Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Cell ; 184(8): 2151-2166.e16, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33765440

ABSTRACT

Cutaneous mast cells mediate numerous skin inflammatory processes and have anatomical and functional associations with sensory afferent neurons. We reveal that epidermal nerve endings from a subset of sensory nonpeptidergic neurons expressing MrgprD are reduced by the absence of Langerhans cells. Loss of epidermal innervation or ablation of MrgprD-expressing neurons increased expression of a mast cell gene module, including the activating receptor, Mrgprb2, resulting in increased mast cell degranulation and cutaneous inflammation in multiple disease models. Agonism of MrgprD-expressing neurons reduced expression of module genes and suppressed mast cell responses. MrgprD-expressing neurons released glutamate which was increased by MrgprD agonism. Inhibiting glutamate release or glutamate receptor binding yielded hyperresponsive mast cells with a genomic state similar to that in mice lacking MrgprD-expressing neurons. These data demonstrate that MrgprD-expressing neurons suppress mast cell hyperresponsiveness and skin inflammation via glutamate release, thereby revealing an unexpected neuroimmune mechanism maintaining cutaneous immune homeostasis.


Subject(s)
Glutamic Acid/metabolism , Mast Cells/metabolism , Neurons/metabolism , Skin/metabolism , Animals , Cells, Cultured , Dermatitis/metabolism , Dermatitis/pathology , Diphtheria Toxin/pharmacology , Disease Models, Animal , Female , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Langerhans Cells/cytology , Langerhans Cells/drug effects , Langerhans Cells/metabolism , Mast Cells/cytology , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Skin/pathology , beta-Alanine/chemistry , beta-Alanine/metabolism , beta-Alanine/pharmacology
2.
J Allergy Clin Immunol ; 151(5): 1169-1177, 2023 05.
Article in English | MEDLINE | ID: mdl-37149370

ABSTRACT

The skin is a barrier organ populated by many types of skin-resident immune cells and sensory neurons. It has become increasingly appreciated that neuroimmune interactions are an important component of inflammatory diseases such as atopic dermatitis and allergic contact dermatitis. Neuropeptides secreted from nerve terminals play an important role in mediating cutaneous immune cell function, and soluble mediators derived from immune cells interact with neurons to induce itch. In this review article, we will explore emerging research describing neuronal effector functions on skin immune cells in mouse models of atopic and contact dermatitis. We will also discuss the contributions of both specific neuronal subsets and secreted immune factors to itch induction and the associated inflammatory processes. Finally, we will explore how treatment strategies have emerged around these findings and discuss the relationship between scratching and dermatitis.


Subject(s)
Dermatitis, Allergic Contact , Dermatitis, Atopic , Mice , Animals , Neuroimmunomodulation , Pruritus , Skin , Sensory Receptor Cells
3.
Immunity ; 41(5): 677-9, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25517608

ABSTRACT

Interactions between potentially pathogenic commensal bacteria and cutaneous immunity are poorly understood. In this issue of Immunity, Skabytska et al. (2014) show that S. aureus-derived TLR2/6 heterodimer ligands can recruit myeloid-derived suppressor cells into the skin, countering rather than promoting inflammation.


Subject(s)
Myeloid Cells/immunology , Skin/immunology , Staphylococcal Skin Infections/immunology , Toll-Like Receptor 2/immunology , Animals , Humans
4.
J Allergy Clin Immunol ; 150(1): 114-130, 2022 07.
Article in English | MEDLINE | ID: mdl-35085664

ABSTRACT

BACKGROUND: Allergic contact dermatitis (CD) is a chronic inflammatory skin disease caused by type 1 biased adaptive immunity for which there is an unmet need for antigen (Ag)-specific immunotherapies. Exposure to skin sensitizers stimulates secretion of the proinflammatory neuropeptides substance P and hemokinin 1, which signal via the neurokinin-1 receptor (NK1R) to promote the innate and adaptive immune responses of CD. Accordingly, mice lacking the NK1R develop impaired CD. Nonetheless, the role and therapeutic opportunities of targeting the NK1R in CD remain to be elucidated. OBJECTIVE: We sought to develop an Ag-specific immunosuppressive approach to treat CD by skin codelivery of hapten and NK1R antagonists integrated in dissolvable microneedle arrays (MNA). METHODS: In vivo mouse models of contact hypersensitivity and ex vivo models of human skin were used to delineate the effects and mechanisms of NK1R signaling and the immunosuppressive effects of the contact sensitizer NK1R antagonist MNA in CD. RESULTS: We demonstrated in mice that CD requires NK1R signaling by substance P and hemokinin 1. Specific deletion of the NK1R in keratinocytes and dendritic cells, but not in mast cells, prevented CD. Skin codelivery of hapten or Ag MNA inhibited neuropeptide-mediated skin inflammation in mouse and human skin, promoted deletion of Ag-specific effector T cells, and increased regulatory T cells, which prevented CD onset and relapses locally and systemically in an Ag-specific manner. CONCLUSIONS: Immunoregulation by engineering localized skin neuroimmune networks can be used to treat cutaneous diseases that like CD are caused by type 1 immunity.


Subject(s)
Dermatitis, Allergic Contact , Neurokinin-1 Receptor Antagonists , Animals , Dermatitis, Allergic Contact/drug therapy , Haptens , Mice , Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-1 , Substance P
5.
J Immunol ; 204(9): 2337-2348, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32213560

ABSTRACT

The signaling protein MALT1 plays a key role in promoting NF-κB activation in Ag-stimulated lymphocytes. In this capacity, MALT1 has two functions, acting as a scaffolding protein and as a substrate-specific protease. MALT1 is also required for NF-κB-dependent induction of proinflammatory cytokines after FcεR1 stimulation in mast cells, implicating a role in allergy. Because MALT1 remains understudied in this context, we sought to investigate how MALT1 proteolytic activity contributes to the overall allergic response. We compared bone marrow-derived mast cells from MALT1 knockout (MALT1-/-) and MALT1 protease-deficient (MALTPD/PD) mice to wild-type cells. We found that MALT1-/- and MALT1PD/PD mast cells are equally impaired in cytokine production following FcεRI stimulation, indicating that MALT1 scaffolding activity is insufficient to drive the cytokine response and that MALT1 protease activity is essential. In addition to cytokine production, acute mast cell degranulation is a critical component of allergic response. Intriguingly, whereas degranulation is MALT1-independent, MALT1PD/PD mice are protected from vascular edema induced by either passive cutaneous anaphylaxis or direct challenge with histamine, a major granule component. This suggests a role for MALT1 protease activity in endothelial cells targeted by mast cell-derived vasoactive substances. Indeed, we find that in human endothelial cells, MALT1 protease is activated following histamine treatment and is required for histamine-induced permeability. We thus propose a dual role for MALT1 protease in allergic response, mediating 1) IgE-dependent mast cell cytokine production, and 2) histamine-induced endothelial permeability. This dual role indicates that therapeutic inhibitors of MALT1 protease could work synergistically to control IgE-mediated allergic disease.


Subject(s)
Endothelial Cells/metabolism , Hypersensitivity/metabolism , Mast Cells/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Line , Cytokines/immunology , Cytokines/metabolism , Endothelial Cells/immunology , Female , Histamine/immunology , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Inflammation/immunology , Inflammation/metabolism , Lymphocyte Activation/immunology , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, IgE/immunology , Receptors, IgE/metabolism
6.
J Allergy Clin Immunol ; 135(4): 1019-1030.e8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25201259

ABSTRACT

BACKGROUND: Efficient development of atopic diseases requires interactions between allergen and adjuvant to initiate and amplify the underlying inflammatory responses. Substance P (SP) and hemokinin-1 (HK-1) are neuropeptides that signal through the neurokinin-1 receptor (NK1R) to promote inflammation. Mast cells initiate the symptoms and tissue effects of atopic disorders, secreting TNF and IL-6 after FcεRI cross-linking by antigen-IgE complexes (FcεRI-activated mast cells [FcεRI-MCs]). Additionally, MCs express the NK1R, suggesting an adjuvant role for NK1R agonists in FcεRI-MC-mediated pathologies; however, in-depth research addressing this relevant aspect of MC biology is lacking. OBJECTIVE: We sought to investigate the effect of NK1R signaling and the individual roles of SP and HK-1 as potential adjuvants for FcεRI-MC-mediated allergic disorders. METHODS: Bone marrow-derived mast cells (BMMCs) from C57BL/6 wild-type (WT) or NK1R(-/-) mice were used to investigate the effects of NK1R signaling on FcεRI-MCs. BMMCs generated from Tac1(-/-) mice or after culture with Tac4 small interfering RNA were used to address the adjuvancy of SP and HK-1. WT, NK1R(-/-), and c-Kit(W-sh/W-sh) mice reconstituted with WT or NK1R(-/-) BMMCs were used to evaluate NK1R signaling on FcεRI-MC-mediated passive local and systemic anaphylaxis and on airway inflammation. RESULTS: FcεRI-activated MCs upregulated NK1R and HK-1 transcripts and protein synthesis, without modifying SP expression. In a positive signaling loop HK-1 promoted TNF and IL-6 secretion by MC degranulation and protein synthesis, the latter through the phosphoinositide 3-kinase/Akt/nuclear factor κB pathways. In vivo NK1R signaling was necessary for the development of passive local and systemic anaphylaxis and airway inflammation. CONCLUSIONS: FcεRI stimulation of MCs promotes autocrine secretion of HK-1, which signals through NK1R to provide adjuvancy for efficient development of FcεRI-MC-mediated disorders.


Subject(s)
Autocrine Communication , Immunoglobulin E/immunology , Inflammation/immunology , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Tachykinins/metabolism , Anaphylaxis/immunology , Anaphylaxis/metabolism , Animals , Disease Models, Animal , Female , Interleukin-6/biosynthesis , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Receptors, IgE/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction , Tumor Necrosis Factors/biosynthesis
7.
Blood ; 121(15): 2923-33, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23365459

ABSTRACT

Substance-P and hemokinin-1 are proinflammatory neuropeptides with potential to promote type 1 immunity through agonistic binding to neurokinin-1 receptor (NK1R). Dendritic cells (DCs) are professional antigen-presenting cells that initiate and regulate the outcome of innate and adaptive immune responses. Immunostimulatory DCs are highly desired for the development of positive immunization techniques. DCs express functional NK1R; however, regardless of their potential DC-stimulatory function, the ability of NK1R agonists to promote immunostimulatory DCs remains unexplored. Here, we demonstrate that NK1R signaling activates therapeutic DCs capable of biasing type 1 immunity by inhibition of interleukin-10 (IL-10) synthesis and secretion, without affecting their low levels of IL-12 production. The potent type 1 effector immune response observed following cutaneous administration of NK1R-signaled DCs required their homing in skin-draining lymph nodes (sDLNs) where they induced inflammation and licensed endogenous-conventional sDLN-resident and -recruited inflammatory DCs to secrete IL-12. Our data demonstrate that NK1R signaling promotes immunostimulatory DCs, and provide relevant insight into the mechanisms used by neuromediators to regulate innate and adaptive immune responses.


Subject(s)
Dendritic Cells/immunology , Immunity, Cellular/immunology , Interleukin-12/immunology , Receptors, Neurokinin-1/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Flow Cytometry , Immunization/methods , Immunophenotyping , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Receptors, Neurokinin-1/agonists , Receptors, Neurokinin-1/metabolism , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
8.
J Immunol ; 188(11): 5227-37, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22508931

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are highly specialized APCs that, in addition to their well-recognized role in anti-viral immunity, also regulate immune responses. Liver-resident pDCs are considerably less immunostimulatory than those from secondary lymphoid tissues and are equipped to promote immune tolerance/regulation through various mechanisms. IL-27 is an IL-12 family cytokine that regulates the function of both APCs and T cells, although little is known about its role in pDC immunobiology. In this study, we show that mouse liver pDCs express higher levels of IL-27p28 and EBV-induced protein 3 (Ebi3) compared with those of splenic pDCs. Both populations of pDCs express the IL-27Rα/WSX-1; however, only liver pDCs significantly upregulate expression of the coregulatory molecule B7 homolog-1 (B7-H1) in response to IL-27. Inhibition of STAT3 activation completely abrogates IL-27-induced upregulation of B7-H1 expression on liver pDCs. Liver pDCs treated with IL-27 increase the percentage of CD4(+)Foxp3(+) T cells in MLR, which is dependent upon expression of B7-H1. pDCs from Ebi3-deficient mice lacking functional IL-27 show increased capacity to stimulate allogeneic T cell proliferation and IFN-γ production in MLR. Liver but not spleen pDCs suppress delayed-type hypersensitivity responses to OVA, an effect that is lost with Ebi3(-/-) and B7-H1(-/-) liver pDCs compared with wild-type liver pDCs. These data suggest that IL-27 signaling in pDCs promotes their immunoregulatory function and that IL-27 produced by pDCs contributes to their capacity to regulate immune responses in vitro and in vivo.


Subject(s)
B7-H1 Antigen/biosynthesis , Dendritic Cells/immunology , Interleukins/biosynthesis , Liver/immunology , STAT3 Transcription Factor/physiology , Up-Regulation/immunology , Animals , B7-H1 Antigen/deficiency , Dendritic Cells/metabolism , Down-Regulation/genetics , Down-Regulation/immunology , Humans , Hypersensitivity, Delayed/genetics , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Liver/cytology , Liver/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Ovalbumin/physiology , Receptors, Cytokine/biosynthesis , Up-Regulation/genetics
9.
J Immunol ; 188(8): 3667-77, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22427640

ABSTRACT

Hepatic stellate cells (HSCs) may play an important role in hepatic immune regulation by producing numerous cytokines/chemokines and expressing Ag-presenting and T cell coregulatory molecules. Due to disruption of the endothelial barrier during cold-ischemic storage and reperfusion of liver grafts, HSCs can interact directly with cells of the immune system. Endotoxin (LPS), levels of which increase in liver diseases and transplantation, stimulates the synthesis of many mediators by HSCs. We hypothesized that LPS-stimulated HSCs might promote hepatic tolerogenicity by influencing naturally occurring immunosuppressive CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Following their portal venous infusion, allogeneic CD4(+) T cells, including Tregs, were found closely associated with HSCs, and this association increased in LPS-treated livers. In vitro, both unstimulated and LPS-stimulated HSCs upregulated Fas (CD95) expression on conventional CD4(+) T cells and induced their apoptosis in a Fas/Fas ligand-dependent manner. By contrast, HSCs induced Treg proliferation, which required cell-cell contact and was MHC class II-dependent. This effect was augmented when HSCs were pretreated with LPS. LPS increased the expression of MHC class II, CD80, and CD86 and stimulated the production of IL-1α, IL-1ß, IL-6, IL-10 and TNF-α by HSCs. Interestingly, production of IL-1α, IL-1ß, IL-6, and TNF-α was strongly inhibited, but that of IL-10 enhanced in LPS-pretreated HSC/Treg cocultures. Adoptively transferred allogeneic HSCs migrated to the secondary lymphoid tissues and induced Treg expansion in lymph nodes. These data implicate endotoxin-stimulated HSCs as important immune regulators in liver transplantation by inducing selective expansion of tolerance-promoting Tregs and reducing inflammation and alloimmunity.


Subject(s)
Cell Communication/immunology , Endotoxins/pharmacology , Gene Expression Regulation/immunology , Liver Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Adoptive Transfer , Animals , Antigens, CD/biosynthesis , Antigens, CD/immunology , Apoptosis , Cell Communication/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Genes, MHC Class II , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , Transcription Factors/biosynthesis , Transcription Factors/immunology , Transplantation, Homologous
10.
J Immunol ; 189(8): 3848-58, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22962681

ABSTRACT

Hepatic stellate cells (HSCs) are critical for hepatic wound repair and tissue remodeling. They also produce cytokines and chemokines that may contribute to the maintenance of hepatic immune homeostasis and the inherent tolerogenicity of the liver. The functional relationship between HSCs and the professional migratory APCs in the liver, that is, dendritic cells (DCs), has not been evaluated. In this article, we report that murine liver DCs colocalize with HSCs in vivo under normal, steady-state conditions, and cluster with HSCs in vitro. In vitro, HSCs secrete high levels of DC chemoattractants, such as MΙP-1α and MCP-1, as well as cytokines that modulate DC activation, including TNF-α, IL-6, and IL-1ß. Culture of HSCs with conventional liver myeloid (m) DCs resulted in increased IL-6 and IL-10 secretion compared with that of either cell population alone. Coculture also resulted in enhanced expression of costimulatory (CD80, CD86) and coinhibitory (B7-H1) molecules on mDCs. HSC-induced mDC maturation required cell-cell contact and could be blocked, in part, by neutralizing MΙP-1α or MCP-1. HSC-induced mDC maturation was dependent on activation of STAT3 in mDCs and, in part, on HSC-secreted IL-6. Despite upregulation of costimulatory molecules, mDCs conditioned by HSCs demonstrated impaired ability to induce allogeneic T cell proliferation, which was independent of B7-H1, but dependent upon HSC-induced STAT3 activation and subsequent upregulation of IDO. In conclusion, by promoting IDO expression, HSCs may act as potent regulators of liver mDCs and function to maintain hepatic homeostasis and tolerogenicity.


Subject(s)
Dendritic Cells/immunology , Down-Regulation/immunology , Hepatic Stellate Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Liver/immunology , Myeloid Cells/immunology , STAT3 Transcription Factor/physiology , Animals , Cells, Cultured , Coculture Techniques , Enzyme Induction/genetics , Enzyme Induction/immunology , Hepatic Stellate Cells/enzymology , Hepatic Stellate Cells/metabolism , Immunophenotyping , Isoantigens/genetics , Isoantigens/physiology , Liver/cytology , Liver/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
11.
Melanoma Res ; 34(1): 1-8, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37924526

ABSTRACT

Mast cells (MCs) accumulate in a broad range of tumors, including melanomas. While MCs are potent initiators of immunity in infection, and in allergic inflammation, the function of MCs in anti-melanoma immunity is unclear. MCs have the potential to release tumoricidal cytokines and proteases, to activate antigen-presenting cells and to promote anti-tumor adaptive immunity. However, within the immunosuppressive tumor microenvironment (TME), MC activation may promote angiogenesis and contribute to tumor growth. In this review, the relationship between MCs and melanomas is discussed with a focus on the impact of the TME on MC activation.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Mast Cells/pathology , Mast Cells/physiology , Melanoma/pathology , Skin Neoplasms/pathology , Cytokines , Inflammation , Tumor Microenvironment
12.
J Immunol ; 186(4): 1970-80, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21257958

ABSTRACT

Freshly isolated hepatic dendritic cells (DC) are comparatively immature, relatively resistant to maturation, and can downmodulate effector T cell responses. Molecular mechanisms that underlie these properties are ill defined. DNAX-activating protein of 12 kDa (DAP12) is an ITAM-bearing transmembrane adaptor protein that integrates signals through several receptors, including triggering receptor expressed on myeloid cells-1, -2, and CD200R. Notably, DC propagated from DAP12-deficient mice exhibit enhanced maturation in response to TLR ligation. Given the constitutive exposure of liver DC to endotoxin draining from the gut, we hypothesized that DAP12 might regulate liver DC maturation. We show that DAP12 is expressed by freshly isolated liver, spleen, kidney, and lung myeloid DC. Moreover, inhibition of DAP12 expression by liver DC using small interfering RNA promotes their phenotypic and functional maturation, resulting in enhanced TNF-α, IL-6, and IL-12p70 production, reduced secretion of IL-10, and enhanced CD4(+) and CD8(+) T cell proliferation. Furthermore, DAP12 silencing correlates with decreased STAT3 phosphorylation in mature liver DC and with diminished expression of the IL-1R-associated kinase-M, a negative regulator of TLR signaling. These findings highlight a regulatory role for DAP12 in hepatic DC maturation, and suggest a mechanism whereby this function may be induced/maintained.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Differentiation/immunology , Dendritic Cells/immunology , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Interleukin-10/biosynthesis , Liver/immunology , Myeloid Cells/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/deficiency , Animals , CHO Cells , Cell Differentiation/genetics , Cells, Cultured , Coculture Techniques , Cricetinae , Cricetulus , Dendritic Cells/cytology , Dendritic Cells/metabolism , Growth Inhibitors/biosynthesis , Growth Inhibitors/deficiency , Growth Inhibitors/physiology , Immune Tolerance/genetics , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-10/antagonists & inhibitors , Interleukin-10/physiology , Liver/cytology , Liver/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
13.
Eur J Immunol ; 41(2): 286-90, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21267998

ABSTRACT

Expression by DCs of co-inhibitory molecules such as programmed death ligand-1 (PD-L1/B7-H1/CD274), a member of the B7 superfamily, is crucial for the downregulation of T-cell responses and the maintenance of immune homeostasis. Exposure of immature DCs to danger-associated molecular patterns (DAMPS) or pathogen-associated molecular patterns (PAMPs) generally results in their maturation and acquisition of immunostimulatory function. However, exposure of DCs to TLR ligands early during their differentiation can inhibit further differentiation and confer tolerogenic properties on these APCs. A report in this issue of The European Journal of Immunology reveals that early inhibition of human DC differentiation from blood monocytes by TLR agonists is associated with a tolerogenic phenotype and Treg generation. The tolerogenic function of these APCs is dependent on MAPK-induced IL-6 and IL-10 production, which drives STAT-3-mediated PD-L1 expression. These observations link IL-10 and IL-6 to PD-L1 expression, providing a new dimension to the anti-inflammatory properties of these cytokines. These findings also have implications for understanding the inherent function of DCs in non-lymphoid tissues such as the liver and lung, where they are exposed to PAMPs that are found constitutively in the local microenvironment.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/physiology , Gene Expression Regulation/immunology , Immune Tolerance/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Antigen-Presenting Cells/cytology , Apoptosis Regulatory Proteins/physiology , B7-H1 Antigen , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Interleukin-10/metabolism , Interleukin-6/metabolism , Lipopolysaccharide Receptors/metabolism , Mitogen-Activated Protein Kinases/metabolism , Models, Immunological , Monocytes/cytology , Monocytes/immunology , Programmed Cell Death 1 Receptor , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptors/agonists
14.
Blood ; 115(23): 4758-69, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20335217

ABSTRACT

Prolonged inhibition of the kinase, mammalian target of rapamycin (mTOR), during myeloid dendritic cell (DC) generation confers resistance to maturation. Recently, however, mTOR inhibition immediately before Toll-like receptor ligation has been found to exert proinflammatory effects on myeloid cells, notably enhanced IL-12p40/p70 production. We show, for the first time, that mouse or human DCs generated under mTOR inhibition exhibit markedly enhanced IL-12p70 production after lipopolysaccharide (LPS) stimulation, despite impaired costimulatory molecule expression and poor T-cell stimulatory ability. Consistent with this finding, we reveal that increased IL-12p40 production occurs predominantly in CD86(lo) immature DCs. High IL-12p40/p70 production by CD86(lo) DC resulted from failed down-regulation of glycogen synthase kinase-3 (GSK-3) activity and could not be ascribed to enhanced Akt function. Despite high IL-12p70 secretion, rapamycin-conditioned, LPS-stimulated DCs remained poor T-cell stimulators, failing to enhance allogeneic Th1 cell responses. We also report that inhibition of GSK-3 impedes the ability of LPS-stimulated DCs to induce forkhead box p3 in CD4(+)CD25(-) T cells, as does the absence of IL-12p40/p70. Thus, GSK-3 activity in DC is regulated via signaling linked to mTOR and modulates their capacity both to produce IL-12p40/p70 and induce forkhead box p3 in CD4(+) T cells under inflammatory conditions.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/immunology , Glycogen Synthase Kinase 3/immunology , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/pharmacology , Myeloid Cells/immunology , Protein Serine-Threonine Kinases/immunology , Th1 Cells/immunology , Animals , B7-2 Antigen/genetics , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases , Th1 Cells/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
15.
J Immunol ; 184(2): 624-36, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20007530

ABSTRACT

Minimization of immunosuppression and donor-specific tolerance to MHC-mismatched organ grafts are important clinical goals. The therapeutic potential of regulatory T cells (Tregs) has been demonstrated, but conditions for optimizing their in vivo function posttransplant in nonlymphocyte-depleted hosts remain undefined. In this study, we address mechanisms through which inhibition of the mammalian target of rapamycin (Rapa) synergizes with alloantigen-specific Treg (AAsTreg) to permit long-term, donor-specific heart graft survival in immunocompetent hosts. Crucially, immature allogeneic dendritic cells allowed AAsTreg selection in vitro, with minimal expansion of unwanted (Th17) cells. The rendered Treg potently inhibited T cell proliferation in an Ag-specific manner. However, these AAsTreg remained unable to control T cells stimulated by allogeneic mature dendritic cells, a phenomenon dependent on the release of proinflammatory cytokines. In vivo, Rapa administration reduced danger-associated IL-6 production, T cell proliferation, and graft infiltration. Based on these observations, AAsTreg were administered posttransplant (day 7) in combination with a short course of Rapa and rendered >80% long-term (>150 d) graft survival, a result superior to that achieved with polyclonal Treg. Moreover, graft protection was alloantigen-specific. Significantly, long-term graft survival was associated with alloreactive T cell anergy. These findings delineate combination of transient mammalian target of Rapa inhibition with appropriate AAsTreg selection as an effective approach to promote long-term organ graft survival.


Subject(s)
Graft Survival/immunology , Heart Transplantation/methods , Intracellular Signaling Peptides and Proteins/drug effects , Protein Serine-Threonine Kinases/drug effects , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes, Regulatory/transplantation , Adjuvants, Immunologic , Animals , Graft Survival/drug effects , Humans , Immunocompetence , Isoantigens/immunology , Lymphocyte Transfusion/methods , Membrane Proteins/administration & dosage , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/cytology , TOR Serine-Threonine Kinases , Treatment Outcome
16.
J Invest Dermatol ; 142(3 Pt B): 841-848, 2022 03.
Article in English | MEDLINE | ID: mdl-34753621

ABSTRACT

Skin-resident mast cells (MCs) and cutaneous sensory neurons both play crucial roles in microbial‒host defense and inflammatory diseases. MCs can be directly activated by pathogens or their products, resulting in the release of numerous mediators that promote innate immune responses and also activate sensory neurons. Cutaneous sensory neurons can also directly detect the presence of pathogens, resulting in the release of neuropeptides that modulate MC function. In this review, we will focus on the reciprocal interactions between cutaneous sensory neurons and MCs and the importance of this cross-talk in skin diseases.


Subject(s)
Inflammation , Mast Cells , Humans , Immunity, Innate , Sensory Receptor Cells , Skin
17.
Front Cell Dev Biol ; 10: 687659, 2022.
Article in English | MEDLINE | ID: mdl-35350380

ABSTRACT

The purinergic receptor P2X7 (P2X7R) is important in inflammasome activation and generally considered to favor proinflammatory immune responses. However, there is still a limited understanding of the role of P2X7R signaling in Th cell differentiation, particularly, Th17 differentiation. Herein, the impact of P2X7R signaling on primary Th17 and Th1 cell responses was examined when P2X7R was expressed specifically on dendritic cells (DCs) and CD4+ T cells. Surprisingly, global genetic ablation and pharmacological inhibition of the P2X7R did not affect the generation of Th17 and Th1 development in response to immunization with Complete Freund's Adjuvant and the model antigens, keyhole limpet hemocyanin or OVA. However, in-depth in vitro and in vivo investigations revealed differences in the balance of Th1/Th17 differentiation when P2X7R blockade was restricted to either DCs or CD4+ T cells. In this regard, in vitro DCs treated with a P2X7R agonist released more IL-6 and IL-1ß and induced a more robust Th17 response in mixed leukocyte reactions when compared to controls. To test the hypothesis that P2X7R signaling specifically in DCs enhances Th17 responses in vivo, DC-specific P2X7R deficient chimeras were immunized with CFA and OVA. In this model, the P2X7R expression on DCs decreased the Th1 response without impacting Th17 responses. Following an assessment of CD4+ T cell P2X7R signaling, it was determined that in vitro P2X7R sufficient T cells develop an increased Th17 and suppressed Th1 differentiation profile. In vivo, P2X7R expression on CD4+ T cells had no effect on Th17 differentiation but likewise significantly suppressed the Th1 response, thereby skewing the immune balance. Interestingly, it appears that WT OT-II Th1 cells are more sensitive to P2X7R-induced cell death as evidence by a decrease in cell number and an increase in T cell death. Overall, these studies indicate that in vitro P2X7R signaling does enhances Th17 responses, which suggests that compensatory Th17 differentiation mechanisms are utilized in vivo in the absence of P2X7R signaling.

18.
iScience ; 25(10): 105045, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36062075

ABSTRACT

Sustainable global immunization campaigns against COVID-19 and other emerging infectious diseases require effective, broadly deployable vaccines. Here, we report a dissolvable microarray patch (MAP) SARS-CoV-2 vaccine that targets the immunoresponsive skin microenvironment, enabling efficacious needle-free immunization. Multicomponent MAPs delivering both SARS-CoV-2 S1 subunit antigen and the TLR3 agonist Poly(I:C) induce robust antibody and cellular immune responses systemically and in the respiratory mucosa. MAP vaccine-induced antibodies bind S1 and the SARS-CoV-2 receptor-binding domain, efficiently neutralize the virus, and persist at high levels for more than a year. The MAP platform reduces systemic toxicity of the delivered adjuvant and maintains vaccine stability without refrigeration. When applied to human skin, MAP vaccines activate skin-derived migratory antigen-presenting cells, supporting the feasibility of human translation. Ultimately, this shelf-stable MAP vaccine improves immunogenicity and safety compared to traditional intramuscular vaccines and offers an attractive alternative for global immunization efforts against a range of infectious pathogens.

19.
J Immunol ; 183(11): 6922-32, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19890047

ABSTRACT

The nucleotide-binding oligomerization domain (NOD)2/CARD15 protein, which senses muramyl dipeptide (MDP), a product of bacterial peptidoglycan, appears to play an important role in regulating intestinal immunity. Although the liver is exposed to gut-derived MDP, the influence of NOD2 ligation on hepatic APC, in particular dendritic cells (DC), is unknown. Freshly isolated mouse liver and spleen plasmacytoid (p)DC expressed higher levels of NOD2 message than conventional myeloid (m)DC. Following MDP stimulation in vivo, liver pDC, but not mDC, up-regulated expression of IFN regulatory factor 4 (IRF-4), a negative regulator of TLR signaling, and induced less allogeneic T cell proliferation and IFN-gamma production. The adoptive transfer of liver pDC from MDP-treated mice failed to prime allogeneic T cells in vivo. By contrast, splenic DC IRF-4 levels and T cell stimulatory activity remained unchanged. Liver pDC from MDP-stimulated mice also displayed greater IkappaBalpha, cell surface B7-H1, and B7-H1 relative to CD86 than control liver pDC. No similar effects were observed for liver mDC or spleen DC. Absence of B7-H1 on liver pDC reversed the inhibitory effect of MDP. After ex vivo stimulation with LPS or CpG, liver pDC but not mDC from MDP-treated animals secreted less IL-12p70, IL-6, and TNF-alpha and induced weaker allogeneic T cell proliferation than those from controls. Moreover, CpG-stimulated liver pDC from MDP-treated mice secreted less IFN-alpha than their splenic counterparts, and systemic levels of IFN-alpha were reduced in MDP-treated animals after CpG administration. These findings suggest that differential effects of NOD2 ligation on liver pDC may play a role in regulating hepatic innate and adaptive immunity.


Subject(s)
B7-1 Antigen/immunology , Dendritic Cells/immunology , Interferon-gamma/immunology , Liver/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Nod2 Signaling Adaptor Protein/immunology , Peptides/immunology , Acetylmuramyl-Alanyl-Isoglutamine/immunology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , B7-1 Antigen/biosynthesis , B7-H1 Antigen , Blotting, Western , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , I-kappa B Proteins/immunology , I-kappa B Proteins/metabolism , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interferon-gamma/biosynthesis , Liver/cytology , Lymphocyte Culture Test, Mixed , Male , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Nod2 Signaling Adaptor Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Up-Regulation
20.
Adv Drug Deliv Rev ; 171: 164-186, 2021 04.
Article in English | MEDLINE | ID: mdl-33539853

ABSTRACT

The COVID-19 pandemic is a serious threat to global health and the global economy. The ongoing race to develop a safe and efficacious vaccine to prevent infection by SARS-CoV-2, the causative agent for COVID-19, highlights the importance of vaccination to combat infectious pathogens. The highly accessible cutaneous microenvironment is an ideal target for vaccination since the skin harbors a high density of antigen-presenting cells and immune accessory cells with broad innate immune functions. Microarray patches (MAPs) are an attractive intracutaneous biocargo delivery system that enables safe, reproducible, and controlled administration of vaccine components (antigens, with or without adjuvants) to defined skin microenvironments. This review describes the structure of the SARS-CoV-2 virus and relevant antigenic targets for vaccination, summarizes key concepts of skin immunobiology in the context of prophylactic immunization, and presents an overview of MAP-mediated cutaneous vaccine delivery. Concluding remarks on MAP-based skin immunization are provided to contribute to the rational development of safe and effective MAP-delivered vaccines against emerging infectious diseases, including COVID-19.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Drug Development/trends , SARS-CoV-2/immunology , Skin/immunology , Transdermal Patch/trends , Administration, Cutaneous , COVID-19/metabolism , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/metabolism , Drug Development/methods , Humans , Immunity, Innate/drug effects , Immunity, Innate/physiology , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Skin/drug effects , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL