Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(36): e2404916121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39207730

ABSTRACT

Identifying tumor-mediated mechanisms that impair immunity is instrumental for the design of new cancer therapies. Regulatory T cells (Tregs) are a key component of cancer-derived immune suppression; however, these lymphocytes are necessary to prevent systemic autoimmunity in mice and humans, and thus, direct targeting of Tregs is not a clinical option for cancer patients. We have previously demonstrated that excising transcription factor Kruppel-like factor 2 (Klf2) within the T cell lineage blocks the generation of peripheral-derived Tregs (pTregs) without impairing production of thymic-derived Tregs. Using this mouse model, we have now demonstrated that eliminating pTregs is sufficient to delay/prevent tumor malignancy without causing autoimmunity. Cancer-bearing mice that expressed KLF2 converted tumor-specific CD4+ T cells into pTregs, which accumulated in secondary lymphoid organs and impaired further T cell effector activity. In contrast, pTreg-deficient mice retained cancer-specific immunity, including improved T cell infiltration into "cold" tumors, reduced T cell exhaustion in tumor beds, restricted generation of tumor-associated myeloid-derived suppressor cells, and the continued production of circulating effector T cells that arose in a cancer-dependent manner. Results indicate that tumor-specific pTregs are critical for early stages of cancer progression and blocking the generation of these inhibitory lymphocytes safely delays/prevents malignancy in preclinical models of melanoma and prostate cancer.


Subject(s)
Kruppel-Like Transcription Factors , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , Mice , Kruppel-Like Transcription Factors/metabolism , Male , Mice, Inbred C57BL , Immune Tolerance/immunology , Humans
2.
3.
Proc Natl Acad Sci U S A ; 120(1): e2214874120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574710

ABSTRACT

Adequate mass and function of adipose tissues (ATs) play essential roles in preventing metabolic perturbations. The pathological reduction of ATs in lipodystrophy leads to an array of metabolic diseases. Understanding the underlying mechanisms may benefit the development of effective therapies. Several cellular processes, including autophagy and vesicle trafficking, function collectively to maintain AT homeostasis. Here, we investigated the impact of adipocyte-specific deletion of the lipid kinase phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3) on AT homeostasis and systemic metabolism in mice. We report that PIK3C3 functions in all ATs and that its absence disturbs adipocyte autophagy and hinders adipocyte differentiation, survival, and function with differential effects on brown and white ATs. These abnormalities cause loss of white ATs, whitening followed by loss of brown ATs, and impaired "browning" of white ATs. Consequently, mice exhibit compromised thermogenic capacity and develop dyslipidemia, hepatic steatosis, insulin resistance, and type 2 diabetes. While these effects of PIK3C3 largely contrast previous findings with the autophagy-related (ATG) protein ATG7 in adipocytes, mice with a combined deficiency in both factors reveal a dominant role of the PIK3C3-deficient phenotype. We have also found that dietary lipid excess exacerbates AT pathologies caused by PIK3C3 deficiency. Surprisingly, glucose tolerance is spared in adipocyte-specific PIK3C3-deficient mice, a phenotype that is more evident during dietary lipid excess. These findings reveal a crucial yet complex role for PIK3C3 in ATs, with potential therapeutic implications.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Mice , Class III Phosphatidylinositol 3-Kinases/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Diabetes Mellitus, Type 2/metabolism , Adipocytes/metabolism , Lipids , Adipose Tissue, Brown/metabolism , Adipocytes, Brown/metabolism
4.
Inflamm Res ; 73(9): 1511-1527, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39028491

ABSTRACT

BACKGROUND: Invariant natural killer T (iNKT) cells play protective or pathogenic roles in a variety of immune and inflammatory diseases. However, whether iNKT cells contribute to the progression of acute neuroinflammation remains unclear. Thus, we addressed this question with a mouse model of lipopolysaccharide (LPS)-induced acute neuroinflammation. METHODS: For induction of acute neuroinflammation, wild-type (WT) C57BL/6 (B6) mice were injected intraperitoneally (i.p.) with LPS for either three or five consecutive days, and then these mice were analyzed for brain-infiltrating leukocytes or mouse behaviors, respectively. To examine the role of iNKT cell activation in LPS-induced neuroinflammation, mice were injected i.p. with the iNKT cell agonist α-galactosylceramide (α-GalCer) seven days prior to LPS treatment. Immune cells infiltrated into the brain during LPS-induced neuroinflammation were determined by flow cytometry. In addition, LPS-induced clinical behavior symptoms such as depressive-like behavior and memory impairment in mice were evaluated by the open field and Y-maze tests, respectively. RESULTS: We found that iNKT cell-deficient Jα18 mutant mice display delayed disease progression and decreased leukocyte infiltration into the brain compared with WT mice, indicating that iNKT cells contribute to the pathogenesis of LPS-induced neuroinflammation. Since it has been reported that pre-treatment with α-GalCer, an iNKT cell agonist, can convert iNKT cells towards anti-inflammatory phenotypes, we next explored whether pre-activation of iNKT cells with α-GalCer can regulate LPS-induced neuroinflammation. Strikingly, we found that α-GalCer pre-treatment significantly delays the onset of clinical symptoms, including depression-like behavior and memory impairment, while decreasing brain infiltration of pro-inflammatory natural killer cells and neutrophils, in this model of LPS-induced neuroinflammation. Such anti-inflammatory effects of α-GalCer pre-treatment closely correlated with iNKT cell polarization towards IL4- and IL10-producing phenotypes. Furthermore, α-GalCer pre-treatment restored the expression of suppressive markers on brain regulatory T cells during LPS-induced neuroinflammation. CONCLUSION: Our findings provide strong evidence that α-GalCer-induced pre-activation of iNKT cells expands iNKT10 cells, mitigating depressive-like behaviors and brain infiltration of inflammatory immune cells induced by LPS-induced acute neuroinflammation. Thus, we suggest the prophylactic potential of iNKT cells and α-GalCer against acute neuroinflammation.


Subject(s)
Brain , Galactosylceramides , Lipopolysaccharides , Mice, Inbred C57BL , Natural Killer T-Cells , Neuroinflammatory Diseases , Animals , Galactosylceramides/pharmacology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/drug effects , Brain/pathology , Brain/drug effects , Brain/immunology , Male , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/chemically induced , Mice , Cytokines/metabolism
5.
J Immunol ; 209(2): 199-207, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35821102

ABSTRACT

Lymphocytes can be functionally partitioned into subsets belonging to the innate or adaptive arms of the immune system. Subsets of innate and innate-like lymphocytes may or may not express Ag-specific receptors of the adaptive immune system, yet they are poised to respond with innate-like speed to pathogenic insults but lack the capacity to develop classical immunological memory. These lymphocyte subsets display a number of common properties that permit them to integrate danger and stress signals dispatched by innate sensor cells to facilitate the generation of specialized effector immune responses tailored toward specific pathogens or other insults. In this review, we discuss the functions of distinct subsets of innate and innate-like lymphocytes. A better understanding of the mechanisms by which these cells are activated in different contexts, their interactions with other immune cells, and their role in health and disease may inform the development of new or improved immunotherapies.


Subject(s)
Immunity, Innate , Lymphocytes , Immunologic Memory , Immunotherapy
6.
Nat Immunol ; 12(11): 1086-95, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964609

ABSTRACT

The presence of immune memory at pathogen-entry sites is a prerequisite for protection. Nevertheless, the mechanisms that warrant immunity at peripheral interfaces are not understood. Here we show that the nonclassical major histocompatibility complex (MHC) class I molecule thymus leukemia antigen (TL), induced on dendritic cells interacting with CD8αα on activated CD8αß(+) T cells, mediated affinity-based selection of memory precursor cells. Furthermore, constitutive expression of TL on epithelial cells led to continued selection of mature CD8αß(+) memory T cells. The memory process driven by TL and CD8αα was essential for the generation of CD8αß(+) memory T cells in the intestine and the accumulation of highly antigen-sensitive CD8αß(+) memory T cells that form the first line of defense at the largest entry port for pathogens.


Subject(s)
Dendritic Cells/metabolism , Listeriosis/immunology , Membrane Glycoproteins/metabolism , Precursor Cells, T-Lymphoid/metabolism , T-Lymphocytes/metabolism , Animals , Antigens/immunology , Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Clonal Selection, Antigen-Mediated , Dendritic Cells/immunology , Dendritic Cells/pathology , Immunity, Mucosal/genetics , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transgenes/genetics
7.
Immunity ; 41(3): 451-464, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25220211

ABSTRACT

Innate immune responses are critical for mucosal immunity. Here we describe an innate lymphocyte population, iCD8α cells, characterized by expression of CD8α homodimers. iCD8α cells exhibit innate functional characteristics such as the capacity to engulf and kill bacteria. Development of iCD8α cells depends on expression of interleukin-2 receptor γ chain (IL-2Rγc), IL-15, and the major histocompatibility complex (MHC) class Ib protein H2-T3, also known as the thymus leukemia antigen or TL. While lineage tracking experiments indicated that iCD8α cells have a lymphoid origin, their development was independent of the transcriptional suppressor Id2, suggesting that these cells do not belong to the family of innate lymphoid cells. Finally, we identified cells with a similar phenotype in humans, which were profoundly depleted in newborns with necrotizing enterocolitis. These findings suggest a critical role of iCD8α cells in immune responses associated with the intestinal epithelium.


Subject(s)
Antigen Presentation/immunology , CD8 Antigens/biosynthesis , Immunity, Mucosal/immunology , Intestinal Mucosa/cytology , Lymphocytes/immunology , Animals , Citrobacter rodentium/immunology , Cytochalasin D/pharmacology , Enterocolitis, Necrotizing , Helicobacter pylori/immunology , Histocompatibility Antigens Class I/biosynthesis , Humans , Inhibitor of Differentiation Protein 2/genetics , Interleukin Receptor Common gamma Subunit/biosynthesis , Interleukin-15/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7/biosynthesis , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Lymphocytes/classification , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Phagocytosis/immunology
8.
Proc Natl Acad Sci U S A ; 117(29): 17156-17165, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32611812

ABSTRACT

Semi-invariant natural killer T (iNKT) cells are self-reactive lymphocytes, yet how this lineage attains self-tolerance remains unknown. iNKT cells constitutively express high levels of Nr4a1-encoded Nur77, a transcription factor that integrates signal strength downstream of the T cell receptor (TCR) within activated thymocytes and peripheral T cells. The function of Nur77 in iNKT cells is unknown. Here we report that sustained Nur77 overexpression (Nur77tg) in mouse thymocytes abrogates iNKT cell development. Introgression of a rearranged Vα14-Jα18 TCR-α chain gene into the Nur77tg (Nur77tg;Vα14tg) mouse rescued iNKT cell development up to the early precursor stage, stage 0. iNKT cells in bone marrow chimeras that reconstituted thymic cellularity developed beyond stage 0 precursors and yielded IL-4-producing NKT2 cell subset but not IFN-γ-producing NKT1 cell subset. Nonetheless, the developing thymic iNKT cells that emerged in these chimeras expressed the exhaustion marker PD1 and responded poorly to a strong glycolipid agonist. Thus, Nur77 integrates signals emanating from the TCR to control thymic iNKT cell tolerance induction, terminal differentiation, and effector functions.


Subject(s)
Cell Differentiation , Immune Tolerance , Natural Killer T-Cells , Nuclear Receptor Subfamily 4, Group A, Member 1 , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Immune Tolerance/genetics , Immune Tolerance/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Antigen, T-Cell , Thymocytes
9.
Int J Mol Sci ; 24(12)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37373104

ABSTRACT

Interleukin-7 (IL-7) plays a vital role in the homeostasis of CD4+ and CD8+ T cells. Although IL-7 has been implicated in T helper (Th)1- and Th17-mediated autoinflammatory diseases, its role in Th2-type allergic disorders, such as atopic dermatitis (AD), remains unclear. Thus, to elucidate the effects of IL-7 deficiency on AD development, we generated IL-7-deficient AD-prone mice by backcrossing IL-7 knockout (KO) B6 mice onto the NC/Nga (NC) mouse strain, a model for human AD. As expected, IL-7 KO NC mice displayed defective development of conventional CD4+ and CD8+ T cells compared with wild type (WT) NC mice. However, IL-7 KO NC mice presented with enhanced AD clinical scores, IgE hyperproduction, and increased epidermal thickness compared with WT NC mice. Moreover, IL-7 deficiency decreased Th1, Th17, and IFN-γ-producing CD8+ T cells but increased Th2 cells in the spleen of NC mice, indicating that a reduced Th1/Th2 ratio correlates with severity of AD pathogenesis. Furthermore, significantly more basophils and mast cells infiltrated the skin lesions of IL-7 KO NC mice. Taken together, our findings suggest that IL-7 could be a useful therapeutic target for treating Th2-mediated skin inflammations, such as AD.


Subject(s)
Dermatitis, Atopic , Skin Diseases , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/pathology , Cytokines , Dermatitis, Atopic/drug therapy , Disease Models, Animal , Interleukin-7/genetics , Interleukin-7/metabolism , Skin/pathology , Skin Diseases/pathology , Th2 Cells
10.
Molecules ; 28(15)2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37570749

ABSTRACT

Beta-glucan (ß-glucan) is a natural polysaccharide produced by fungi, bacteria, and plants. Although it has been reported that ß-glucan enhances innate immune memory responses, it is unclear whether different types of ß-glucans display similar immune effects. To address this issue, we employed zymosan (ß-1,3-glycosidic linkage) and pustulan (ß-1,6-glycosidic linkage) to investigate their in vivo effects on innate memory immune responses. We examined the changes of innate memory-related markers in macrophages and natural killer (NK) cells, two immune cell types that display innate memory characteristics, at two different time points (16 h and 7 days) after ß-glucan stimulation. We found that short-term (16 h) zymosan treatment significantly induced macrophages to upregulate IL15 production and increased surface IL15Rα expression on NK cells. In addition, long-term (7 days) zymosan treatment significantly induced macrophages to upregulate the expression of innate memory-related markers (e.g., TNFα, HIF1α, and mTOR) and induced NK cells to express enhanced levels of KLRG1, known as an innate memory-like marker. Our results provide support that zymosan can be an effective adjuvant to promote innate memory immune responses, providing a bridge between innate and adaptive immune cells to enhance various immune responses such as those directed against tumors.


Subject(s)
Interleukin-15 , beta-Glucans , Mice , Animals , Zymosan/pharmacology , Macrophages , beta-Glucans/pharmacology , Killer Cells, Natural , Immunity, Innate
11.
Immunology ; 165(4): 414-427, 2022 04.
Article in English | MEDLINE | ID: mdl-35137411

ABSTRACT

Obesity is accompanied by and accelerated with chronic inflammation in adipose tissue, especially visceral adipose tissue (VAT). This low-level inflammation predisposes the host to the development of metabolic disease, most notably type 2 diabetes. We have focused on the capacity of glycolipid-reactive, CD1d-restricted natural killer T (NKT) cells to modulate obesity and its associated metabolic sequelae. We previously reported that CD1d knockout (KO) mice are partially protected against the development of obesity-associated insulin resistance, and these findings were recapitulated in mice with an adipocyte-specific CD1d deficiency, suggesting that NKT cell-adipocyte interactions play a critical role in exacerbating disease. However, many other CD1d-expressing cells contribute to the in vivo responses of NKT cells to lipid antigens. In the present study, we examined the role of CD1d expression by macrophages (Mϕ) in the development of obesity-associated metabolic inflammation using LysMcre-cd1d1f/f mice where the CD1d1 gene is disrupted in a Mϕ-specific manner. Unexpectedly, these animals contained a higher frequency of T-bet+ CD4+ T cells in VAT with increased production of Th1 cytokines that aggravated VAT inflammation. Mϕ from mutant mice displayed increased production of IL-12p40, suggesting M1 polarization. These findings indicate that interactions of CD1d on Mϕ with NKT cells play a beneficial role in obesity-associated VAT inflammation and insulin resistance with a sharp contrast to an aggravating role of CD1d in another type of antigen-presenting cell, dendritic cells.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Natural Killer T-Cells , Adipose Tissue/metabolism , Animals , Antigens, CD1d , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism
12.
PLoS Pathog ; 16(5): e1008356, 2020 05.
Article in English | MEDLINE | ID: mdl-32437421

ABSTRACT

Tuberculosis (TB) is one of the deadliest diseases, claiming ~2 million deaths annually worldwide. The majority of people in TB endemic regions are vaccinated with Bacillus Calmette Guerin (BCG), which is the only usable vaccine available. BCG is efficacious against meningeal and disseminated TB in children, but protective responses are relatively short-lived and fail to protect against adult pulmonary TB. The longevity of vaccine efficacy critically depends on the magnitude of long-lasting central memory T (TCM) cells, a major source of which is stem cell-like memory T (TSM) cells. These TSM cells exhibit enhanced self-renewal capacity as well as to rapidly respond to antigen and generate protective poly-functional T cells producing IFN-γ, TNF-α, IL-2 and IL-17. It is now evident that T helper Th 1 and Th17 cells are essential for host protection against TB. Recent reports have indicated that Th17 cells preserve the molecular signature for TSM cells, which eventually differentiate into IFN-γ-producing effector cells. BCG is ineffective in inducing Th17 cell responses, which might explain its inadequate vaccine efficacy. Here, we show that revaccination with BCG along with clofazimine treatment promotes TSM differentiation, which continuously restores TCM and T effector memory (TEM) cells and drastically increases vaccine efficacy in BCG-primed animals. Analyses of these TSM cells revealed that they are predominantly precursors to host protective Th1 and Th17 cells. Taken together, these findings revealed that clofazimine treatment at the time of BCG revaccination provides superior host protection against TB by increasing long-lasting TSM cells.


Subject(s)
BCG Vaccine/immunology , BCG Vaccine/metabolism , Clofazimine/pharmacology , Immunologic Memory/immunology , Animals , BCG Vaccine/pharmacology , Clofazimine/metabolism , Drug Therapy, Combination/methods , Female , Immunization, Secondary/methods , Immunogenicity, Vaccine/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Stem Cells/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Tuberculosis/immunology , Tuberculosis, Pulmonary/immunology
13.
Crit Rev Immunol ; 41(4): 55-88, 2021.
Article in English | MEDLINE | ID: mdl-35381143

ABSTRACT

It is now appreciated that a group of lymphoid lineage cells, collectively called innate-like effector lymphocytes, have evolved to integrate information relayed by the innate sensory immune system about the state of the local tissue environment and to pass on this context to downstream effector innate and adaptive immune responses. Thereby, innate functions engrained into such innate-like lymphoid lineage cells during development can control the quality and magnitude of an immune response to a tissue-altering pathogen and facilitate the formation of memory engrams within the immune system. These goals are accomplished by the innate lymphoid cells that lack antigen-specific receptors, γδ T cell receptor (TCR)-expressing T cells, and several αß TCR-expressing T cell subsets-such as natural killer T cells, mucosal-associated invariant T cells, et cetera. Whilst we briefly consider the commonalities in the origins and functions of these diverse lymphoid subsets to provide context, the primary topic of this review is to discuss how the semi-invariant natural killer T cells got this way in evolution through lineage commitment and onward ontogeny. What emerges from this discourse is the question: Has a "limbic immune system" emerged (screaming quietly in plain sight!) out of what has been dubbed "in-betweeners"?


Subject(s)
Natural Killer T-Cells , Humans , Immunity, Innate , Killer Cells, Natural , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets
14.
Proc Natl Acad Sci U S A ; 116(43): 21673-21684, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31591231

ABSTRACT

Acute myocardial infarction (MI) provokes an inflammatory response in the heart that removes damaged tissues to facilitate tissue repair/regeneration. However, overactive and prolonged inflammation compromises healing, which may be counteracted by antiinflammatory mechanisms. A key regulatory factor in an inflammatory response is the antiinflammatory cytokine IL-10, which can be produced by a number of immune cells, including subsets of B lymphocytes. Here, we investigated IL-10-producing B cells in pericardial adipose tissues (PATs) and their role in the healing process following acute MI in mice. We found that IL-10-producing B cells were enriched in PATs compared to other adipose depots throughout the body, with the majority of them bearing a surface phenotype consistent with CD5+ B-1a cells (CD5+ B cells). These cells were detected early in life, maintained a steady presence during adulthood, and resided in fat-associated lymphoid clusters. The cytokine IL-33 and the chemokine CXCL13 were preferentially expressed in PATs and contributed to the enrichment of IL-10-producing CD5+ B cells. Following acute MI, the pool of CD5+ B cells was expanded in PATs. These cells accumulated in the infarcted heart during the resolution of MI-induced inflammation. B cell-specific deletion of IL-10 worsened cardiac function, exacerbated myocardial injury, and delayed resolution of inflammation following acute MI. These results revealed enrichment of IL-10-producing B cells in PATs and a significant contribution of these cells to the antiinflammatory processes that terminate MI-induced inflammation. Together, these findings have identified IL-10-producing B cells as therapeutic targets to improve the outcome of MI.


Subject(s)
Adipose Tissue/metabolism , B-Lymphocytes/immunology , Interleukin-10/metabolism , Myocardial Infarction/immunology , Pericardium/metabolism , Adipose Tissue/cytology , Animals , Chemokine CXCL13/metabolism , Female , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-33/metabolism , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Pericardium/cytology , Regeneration/physiology
15.
Int J Mol Sci ; 23(23)2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36499642

ABSTRACT

We have previously shown that a deficiency of CD1d-restricted invariant natural killer T (iNKT) cells exacerbates dextran sulfate sodium (DSS)-induced colitis in Yeti mice that exhibit IFNγ-mediated hyper-inflammation. Although iNKT cell-deficiency resulted in reduced Foxp3 expression by mesenteric lymph node (MLN) CD4+ T cells in DSS-treated Yeti mice, the cellular mechanisms that regulate Foxp3 expression by CD25+CD4+ T cells during intestinal inflammation remain unclear. We found that Foxp3-CD25+CD4+ T cells expressing Th1 and Th17 phenotypic hallmarks preferentially expanded in the MLNs of DSS-treated Yeti/CD1d knockout (KO) mice. Moreover, adoptive transfer of Yeti iNKT cells into iNKT cell-deficient Jα18 KO mice effectively suppressed the expansion of MLN Foxp3-CD25+CD4+ T cells during DSS-induced colitis. Interestingly, MLN dendritic cells (DCs) purified from DSS-treated Yeti/CD1d KO mice promoted the differentiation of naive CD4+ T cells into Foxp3-CD25+CD4+ T cells rather than regulatory T (Treg) cells, indicating that MLN DCs might mediate Foxp3+CD25+CD4+ T cell expansion in iNKT cell-sufficient Yeti mice. Furthermore, we showed that Foxp3-CD25+CD4+ T cells were pathogenic in DSS-treated Yeti/CD1d KO mice. Our result suggests that pro-inflammatory DCs and CD1d-restricted iNKT cells play opposing roles in Foxp3 expression by MLN CD25+CD4+ T cells during IFNγ-mediated intestinal inflammation, with potential therapeutic implications.


Subject(s)
Colitis , Dendritic Cells , Natural Killer T-Cells , Animals , Mice , Colitis/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Interferon-gamma/metabolism , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , CD4-Positive T-Lymphocytes/immunology , Interleukin-2 Receptor alpha Subunit , Dendritic Cells/immunology , Dendritic Cells/metabolism
16.
Molecules ; 27(24)2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36557876

ABSTRACT

Although 3-aminopropyl functionalized magnesium phyllosilicate nanoparticles (hereafter aminoclay nanoparticles, ACNs) are well-known nanomaterials employed as drug carriers, their effects on immune cells remain unclear. To address this issue, we explored murine dendritic cells (DCs) as these cells belong to the innate arm of the immune system and function as antigen-presenting cells to elicit adaptive immune responses. We examined the in vitro effects of ACNs on DCs isolated from B6 mice. ACN treatment significantly down-regulated the expression of inflammasome-related markers, including NLRP3, caspase-1, and IL1ß. The ACNs-induced anti-inflammatory DC phenotype was further confirmed by down-regulation of the AKT/mTOR/HIF1α signaling pathway. Such anti-inflammatory effects of ACNs on DCs occurred independently of DC subtypes. To document the effects of ACNs on DCs more clearly, we examined their anti-inflammatory effects on lipopolysaccharide (LPS)-activated DCs. As expected, excessive inflammatory responses (increased mitochondrial ROS and Th1-type cytokines such as IL12 and IL1ß) of LPS-activated DCs were dramatically attenuated by ACN treatment. Furthermore, ACNs down-regulated IFNγ production by antigen-specific CD4+ T cells, which is consistent with a reduced inflammatory phenotype of DCs. Overall, our results provide support for employing ACNs as drug delivery materials with therapeutic potential to control inflammatory disorders.


Subject(s)
Lipopolysaccharides , Nanoparticles , Animals , Mice , Lipopolysaccharides/pharmacology , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Immunity , Dendritic Cells
17.
Trends Immunol ; 39(4): 264-275, 2018 04.
Article in English | MEDLINE | ID: mdl-29221933

ABSTRACT

Intestinal intraepithelial lymphocytes (IELs) are a large and diverse population of lymphoid cells that reside between the intestinal epithelial cells (IECs) that form the intestinal mucosal barrier. Although IEL biology has traditionally focused on T cells, recent studies have identified several subsets of T cell receptor (TCR)-negative IELs with intriguing properties. New insight into the development, homeostasis, and functions of distinct IEL subsets has recently been provided. Additional studies have revealed intricate interactions between different IEL subsets, reciprocal interactions between IELs and IECs, and communication of IELs with immune cells that reside outside the intestinal epithelium. We review here sentinel functions of IELs in the maintenance of the mucosal barrier integrity, as well as how dysregulated IEL responses can contribute to pathology.


Subject(s)
Inflammation/immunology , Intestinal Mucosa/physiology , Intraepithelial Lymphocytes/immunology , Animals , Cell Communication , Cell Differentiation , Homeostasis , Humans , Immunity, Mucosal
18.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513946

ABSTRACT

We have previously shown that CD1d-restricted iNKT cells suppress dysregulated IFNγ expression and intestinal inflammation in Yeti mice on the C57BL/6 background. Since type 3 innate lymphoid cells (ILC3s) in mesenteric lymph nodes (MLN) protect against intestinal inflammation in a CD1d-associated manner, we investigated whether crosstalk between iNKT cells and MLN ILC3s controls IFNγ-mediated intestinal inflammation in Yeti mice. We found that Yeti mice display increased levels of ILC3s and that iNKT cell deficiency in Yeti/CD1d KO mice decreases levels of IL22-producing ILC3s during DSS-induced colitis. This finding indicates that iNKT cells and ILC3s cooperate to regulate intestinal inflammation in Yeti mice. Yeti iNKT cells displayed a pronounced anti-inflammatory (IL4- or IL9-producing) phenotype during colitis. Their adoptive transfer to iNKT cell-deficient animals induced a significant increase in IL22 production by ILC3s, indicating that crosstalk between iNKT cells and ILC3s plays a critical role in modulating colitis in Yeti mice. Moreover, we showed that the IL9-producing subset of iNKT cells potently enhances IL22-producing ILC3s in vivo. Taken together, our results identify a central role of the iNKT cell-ILC3 axis in ameliorating IFNγ-mediated intestinal inflammation.


Subject(s)
Antigens, CD1d/genetics , Inflammation/genetics , Interferon-gamma/genetics , Interleukins/genetics , Lymphocytes/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Dextran Sulfate/toxicity , Humans , Immunity, Innate/genetics , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/pathology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Splanchnic Circulation/genetics , Interleukin-22
19.
Int J Mol Sci ; 22(6)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33809795

ABSTRACT

We previously showed that ubiquitous overexpression of the chromatin remodeling factor SWItch3-related gene (SRG3) promotes M2 macrophage differentiation, resulting in anti-inflammatory responses in the experimental autoimmune encephalomyelitis model of multiple sclerosis. Since hepatic macrophages are responsible for sepsis-induced liver injury, we investigated herein the capacity of transgenic SRG3 overexpression (SRG3ß-actin mice) to modulate sepsis in mice exposed to lipopolysaccharide (LPS) plus d-galactosamine (d-GalN). Our results demonstrated that ubiquitous SRG3 overexpression significantly protects mice from LPS/d-GalN-induced lethality mediated by hepatic M1 macrophages. These protective effects of SRG3 overexpression correlated with the phenotypic conversion of hepatic macrophages from an M1 toward an M2 phenotype. Furthermore, SRG3ß-actin mice had decreased numbers and activation of natural killer (NK) cells but not natural killer T (NKT) cells in the liver during sepsis, indicating that SRG3 overexpression might contribute to cross-talk between NK cells and macrophages in the liver. Finally, we demonstrated that NKT cell-deficient CD1d KO/SRG3ß-actin mice are protected from LPS/d-GalN-induced sepsis, indicating that NKT cells are dispensable for SRG3-mediated sepsis suppression. Taken together, our findings provide strong evidence that SRG3 overexpression may serve as a therapeutic approach to control overwhelming inflammatory diseases such as sepsis.


Subject(s)
Chromatin/metabolism , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Liver/pathology , Macrophages/metabolism , Natural Killer T-Cells/metabolism , Sepsis/chemically induced , Sepsis/prevention & control , Transcription Factors/metabolism , Actins/genetics , Animals , Chromatin Assembly and Disassembly , Dendritic Cells/metabolism , Galactosamine , Inflammation Mediators/metabolism , Lipopolysaccharides , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protective Agents/metabolism , Sepsis/immunology , Sepsis/pathology , Severity of Illness Index
20.
Int J Mol Sci ; 22(4)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557054

ABSTRACT

The SWItch (SWI)3-related gene (SRG3) product, a SWI/Sucrose Non-Fermenting (SNF) chromatin remodeling subunit, plays a critical role in regulating immune responses. We have previously shown that ubiquitous SRG3 overexpression attenuates the progression of Th1/Th17-mediated experimental autoimmune encephalomyelitis. However, it is unclear whether SRG3 overexpression can affect the pathogenesis of inflammatory skin diseases such as atopic dermatitis (AD), a Th2-type immune disorder. Thus, to elucidate the effects of SRG3 overexpression in AD development, we bred NC/Nga (NC) mice with transgenic mice where SRG3 expression is driven by the ß-actin promoter (SRG3ß-actin mice). We found that SRG3ß-actin NC mice exhibit increased AD development (e.g., a higher clinical score, immunoglobulin E (IgE) hyperproduction, and an increased number of infiltrated mast cells and basophils in skin lesions) compared with wild-type NC mice. Moreover, the severity of AD pathogenesis in SRG3ß-actin NC mice correlated with expansion of interleukin 4 (IL4)-producing basophils and mast cells, and M2 macrophages. Furthermore, this accelerated AD development is strongly associated with Treg cell suppression. Collectively, our results have identified that modulation of SRG3 function can be applied as one of the options to control AD pathogenesis.


Subject(s)
Chromatin Assembly and Disassembly , Dermatitis, Atopic/etiology , Gene Expression , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription Factors/genetics , Actins/metabolism , Animals , Biopsy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/metabolism , Disease Models, Animal , Disease Susceptibility , Immunity, Cellular , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL