widespread immune dysregulation. Thus, despite severely curtailed CD25 expression and function, Treg cells retain selective access to IL-2 that supports their anti-inflammatory functions in vivo. Ab-mediated targeting of CD25 is being actively pursued for treatment of autoimmune disease and prevention of allograft rejection, and our findings help inform therapeutic manipulation and design for optimal patient outcomes.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cell Proliferation , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immune Tolerance/drug effects , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Models, Animal , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
12.
Am J Otolaryngol ; 43(4): 103483, 2022.
Article in English | MEDLINE | ID: mdl-35580419

ABSTRACT

OBJECTIVE: To compare the experiences of patients who received sialendoscopy under general anesthesia (GA) with those who received monitored anesthesia care (MAC). METHODS: Patients who underwent sialendoscopy for sialadenitis or sialolithiasis from July 1, 2020, to July 31, 2021, were offered inclusion to this prospective observational study. A survey was sent to consenting patients on post-operative day 1 to record aspects of their pre-, intra-, and post-operative experience. The primary outcome was overall satisfaction. Secondary outcomes included pain tolerability and preference for similar anesthetic modality in the future. RESULTS: Seventy-five patients completed the post-operative survey (86% response rate), of which 39 patients received GA and 36 received MAC. Patient overall satisfaction was similar between groups (GA: "Poor/Average/Good" = 23%, "Excellent" = 77%; MAC: "Poor/Average/Good" = 25%, "Excellent" = 75%, p = 1.00). Tolerability of immediate post-operative pain was likewise similar between the GA (82%) and MAC (97%) groups (p = 0.058). Patients who received MAC reported intra-operative pain as "none/tolerable" 72% of the time and "uncomfortable" 28% of the time. Patients who received GA would prefer the same anesthetic in the future more often than in the MAC group (85% versus 61%, respectively, OR 3.50, 95% CI 1.17-10.50, p = 0.035). CONCLUSION: In regard to patient satisfaction, both MAC and GA are acceptable anesthetic choices in sialendoscopy for appropriate cases. Patients report similar overall satisfaction and post-operative pain tolerance under either anesthetic modality. Patients who undergo GA report higher rates of preference for similar anesthetic modality in the future. Further study is needed to determine the most appropriate criteria for anesthesia modality selection.


Subject(s)
Anesthesia, General , Salivary Gland Calculi , Humans , Pain, Postoperative , Patient Outcome Assessment , Retrospective Studies , Salivary Gland Calculi/surgery , Treatment Outcome
13.
Nat Immunol ; 10(6): 595-602, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19412181

ABSTRACT

Several subsets of Foxp3(+) regulatory T cells (T(reg) cells) work in concert to maintain immune homeostasis. However, the molecular bases underlying the phenotypic and functional diversity of T(reg) cells remain obscure. We show that in response to interferon-gamma, Foxp3(+) T(reg) cells upregulated the T helper type 1 (T(H)1)-specifying transcription factor T-bet. T-bet promoted expression of the chemokine receptor CXCR3 on T(reg) cells, and T-bet(+) T(reg) cells accumulated at sites of T(H)1 cell-mediated inflammation. Furthermore, T-bet expression was required for the homeostasis and function of T(reg) cells during type 1 inflammation. Thus, in a subset of CD4(+) T cells, the activities of the transcription factors Foxp3 and T-bet are overlaid, which results in T(reg) cells with unique homeostatic and migratory properties optimized for the suppression of T(H)1 responses in vivo.


Subject(s)
Inflammation/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Homeostasis , Inflammation/metabolism , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/metabolism , T-Box Domain Proteins/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Up-Regulation
14.
Immunity ; 37(3): 501-10, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22960221

ABSTRACT

Foxp3(+) regulatory T (Treg) cells limit inflammatory responses and maintain immune homeostasis. Although comprised of several phenotypically and functionally distinct subsets, the differentiation of specialized Treg cell populations within the periphery is poorly characterized. We demonstrate that the development of T-bet(+) Treg cells that potently inhibit T helper 1 (Th1) cell responses was dependent on the transcription factor STAT1 and occurred directly in response to interferon-γ produced by effector T cells. Additionally, delayed induction of the IL-12Rß2 receptor component after STAT1 activation helped ensure that Treg cells do not readily complete STAT4-dependent Th1 cell development and lose their ability to suppress effector T cell proliferation. Thus, we define a pathway of abortive Th1 cell development that results in the specialization of peripheral Treg cells and demonstrate that impaired expression of a single cytokine receptor helps maintain Treg cell-suppressive function in the context of inflammatory Th1 cell responses.


Subject(s)
Cell Differentiation , Interleukin-12 Receptor beta 2 Subunit/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-12/pharmacology , Interleukin-12 Receptor beta 2 Subunit/genetics , Interleukin-12 Receptor beta 2 Subunit/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Time Factors
15.
J Immunol ; 202(1): 31-36, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30518568

ABSTRACT

Foxp3+ regulatory T (TR) cells are phenotypically and functionally diverse and broadly distributed in lymphoid and nonlymphoid tissues. However, the pathways guiding the differentiation of tissue-resident TR cell populations have not been well defined. By regulating E-protein function, Id3 controls the differentiation of CD8+ effector T cells and is essential for TR cell maintenance and function. We show that dynamic expression of Id3 helps define three distinct mouse TR cell populations: Id3+CD62LhiCD44lo central TR cells, Id3+CD62LloCD44hi effector TR (eTR) cells, and Id3- eTR cells. Adoptive transfer experiments and transcriptome analyses support a stepwise model of differentiation from Id3+ central TR to Id3+ eTR to Id3- eTR cells. Furthermore, Id3- eTR cells have high expression of functional inhibitory markers and a transcriptional signature of tissue-resident TR cells. Accordingly, Id3- eTR cells are highly enriched in nonlymphoid organs but virtually absent from blood and lymph. Thus, we propose that tissue-resident TR cells develop in a multistep process associated with Id3 downregulation.


Subject(s)
Blood Cells/immunology , Inhibitor of Differentiation Proteins/metabolism , Lymph Nodes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cell Differentiation , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation , Inhibitor of Differentiation Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
J Immunol ; 202(5): 1373-1382, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30683697

ABSTRACT

Abatacept is a CTLA-4-Ig fusion protein that binds to the costimulatory ligands CD80 and CD86 and blocks their interaction with the CD28 and CTLA-4 receptors expressed by T cells, therefore inhibiting T cell activation and function. Abatacept has shown clinical efficacy in treating some autoimmune diseases but has failed to show clinical benefit in other autoimmune conditions. The reasons for these disparate results are not clear and warrant further investigation of abatacept's mode of action. Longitudinal specimens from the Immune Tolerance Network's A Cooperative Clinical Study of Abatacept in Multiple Sclerosis trial were used to examine the effects of abatacept treatment on the frequency and transcriptional profile of specific T cell populations in peripheral blood. We found that the relative abundance of CD4+ T follicular helper (Tfh) cells and regulatory T cells was selectively decreased in participants following abatacept treatment. Within both cell types, abatacept reduced the proportion of activated cells expressing CD38 and ICOS and was associated with decreased expression of genes that regulate cell-cycle and chromatin dynamics during cell proliferation, thereby linking changes in costimulatory signaling to impaired activation, proliferation, and decreased abundance. All cellular and molecular changes were reversed following termination of abatacept treatment. These data expand upon the mechanism of action of abatacept reported in other autoimmune diseases and identify new transcriptional targets of CD28-mediated costimulatory signaling in human regulatory T and Tfh cells, further informing on its potential use in diseases associated with dysregulated Tfh activity.


Subject(s)
Abatacept/pharmacology , Immunosuppressive Agents/pharmacology , Multiple Sclerosis/drug therapy , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Cell Proliferation/drug effects , Double-Blind Method , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
18.
Immunity ; 35(3): 313-5, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21943484

ABSTRACT

The factors that control regulatory T (Treg) cell homeostasis and function are still being defined. In this issue of Immunity, Wang et al. (2011) demonstrate that the Th2 cell-associated transcription factor GATA-3 helps control Foxp3 expression in Treg cells and is required for their proper functional activity in vivo.

19.
J Immunol ; 197(7): 2635-45, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27566821

ABSTRACT

Phenotypically and functionally diverse regulatory T (Tr) cell subsets populate lymphoid and nonlymphoid tissues, where their maintenance and function are governed by unique homeostatic signals. Whereas Tr cells resident in nonlymphoid tissues depend on continual TCR signaling for their survival and function, phenotypically naive Tr cells occupying secondary lymphoid organs are largely supported by paracrine IL-2 signaling. Crucially, the absence of either of these distinct Tr cell subsets results in pathogenic autoimmunity, underscoring their nonredundant roles in the preservation of self-tolerance. However, the cellular and molecular factors precipitating IL-2 release and subsequent maintenance of secondary lymphoid organ-resident Tr cells are still poorly understood. In this study, we report that IL-2-dependent Tr cells in the spleen compete for a limiting supply of paracrine IL-2 generated by autoreactive CD4(+) T cells in response to MHC class II-restricted autoantigen activation by 33D1(+)CD11b(int) dendritic cells. Manipulating this cellular circuit culminating in IL-2 production could have clinical benefits in settings in which diminished Tr cell abundance is desired.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interleukin-2/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Coculture Techniques , Interleukin-2/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Spleen/cytology
20.
J Immunol ; 197(12): 4541-4551, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27815425

ABSTRACT

Human asthma is a heterogeneous disease characterized by the expression of both Th2 and Th17 cytokines. In vitro and in vivo studies have shown a reciprocal regulation between Th2 and Th17 pathways, suggesting a potential induction of neutrophil-promoting Th17 inflammation in the absence of a Th2 response. Alternaria alternata is a clinically relevant allergen that is associated with severe and fatal asthma exacerbations. Exposure to A. alternata is characterized by a predominant Th2 response, but can also induce the production of factors associated with Th17 responses (e.g., CXCL8) from epithelial cells. Using a mouse model, we found that wild-type mice develop an eosinophilic Th2 airway disease in response to A. alternata exposure, whereas IL-4-, IL-13-, and STAT6-deficient mice exhibit a primarily neutrophilic response. Neutrophilic asthma in STAT6-/- mice was accompanied by elevated lung levels of TNF-α, CXCL1, CXCL2, and CXCL5, and was steroid resistant. Neutralization of Th17 signaling only partially reduced neutrophil numbers and total airway inflammation. Airway neutrophilia developed in RAG-deficient and CD4-depleted BALB/c mice, suggesting that the suppression of neutrophil responses is dependent on Th2 cytokine production by T cells and that airway neutrophilia is primarily an innate response to allergen. These results highlight the importance of combination therapies for treatment of asthma and establish a role for factors other than IL-17 as targets for neutrophilic asthma.


Subject(s)
Alternaria/immunology , Asthma/immunology , Eosinophils/immunology , Interleukin-13/metabolism , Interleukin-4/metabolism , Neutrophils/immunology , STAT6 Transcription Factor/metabolism , Allergens/immunology , Animals , Antigens, Fungal/immunology , Cell Extracts/immunology , Disease Models, Animal , Female , Interleukin-13/genetics , Interleukin-4/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , STAT6 Transcription Factor/genetics , Transplantation Chimera
SELECTION OF CITATIONS
SEARCH DETAIL