Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Immunity ; 56(5): 1064-1081.e10, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36948193

ABSTRACT

The recent revolution in tissue-resident macrophage biology has resulted largely from murine studies performed in C57BL/6 mice. Here, using both C57BL/6 and BALB/c mice, we analyze immune cells in the pleural cavity. Unlike C57BL/6 mice, naive tissue-resident large-cavity macrophages (LCMs) of BALB/c mice failed to fully implement the tissue-residency program. Following infection with a pleural-dwelling nematode, these pre-existing differences were accentuated with LCM expansion occurring in C57BL/6, but not in BALB/c mice. While infection drove monocyte recruitment in both strains, only in C57BL/6 mice were monocytes able to efficiently integrate into the resident pool. Monocyte-to-macrophage conversion required both T cells and interleukin-4 receptor alpha (IL-4Rα) signaling. The transition to tissue residency altered macrophage function, and GATA6+ tissue-resident macrophages were required for host resistance to nematode infection. Therefore, during tissue nematode infection, T helper 2 (Th2) cells control the differentiation pathway of resident macrophages, which determines infection outcome.


Subject(s)
Filariasis , Filarioidea , Nematode Infections , Mice , Animals , Filarioidea/physiology , Th2 Cells , Monocytes , Pleural Cavity , Mice, Inbred C57BL , Macrophages/physiology , Cell Differentiation , Mice, Inbred BALB C
2.
Immunity ; 46(4): 660-674, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423340

ABSTRACT

Regulatory T cells (Treg cells) perform suppressive functions in disparate tissue environments and against many inflammatory insults, yet the tissue-enriched factor(s) that influence Treg cell phenotype and function remain largely unknown. We have shown a vital role for transforming growth factor-ß (TGF-ß) signals in safe-guarding specific Treg cell functions. TGF-ß signals were dispensable for steady-state Treg cell homeostasis and for Treg cell suppression of T cell proliferation and T helper-1 (Th1) cell differentiation. However, Treg cells require TGF-ß signals to appropriately dampen Th17 cells and regulate responses in the gastrointestinal tract. TGF-ß signaling maintains CD103 expression, promotes expression of the colon-specific trafficking molecule GPR15, and inhibits expression of GPR174, a receptor for lysophosphatidylserine, on Treg cells, collectively supporting the accumulation and retention of Treg cells in the colon and control of colitogenic responses. Thus, we reveal an unrecognized function for TGF-ß signaling as an upstream factor controlling Treg cell activity in specific tissue environments.


Subject(s)
Organ Specificity/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Homeostasis/immunology , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Receptors, Transforming Growth Factor beta/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Transforming Growth Factor beta/metabolism
3.
Immunity ; 46(1): 133-147, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28087239

ABSTRACT

Immuno-surveillance networks operating at barrier sites are tuned by local tissue cues to ensure effective immunity. Site-specific commensal bacteria provide key signals ensuring host defense in the skin and gut. However, how the oral microbiome and tissue-specific signals balance immunity and regulation at the gingiva, a key oral barrier, remains minimally explored. In contrast to the skin and gut, we demonstrate that gingiva-resident T helper 17 (Th17) cells developed via a commensal colonization-independent mechanism. Accumulation of Th17 cells at the gingiva was driven in response to the physiological barrier damage that occurs during mastication. Physiological mechanical damage, via induction of interleukin 6 (IL-6) from epithelial cells, tailored effector T cell function, promoting increases in gingival Th17 cell numbers. These data highlight that diverse tissue-specific mechanisms govern education of Th17 cell responses and demonstrate that mechanical damage helps define the immune tone of this important oral barrier.


Subject(s)
Gingiva/immunology , Immunity, Mucosal/immunology , Immunologic Surveillance/immunology , Mouth Mucosa/immunology , Th17 Cells/immunology , Animals , Flow Cytometry , Gingiva/microbiology , Humans , Mastication , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Mouth Mucosa/microbiology , Real-Time Polymerase Chain Reaction
4.
FASEB J ; 37(1): e22704, 2023 01.
Article in English | MEDLINE | ID: mdl-36520064

ABSTRACT

The gut microbiota is important for host health and immune system function. Moreover autoimmune diseases, such as rheumatoid arthritis, are associated with significant gut microbiota dysbiosis, although the causes and consequences of this are not fully understood. It has become clear that the composition and metabolic outputs of the microbiome exhibit robust 24 h oscillations, a result of daily variation in timing of food intake as well as rhythmic circadian clock function in the gut. Here, we report that experimental inflammatory arthritis leads to a re-organization of circadian rhythmicity in both the gut and associated microbiome. Mice with collagen induced arthritis exhibited extensive changes in rhythmic gene expression in the colon, and reduced barrier integrity. Re-modeling of the host gut circadian transcriptome was accompanied by significant alteration of the microbiota, including widespread loss of rhythmicity in symbiont species of Lactobacillus, and alteration in circulating microbial derived factors, such as tryptophan metabolites, which are associated with maintenance of barrier function and immune cell populations within the gut. These findings highlight that altered circadian rhythmicity during inflammatory disease contributes to dysregulation of gut integrity and microbiome function.


Subject(s)
Arthritis, Experimental , Gastrointestinal Microbiome , Microbiota , Mice , Animals , Gastrointestinal Microbiome/physiology , Dysbiosis/etiology , Arthritis, Experimental/complications , Collagen
5.
Immunity ; 43(2): 251-63, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26275994

ABSTRACT

Regulatory T (Treg) cells are essential for maintenance of immune homeostasis. Here we found that hydrogen sulfide (H2S) was required for Foxp3(+) Treg cell differentiation and function and that H2S deficiency led to systemic autoimmune disease. H2S maintained expression of methylcytosine dioxygenases Tet1 and Tet2 by sulfhydrating nuclear transcription factor Y subunit beta (NFYB) to facilitate its binding to Tet1 and Tet2 promoters. Transforming growth factor-ß (TGF-ß)-activated Smad3 and interleukin-2 (IL-2)-activated Stat5 facilitated Tet1 and Tet2 binding to Foxp3. Tet1 and Tet2 catalyzed conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in Foxp3 to establish a Treg-cell-specific hypomethylation pattern and stable Foxp3 expression. Consequently, Tet1 and Tet2 deletion led to Foxp3 hypermethylation, impaired Treg cell differentiation and function, and autoimmune disease. Thus, H2S promotes Tet1 and Tet2 expression, which are recruited to Foxp3 by TGF-ß and IL-2 signaling to maintain Foxp3 demethylation and Treg-cell-associated immune homeostasis.


Subject(s)
Colitis/immunology , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Hydrogen Sulfide/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CCAAT-Binding Factor/metabolism , Cell Differentiation/genetics , Colitis/genetics , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Forkhead Transcription Factors/genetics , Homeostasis/genetics , Homeostasis/immunology , Humans , Interleukin-2/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , STAT5 Transcription Factor/metabolism , Smad3 Protein/metabolism , T-Lymphocytes, Regulatory/transplantation , Transforming Growth Factor beta/immunology
6.
Immunity ; 42(6): 1130-42, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26070484

ABSTRACT

Tissue-infiltrating Ly6C(hi) monocytes play diverse roles in immunity, ranging from pathogen killing to immune regulation. How and where this diversity of function is imposed remains poorly understood. Here we show that during acute gastrointestinal infection, priming of monocytes for regulatory function preceded systemic inflammation and was initiated prior to bone marrow egress. Notably, natural killer (NK) cell-derived IFN-γ promoted a regulatory program in monocyte progenitors during development. Early bone marrow NK cell activation was controlled by systemic interleukin-12 (IL-12) produced by Batf3-dependent dendritic cells (DCs) in the mucosal-associated lymphoid tissue (MALT). This work challenges the paradigm that monocyte function is dominantly imposed by local signals after tissue recruitment, and instead proposes a sequential model of differentiation in which monocytes are pre-emptively educated during development in the bone marrow to promote their tissue-specific function.


Subject(s)
Bone Marrow Cells/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Antigens, Ly/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Bone Marrow Cells/parasitology , Cell Differentiation , Cells, Cultured , Interferon-gamma/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Intestinal Mucosa/parasitology , Killer Cells, Natural/parasitology , Leukocytes, Mononuclear/parasitology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Organ Specificity/immunology , Repressor Proteins/genetics , Repressor Proteins/metabolism
7.
Eur Respir J ; 61(5)2023 05.
Article in English | MEDLINE | ID: mdl-36922030

ABSTRACT

BACKGROUND: COVID-19 is associated with a dysregulated immune response but it is unclear how immune dysfunction contributes to the chronic morbidity persisting in many COVID-19 patients during convalescence (long COVID). METHODS: We assessed phenotypical and functional changes of monocytes in COVID-19 patients during hospitalisation and up to 9 months of convalescence following COVID-19, respiratory syncytial virus or influenza A. Patients with progressive fibrosing interstitial lung disease were included as a positive control for severe, ongoing lung injury. RESULTS: Monocyte alterations in acute COVID-19 patients included aberrant expression of leukocyte migration molecules, continuing into convalescence (n=142) and corresponding with specific symptoms of long COVID. Long COVID patients with unresolved lung injury, indicated by sustained shortness of breath and abnormal chest radiology, were defined by high monocyte expression of C-X-C motif chemokine receptor 6 (CXCR6) (p<0.0001) and adhesion molecule P-selectin glycoprotein ligand 1 (p<0.01), alongside preferential migration of monocytes towards the CXCR6 ligand C-X-C motif chemokine ligand 16 (CXCL16) (p<0.05), which is abundantly expressed in the lung. Monocyte CXCR6 and lung CXCL16 were heightened in patients with progressive fibrosing interstitial lung disease (p<0.001), confirming a role for the CXCR6-CXCL16 axis in ongoing lung injury. Conversely, monocytes from long COVID patients with ongoing fatigue exhibited a sustained reduction of the prostaglandin-generating enzyme cyclooxygenase 2 (p<0.01) and CXCR2 expression (p<0.05). These monocyte changes were not present in respiratory syncytial virus or influenza A convalescence. CONCLUSIONS: Our data define unique monocyte signatures that define subgroups of long COVID patients, indicating a key role for monocyte migration in COVID-19 pathophysiology. Targeting these pathways may provide novel therapeutic opportunities in COVID-19 patients with persistent morbidity.


Subject(s)
COVID-19 , Influenza, Human , Lung Injury , Humans , Monocytes/metabolism , Chemokines, CXC/metabolism , Receptors, Virus/metabolism , Receptors, CXCR6 , Receptors, Chemokine/metabolism , Post-Acute COVID-19 Syndrome , Ligands , Convalescence , Receptors, Scavenger/metabolism , Chemokine CXCL16 , Patient Acuity
8.
Nat Immunol ; 12(4): 312-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21297643

ABSTRACT

The molecular mechanisms that direct the development of TCRαß+CD8αα+ intestinal intraepithelial lymphocytes (IELs) are not thoroughly understood. Here we show that transforming growth factor-ß (TGF-ß) controls the development of TCRαß+CD8αα+ IELs. Mice with either a null mutation in the gene encoding TGF-ß1 or T cell-specific deletion of TGF-ß receptor I lacked TCRαß+CD8αα+ IELs, whereas mice with transgenic overexpression of TGF-ß1 had a larger population of TCRαß+CD8αα+ IELs. We observed defective development of the TCRαß+CD8αα+ IEL thymic precursors (CD4⁻CD8⁻TCRαß+CD5+) in the absence of TGF-ß. In addition, we found that TGF-ß signaling induced CD8α expression in TCRαß+CD8αα+ IEL thymic precursors and induced and maintained CD8α expression in peripheral populations of T cells. Our data demonstrate a previously unrecognized role for TGF-ß in the development of TCRαß+CD8αα+ IELs and the expression of CD8α in T cells.


Subject(s)
CD8 Antigens/metabolism , Lymphocytes/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Transforming Growth Factor beta1/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Flow Cytometry , Gene Expression/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Lymphocyte Count , Lymphocytes/cytology , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology
9.
Nat Immunol ; 12(1): 86-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21131965

ABSTRACT

The molecular mechanisms that direct transcription of the gene encoding the transcription factor Foxp3 in CD4(+) T cells remain ill-defined. We show here that deletion of the DNA-binding inhibitor Id3 resulted in the defective generation of Foxp3(+) regulatory T cells (T(reg) cells). We identify two transforming growth factor-ß1 (TGF-ß1)-dependent mechanisms that were vital for activation of Foxp3 transcription and were defective in Id3(-/-) CD4(+) T cells. Enhanced binding of the transcription factor E2A to the Foxp3 promoter promoted Foxp3 transcription. Id3 was required for relief of inhibition by the transcription factor GATA-3 at the Foxp3 promoter. Furthermore, Id3(-/-) T cells showed greater differentiation into the T(H)17 subset of helper T cells in vitro and in a mouse asthma model. Therefore, a network of factors acts in a TGF-ß-dependent manner to control Foxp3 expression and inhibit the development of T(H)17 cells.


Subject(s)
Asthma/metabolism , Forkhead Transcription Factors/metabolism , Inhibitor of Differentiation Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/genetics , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Sequence Deletion/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Transcriptional Activation/genetics , Transforming Growth Factor beta1/metabolism
10.
Trends Immunol ; 39(4): 276-287, 2018 04.
Article in English | MEDLINE | ID: mdl-28923364

ABSTRACT

The oral mucosal barrier is constantly exposed to a plethora of triggers requiring immune control, including a diverse commensal microbiome, ongoing damage from mastication, and dietary and airborne antigens. However, how these tissue-specific cues participate in the training of immune responsiveness at this site is minimally understood. Moreover, the mechanisms mediating homeostatic immunity at this interface are not yet fully defined. Here we present basic aspects of the oral mucosal barrier and discuss local cues that may modulate and train local immune responsiveness. We particularly focus on the immune cell network mediating immune surveillance at a specific oral barrier, the gingiva - a constantly stimulated and dynamic environment where homeostasis is often disrupted, resulting in the common inflammatory disease periodontitis.


Subject(s)
Immunity, Mucosal , Microbiota/immunology , Mouth/immunology , Periodontitis/immunology , Animals , Host-Parasite Interactions , Humans , Intestinal Mucosa , Organ Specificity
11.
Proc Natl Acad Sci U S A ; 115(42): 10738-10743, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30279177

ABSTRACT

γδ T cells are enriched at barrier sites such as the gut, skin, and lung, where their roles in maintaining barrier integrity are well established. However, how these cells contribute to homeostasis at the gingiva, a key oral barrier and site of the common chronic inflammatory disease periodontitis, has not been explored. Here we demonstrate that the gingiva is policed by γδ T cells with a T cell receptor (TCR) repertoire that diversifies during development. Gingival γδ T cells accumulated rapidly after birth in response to barrier damage, and strikingly, their absence resulted in enhanced pathology in murine models of the oral inflammatory disease periodontitis. Alterations in bacterial communities could not account for the increased disease severity seen in γδ T cell-deficient mice. Instead, gingival γδ T cells produced the wound healing associated cytokine amphiregulin, administration of which rescued the elevated oral pathology of tcrδ-/- mice. Collectively, our results identify γδ T cells as critical constituents of the immuno-surveillance network that safeguard gingival tissue homeostasis.


Subject(s)
Amphiregulin/metabolism , Homeostasis , Mouth/immunology , Periodontitis/immunology , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocyte Subsets/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouth/metabolism , Periodontitis/metabolism , Periodontitis/pathology , T-Lymphocyte Subsets/metabolism
12.
Pflugers Arch ; 469(3-4): 527-539, 2017 04.
Article in English | MEDLINE | ID: mdl-28283748

ABSTRACT

Monocyte-derived mononuclear phagocytes, particularly macrophages, are crucial to maintain gastrointestinal homeostasis in the steady state but are also important for protection against certain pathogens. However, when uncontrolled, they can promote immunopathology. Broadly two subsets of macrophages can be considered to perform the vast array of functions to complete these complex tasks: resident macrophages that dominate in the healthy gut and inflammation-elicited (inflammatory) macrophages that derive from circulating monocytes infiltrating inflamed tissue. Here, we discuss the features of resident and inflammatory intestinal macrophages, complexities in identifying and defining these populations and the mechanisms involved in their differentiation. In particular, focus will be placed on describing their unique ontogeny as well as local gastrointestinal signals that instruct specialisation of resident macrophages in healthy tissue. We then explore the very different roles of inflammatory macrophages and describe new data suggesting that they may be educated not only by the gut microenvironment but also by signals they receive during development in the bone marrow. Given the high degree of plasticity of gut macrophages and their multifaceted roles in both healthy and inflamed tissue, understanding the mechanisms controlling their differentiation could inform development of improved therapies for inflammatory diseases such as inflammatory bowel disease (IBD).


Subject(s)
Homeostasis/physiology , Inflammation/pathology , Intestines/pathology , Intestines/physiology , Macrophages/physiology , Animals , Cell Differentiation/physiology , Humans , Inflammatory Bowel Diseases/pathology
13.
Proc Natl Acad Sci U S A ; 111(4): E465-73, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474796

ABSTRACT

Maintenance of immune tolerance critically depends upon regulatory T cells that express the transcription factor forkhead box P3 (Foxp3). These CD4(+) T cells can be generated in the thymus, termed thymus-derived regulatory T cells (tTregs), but their developmental pathway remains incompletely understood. tTreg development has been shown to be delayed compared with that of CD4(+) single positive (SP) thymocytes, with tTregs being detected only in neonatal thymi by day 3 after birth. Here, we outline the reasons for this delayed emergence of Foxp3(+) tTregs and demonstrate that thymocyte apoptosis is intrinsically tied to tTreg development. We show that thymic apoptosis leads to the production of TGFß intrathymically from thymic macrophages, dendritic cells, and epithelial cells. This TGFß then induces foxp3 expression and drives tTreg generation. Thymocyte apoptosis has previously been shown to accelerate after birth, which drives increases in TGFß in the neonatal thymus. We highlight a paucity of TGFß in the neonatal thymus, accounting for the delayed development of tTregs compared with CD4(+) SP thymocytes. Importantly, we show that enhanced levels of apoptosis in the thymus result in an augmented tTreg population and, moreover, that decreasing thymic apoptosis results in reduced tTregs. In addition to this, we also show that T-cell receptor (TCR) signals of different affinity were all capable of driving tTreg development; however, to achieve this TGFß signals must also be received concomitant with the TCR signal. Collectively, our results indicate that thymic apoptosis is a key event in tTreg generation and reveal a previously unrecognized apoptosis-TGFß-Foxp3 axis that mediates the development of tTregs.


Subject(s)
Apoptosis , T-Lymphocytes, Regulatory/immunology , Thymocytes/cytology , Transforming Growth Factor beta/physiology , Animals , Animals, Newborn , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Thymocytes/immunology
14.
Eur J Immunol ; 45(4): 958-65, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25684698

ABSTRACT

CD4(+) regulatory T cells expressing the transcription factor Foxp3 can be generated in the thymus (tTreg cells), but the cellular and molecular pathways driving their development remain incompletely understood. TGF-ß is essential for the generation of Foxp3(+) Treg cells converted from peripheral naïve CD4(+) T cells (pTreg cells), yet a role for TGF-ß in tTreg-cell development was initially refuted. Nevertheless, recent studies have unmasked a requirement for TGF-ß in the generation of tTreg cells. Experimental evidence reveals that TGF-ß in the context of TCR stimulation induces Foxp3 gene transcription in thymic Treg precursors, CD4(+) CD8(-) CD25(-) semimature and mature single-positive thymocytes. Intriguingly, thymic apoptosis was found to be intrinsically linked to the generation of tTreg cells, as apoptosis induced expression of TGF-ß intrathymically. In this short review, we will highlight key data, discuss the experimental evidence and propose a modified model of tTreg-cell development involving TGF-ß. We will also outline the remaining unresolved questions concerning generation of thymic Foxp3(+) Treg cells and provide our personal perspectives on the mechanisms controlling tTreg-cell development.


Subject(s)
Apoptosis/immunology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Transforming Growth Factor beta/immunology , Cell Differentiation/immunology , Forkhead Transcription Factors/genetics , Humans , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/cytology , Thymocytes/immunology
15.
Nature ; 467(7318): 967-71, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20962846

ABSTRACT

CD4(+) T-helper cells that selectively produce interleukin (IL)-17 (T(H)17), are critical for host defence and autoimmunity. Although crucial for T(H)17 cells in vivo, IL-23 has been thought to be incapable of driving initial differentiation. Rather, IL-6 and transforming growth factor (TGF)-ß1 have been proposed to be the factors responsible for initiating specification. Here we show that T(H)17 differentiation can occur in the absence of TGF-ß signalling. Neither IL-6 nor IL-23 alone efficiently generated T(H)17 cells; however, these cytokines in combination with IL-1ß effectively induced IL-17 production in naive precursors, independently of TGF-ß. Epigenetic modification of the Il17a, Il17f and Rorc promoters proceeded without TGF-ß1, allowing the generation of cells that co-expressed RORγt (encoded by Rorc) and T-bet. T-bet(+)RORγt(+) T(H)17 cells are generated in vivo during experimental allergic encephalomyelitis, and adoptively transferred T(H)17 cells generated with IL-23 without TGF-ß1 were pathogenic in this disease model. These data indicate an alternative mode for T(H)17 differentiation. Consistent with genetic data linking IL23R with autoimmunity, our findings re-emphasize the importance of IL-23 and therefore may have therapeutic implications.


Subject(s)
Signal Transduction , Th17 Cells/pathology , Transforming Growth Factor beta , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmunity/immunology , Cell Differentiation/drug effects , Central Nervous System/pathology , Inflammation , Interleukin-10 , Interleukin-17/metabolism , Interleukin-1beta/immunology , Interleukin-23/immunology , Interleukin-23/pharmacology , Interleukin-6/immunology , Interleukin-9 , Interleukins/biosynthesis , Mice , Mice, Inbred C57BL , Mucous Membrane/cytology , Mucous Membrane/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Interleukin/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism , Interleukin-22
16.
Blood ; 122(13): 2224-32, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23940283

ABSTRACT

Transforming growth factor-ß (TGF-ß) receptors (TßRs) are essential components for TGF-ß signal transduction in T cells, yet the mechanisms by which the receptors are regulated remain poorly understood. We show here that Poly(ADP-ribose) polymerase-1 (PARP-1) regulates TGF-ß receptor I (TßRI) and II (TßRII) expression in CD4(+) T cells and subsequently affects Smad2/3-mediated TGF-ß signal transduction. Inhibition of PARP-1 led to the upregulation of both TßRI and TßRII, yet the underlying molecular mechanisms were distinct. PARP-1 selectively bound to the promoter of TßRII, whereas the enzymatic activity of PARP-1 was responsible for the inhibition of TßRI expression. Importantly, inhibition of PARP-1 also enhanced expression of TßRs in human CD4(+) T cells. Thus, PARP-1 regulates TßR expression and TGF-ß signaling in T cells.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Receptors, Transforming Growth Factor beta/biosynthesis , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/immunology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
17.
Semin Immunol ; 23(6): 418-23, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21752667

ABSTRACT

Induction of Foxp3 gene expression and acquisition of regulatory T cell fate is, understandably, a highly controlled process and one which many investigators want to illuminate. In studying the regulation of Foxp3 gene expression, several conserved non-coding regions have been identified and the role of various transcription factors at these sites has been explored. What emerges is that many factors, some positive, some negative, interact to collectively drive Foxp3 gene expression and then maintain its expression in Foxp3(+) regulatory T cells. TCR signaling is imperative for Foxp3 gene expression and TGF-ß is a key cytokine for initiating Foxp3 gene expression in naïve T cells. But other signaling pathways are also known to play a role in properly orchestrating Foxp3 gene expression and regulatory T cell expansion. Here we review the recent progress in understanding the complex molecular events that drive Foxp3 gene expression and allow functional regulatory T cells to develop.


Subject(s)
Forkhead Transcription Factors/immunology , Gene Expression Regulation , T-Lymphocytes, Regulatory/immunology , Animals , Cell Lineage , Forkhead Transcription Factors/genetics , Humans , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
18.
J Biol Chem ; 288(44): 32074-92, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24056369

ABSTRACT

Three homologues of TGF-ß exist in mammals as follows: TGF-ß1, TGF-ß2, and TGF-ß3. All three proteins share high homology in their amino acid sequence, yet each TGF-ß isoform has unique heterologous motifs that are highly conserved during evolution. Although these TGF-ß proteins share similar properties in vitro, isoform-specific properties have been suggested through in vivo studies and by the unique phenotypes for each TGF-ß knock-out mouse. To test our hypothesis that each of these homologues has nonredundant functions, and to identify such isoform-specific roles, we genetically exchanged the coding sequence of the mature TGF-ß1 ligand with a sequence from TGF-ß3 using targeted recombination to create chimeric TGF-ß1/3 knock-in mice (TGF-ß1(Lß3/Lß3)). In the TGF-ß1(Lß3/Lß3) mouse, localization and activation still occur through the TGF-ß1 latent associated peptide, but cell signaling is triggered through the TGF-ß3 ligand that binds to TGF-ß receptors. Unlike TGF-ß1(-/-) mice, the TGF-ß1(Lß3/Lß3) mice show neither embryonic lethality nor signs of multifocal inflammation, demonstrating that knock-in of the TGF-ß3 ligand can prevent the vasculogenesis defects and autoimmunity associated with TGF-ß1 deficiency. However, the TGF-ß1(Lß3/Lß3) mice have a shortened life span and display tooth and bone defects, indicating that the TGF-ß homologues are not completely interchangeable. Remarkably, the TGF-ß1(Lß3/Lß3) mice display an improved metabolic phenotype with reduced body weight gain and enhanced glucose tolerance by induction of beneficial changes to the white adipose tissue compartment. These findings reveal both redundant and unique nonoverlapping functional diversity in TGF-ß isoform signaling that has relevance to the design of therapeutics aimed at targeting the TGF-ß pathway in human disease.


Subject(s)
Glucose/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/metabolism , Animals , COS Cells , Chlorocebus aethiops , Gene Knock-In Techniques , Glucose/genetics , Hep G2 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neovascularization, Physiologic/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Swine , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta3/genetics
19.
Mucosal Immunol ; 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39038755

ABSTRACT

Immune cells residing at the gingiva experience diverse and unique signals, tailoring their functions to enable them to appropriately respond to immunological challenges and maintain tissue integrity. The gingiva, defined as the mucosal barrier that surrounds and supports the teeth, is the only barrier site completely transected by a hard structure, the tooth. The tissue is damaged in early life during tooth eruption and chronically throughout life by the process of mastication. This occurs alongside challenges typical of barrier sites, including exposure to invading pathogens, the local commensal microbial community and environmental antigens. This review will focus on the immune network safeguarding gingival integrity, which is far less understood than that resident at other barrier sites. A detailed understanding of the gingiva-resident immune network is vital as it is the site of the inflammatory disease periodontitis, the most common chronic inflammatory condition in humans which has well-known detrimental systemic effects. Furthering our understanding of how the immune populations within the gingiva develop, are tailored in health, and how this is dysregulated in disease would further the development of effective therapies for periodontitis.

20.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38819409

ABSTRACT

Th17 cell plasticity is crucial for development of autoinflammatory disease pathology. Periodontitis is a prevalent inflammatory disease where Th17 cells mediate key pathological roles, yet whether they exhibit any functional plasticity remains unexplored. We found that during periodontitis, gingival IL-17 fate-mapped T cells still predominantly produce IL-17A, with little diversification of cytokine production. However, plasticity of IL-17 fate-mapped cells did occur during periodontitis, but in the gingiva draining lymph node. Here, some Th17 cells acquired features of Tfh cells, a functional plasticity that was dependent on IL-6. Notably, Th17-to-Tfh diversification was important to limit periodontitis pathology. Preventing Th17-to-Tfh plasticity resulted in elevated periodontal bone loss that was not simply due to increased proportions of conventional Th17 cells. Instead, loss of Th17-to-Tfh cells resulted in reduced IgG levels within the oral cavity and a failure to restrict the biomass of the oral commensal community. Thus, our data identify a novel protective function for a subset of otherwise pathogenic Th17 cells during periodontitis.


Subject(s)
Cell Plasticity , Interleukin-17 , Periodontitis , Th17 Cells , Th17 Cells/immunology , Animals , Periodontitis/immunology , Periodontitis/pathology , Cell Plasticity/immunology , Interleukin-17/metabolism , Interleukin-17/immunology , Mice , Interleukin-6/metabolism , Mice, Inbred C57BL , T Follicular Helper Cells/immunology , Gingiva/immunology , Gingiva/pathology , Immunoglobulin G/immunology , Alveolar Bone Loss/immunology , Alveolar Bone Loss/pathology
SELECTION OF CITATIONS
SEARCH DETAIL