Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
PNAS Nexus ; 2(10): pgad305, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37781098

ABSTRACT

Actin-binding protein Profilin1 is an important regulator of actin cytoskeletal dynamics in cells and critical for embryonic development in higher eukaryotes. The objective of the present study was to examine the consequence of loss-of-function of Pfn1 in vascular endothelial cells (ECs) in vivo. We utilized a mouse model engineered for tamoxifen-inducible biallelic inactivation of the Pfn1 gene selectively in EC (Pfn1EC-KO). Widespread deletion of EC Pfn1 in adult mice leads to severe health complications presenting overt pathologies (endothelial cell death, infarct, and fibrosis) in major organ systems and evidence for inflammatory infiltrates, ultimately compromising the survival of animals within 3 weeks of gene ablation. Mice deficient in endothelial Pfn1 exhibit selective bias toward the proinflammatory myeloid-derived population of immune cells, a finding further supported by systemic elevation of proinflammatory cytokines. We further show that triggering Pfn1 depletion not only directly upregulates proinflammatory cytokine/chemokine gene expression in EC but also potentiates the paracrine effect of EC on proinflammatory gene expression in macrophages. Consistent with these findings, we provide further evidence for increased activation of Interferon Regulatory Factor 7 (IRF7) and STAT1 in EC when depleted of Pfn1. Collectively, these findings for the first time demonstrate a prominent immunological consequence of loss of endothelial Pfn1 and an indispensable role of endothelial Pfn1 in mammalian survival unlike tolerable phenotypes of Pfn1 loss in other differentiated cell types.

2.
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
3.
Sci Transl Med ; 13(585)2021 03 17.
Article in English | MEDLINE | ID: mdl-33731430

ABSTRACT

Despite the role of donor-specific antibodies (DSAs) in recognizing major histocompatibility complex (MHC) antigens and mediating transplant rejection, how and where recipient B cells in lymphoid tissues encounter donor MHC antigens remains unclear. Contrary to the dogma, we demonstrated here that migration of donor leukocytes out of skin or heart allografts is not necessary for B or T cell allosensitization in mice. We found that mouse skin and cardiac allografts and human skin grafts release cell-free donor MHC antigens via extracellular vesicles (EVs) that are captured by subcapsular sinus (SCS) macrophages in lymph nodes or analog macrophages in the spleen. Donor EVs were transported across the SCS macrophages, and donor MHC molecules on the EVs were recognized by alloreactive B cells. This triggered B cell activation and DSA production, which were both prevented by SCS macrophage depletion. These results reveal an unexpected role for graft-derived EVs and open venues to interfere with EV biogenesis, trafficking, or function to restrain priming or reactivation of alloreactive B cells.


Subject(s)
Extracellular Vesicles , Heart Transplantation , Animals , B-Lymphocytes , Graft Rejection , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
Immunity ; 53(1): 16-18, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32668224

ABSTRACT

The role of keratinocyte metabolism in psoriasis is not fully elucidated. In this issue of Immunity, Lou et al. describe that interleukin-17 (IL-17) re-programs the urea cycle in keratinocytes increasing polyamines that stabilize RNA-Ag-complexes that upon cellular turnover activate dendritic cells, which amplify psoriasis inflammation.


Subject(s)
Psoriasis , RNA , Dendritic Cells , Humans , Keratinocytes , Polyamines
5.
Cell Rep ; 30(10): 3448-3465.e8, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160549

ABSTRACT

Efficient Ca2+ flux induced during cognate T cell activation requires signaling the T cell receptor (TCR) and unidentified G-protein-coupled receptors (GPCRs). T cells express the neurokinin-1 receptor (NK1R), a GPCR that mediates Ca2+ flux in excitable and non-excitable cells. However, the role of the NK1R in TCR signaling remains unknown. We show that the NK1R and its agonists, the neuropeptides substance P and hemokinin-1, co-localize within the immune synapse during cognate activation of T cells. Simultaneous TCR and NK1R stimulation is necessary for efficient Ca2+ flux and Ca2+-dependent signaling that sustains the survival of activated T cells and helper 1 (Th1) and Th17 bias. In a model of contact dermatitis, mice with T cells deficient in NK1R or its agonists exhibit impaired cellular immunity, due to high mortality of activated T cells. We demonstrate an effect of the NK1R in T cells that is relevant for immunotherapies based on pro-inflammatory neuropeptides and its receptors.


Subject(s)
Calcium/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction , T-Lymphocytes/immunology , Animals , Autocrine Communication/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Polarity/drug effects , Cell Survival/drug effects , Immunological Synapses/drug effects , Immunological Synapses/metabolism , Interleukin-2/metabolism , Lymphocyte Activation/drug effects , Mice , NF-kappa B/metabolism , Receptors, Neurokinin-1/agonists , Signal Transduction/drug effects , Substance P/pharmacology , T-Lymphocytes/drug effects , Tachykinins/pharmacology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
6.
Curr Opin Organ Transplant ; 24(6): 670-678, 2019 12.
Article in English | MEDLINE | ID: mdl-31592838

ABSTRACT

PURPOSE OF REVIEW: Extracellular vesicles released by prokaryote or eukaryote cells are emerging as mechanisms of cell-to-cell communication, by either physically interacting with the surface of target cells or transferring proteins/peptides, lipids, carbohydrates, and nuclei acids to acceptor cells. Accumulating evidence indicates that extracellular vesicles, among other functions, regulate innate and adaptive immune responses. We revisit here the effects that extracellular vesicles of various origins have on innate immunity. RECENT FINDINGS: Extracellular vesicles comprise a heterogeneous group of vesicles with different biogenesis, composition and biological properties, which include exosomes, microvesicles, apoptotic cell-derived extracellular vesicles, and other extracellular vesicles still not well characterized. Extracellular vesicles released by pathogens, leukocytes, nonhematopoietic cells, tumor cells, and likely allografts, can either stimulate or suppress innate immunity via multiple mechanisms. These include transfer to target leukocytes of pro-inflammatory or anti-inflammatory mediators, membrane receptors, enzymes, mRNAs, and noncoding RNAs; and interaction of extracellular vesicles with the complement and coagulation systems. As a result, extracellular vesicles affect differentiation, polarization, activation, tissue recruitment, cytokine and chemokine production, cytolytic and phagocytic function, and antigen transfer ability, of different types of innate immune cells. SUMMARY: The field of intercellular communication via extracellular vesicles is a rapid evolving area and the effects of pathogen-derived and host-derived extracellular vesicles on innate immunity in particular, have received increasing attention during the past decade. Future studies will be necessary to assess the full potential of the crosstalk between extracellular vesicles and the innate immune system and its use for therapeutic applications to treat chronic inflammation-based diseases and cancer growth and dissemination, among the growing list of disorders in which the innate immune system plays a critical role.


Subject(s)
Extracellular Vesicles/immunology , Immunity, Innate/immunology , Humans
7.
Eur J Immunol ; 49(11): 2095-2102, 2019 11.
Article in English | MEDLINE | ID: mdl-31334839

ABSTRACT

There is increasing evidence of the relevant connection and regulation between the gut and skin immune axis. In fact, oral administration of lipoteichoic acid (LTA) from Lactobacillus rhamnosus GG (LGG) prevents the development of UV-induced skin tumors in chronically exposed mice. Here we aim to evaluate whether this LTA is able to revert UV-induced immunosuppression as a mechanism involved in its anti-tumor effect and whether it has an immunotherapeutic effect against cutaneous squamous cell carcinoma. Using a mouse model of contact hypersensitivity, we demonstrate that LTA overcomes UV-induced skin immunosuppression. This effect was in part achieved by modulating the phenotype of lymph node resident dendritic cells (DC) and the homing of skin migratory DC. Importantly, oral LTA reduced significantly the growth of established skin tumors once UV radiation was discontinued, demonstrating that it has a therapeutic, besides the already demonstrated preventive antitumor effect. The data presented here strongly indicates that oral administration of LTA represents a promising immunotherapeutic approach for different conditions in which the skin immune system is compromised.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Lacticaseibacillus rhamnosus/chemistry , Lipopolysaccharides/pharmacology , Skin Neoplasms/drug therapy , Teichoic Acids/pharmacology , Ultraviolet Rays/adverse effects , Administration, Oral , Animals , Antineoplastic Agents/isolation & purification , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Movement/drug effects , Cell Movement/immunology , Cell Movement/radiation effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/radiation effects , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/radiation effects , Lipopolysaccharides/isolation & purification , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/radiation effects , Mice , Mice, Inbred C57BL , Skin/drug effects , Skin/immunology , Skin/pathology , Skin/radiation effects , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Teichoic Acids/isolation & purification
8.
J Invest Dermatol ; 137(5): 1042-1050, 2017 05.
Article in English | MEDLINE | ID: mdl-28012718

ABSTRACT

Loss of CD28 expression by CD8+ T cells occurs with age and during chronic inflammatory conditions. CD8+CD28- T cells are a heterogeneous cell subpopulation whose function ranges from immunosuppressive to effector. Here we analyzed the role of CD8+CD28- T cells in the pathogenesis of systemic sclerosis (SSc), a connective tissue disorder characterized by autoimmunity, vasculopathy, and extensive cutaneous and visceral fibrosis. We show that the frequency of CD8+CD28- T cells is increased in the blood and affected skin of SSc patients, independent of patient age, and correlates with the extent of skin fibrosis. We found that most skin-tropic CD8+CD28- T cells are resident in the skin lesions of patients in the early stage of the disease, exhibit an effector memory phenotype, and present a strong cytolytic activity ex vivo. Skin-resident and circulating SSc CD8+CD28- T cells produce high levels of the profibrotic cytokine IL-13, which induces collagen production by normal and SSc dermal fibroblasts. Thus, our findings indicate that CD8+CD28- T cells represent a pathogenic T-cell subset in SSc and likely play a critical role in the early stage of SSc skin disease.


Subject(s)
CD28 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-13/metabolism , Scleroderma, Systemic/physiopathology , Skin/pathology , Adult , Aged , Case-Control Studies , Collagen/metabolism , Female , Fibrosis , Humans , Male , Middle Aged , Phenotype , Scleroderma, Systemic/immunology , Skin/immunology , Young Adult
9.
J Clin Invest ; 126(8): 2805-20, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27348586

ABSTRACT

The immune response against transplanted allografts is one of the most potent reactions mounted by the immune system. The acute rejection response has been attributed to donor dendritic cells (DCs), which migrate to recipient lymphoid tissues and directly activate alloreactive T cells against donor MHC molecules. Here, using a murine heart transplant model, we determined that only a small number of donor DCs reach lymphoid tissues and investigated how this limited population of donor DCs efficiently initiates the alloreactive T cell response that causes acute rejection. In our mouse model, efficient passage of donor MHC molecules to recipient conventional DCs (cDCs) was dependent on the transfer of extracellular vesicles (EVs) from donor DCs that migrated from the graft to lymphoid tissues. These EVs shared characteristics with exosomes and were internalized or remained attached to the recipient cDCs. Recipient cDCs that acquired exosomes became activated and triggered full activation of alloreactive T cells. Depletion of recipient cDCs after cardiac transplantation drastically decreased presentation of donor MHC molecules to directly alloreactive T cells and delayed graft rejection in mice. These findings support a key role for transfer of donor EVs in the generation of allograft-targeting immune responses and suggest that interrupting this process has potential to dampen the immune response to allografts.


Subject(s)
Allografts/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Exosomes/metabolism , Immune Tolerance/immunology , Animals , Cell Movement , Graft Rejection , Graft Survival , Heart Transplantation , Major Histocompatibility Complex/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Skin Transplantation , Spleen/metabolism , T-Lymphocytes/cytology , Transplantation, Homologous
10.
Stem Cells ; 34(5): 1142-50, 2016 05.
Article in English | MEDLINE | ID: mdl-26865545

ABSTRACT

The main limitations to the success of transplantation are the antigraft response developed by the recipient immune system, and the adverse side effects of chronic immunosuppression. Graft-versus-host disease (GVHD) triggered by donor-derived T lymphocytes against the recipient tissues is another serious obstacle in the field of hematopoietic stem cell transplantation. Several laboratories have tested the possibility of promoting antigen (Ag)-specific tolerance for therapy of graft rejection, GVHD, and autoimmune disorders, by developing methodologies that mimic the mechanisms by which the immune system maintains peripheral tolerance in the steady state. It has been long recognized that the silent clearance of cells undergoing apoptosis exerts potent immune-regulatory effects and provides apoptotic cell-derived Ags to those Ag-presenting cells (APCs) that internalize them, in particular macrophages and dendritic cells. Therefore, in situ-targeting of recipient APCs by systemic administration of leukocytes in early apoptosis and bearing donor Ags represents a relatively simple approach to control the antidonor response against allografts. Here, we review the mechanisms by which apoptotic cells are silently cleared by phagocytes, and how such phenomenon leads to down-regulation of the innate and adaptive immunity. We discuss the evolution of apoptotic cell-based therapies from murine models of organ/tissue transplantation and GVHD, to clinical trials. We make emphasis on potential limitations and areas of concern of apoptotic cell-based therapies, and on how other immune-suppressive therapies used in the clinics or tested experimentally likely also function through the silent clearance of apoptotic cells by the immune system. Stem Cells 2016;34:1142-1150.


Subject(s)
Apoptosis , Cell- and Tissue-Based Therapy , Graft Rejection/therapy , Graft vs Host Disease/therapy , Animals , Graft Rejection/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immunosuppression Therapy
11.
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
12.
Methods Mol Biol ; 1186: 87-102, 2014.
Article in English | MEDLINE | ID: mdl-25149305

ABSTRACT

Exosomes are membrane nanovesicles (approximately <120 nm in size) released by most, if not all, living cells and in particular by leukocytes. They originate within the endocytic compartment by invagination of the endosome membrane. Therefore, they have a different biogenesis and molecular composition than microvesicles (>0.2 µm) shed from the plasma membrane. Although the functions of exosomes in vivo are beginning to be elucidated, increasing evidence suggests that exosomes constitute a mechanism of cell-to-cell communication, transferring antigens, proteins, mRNAs, and noncoding RNAs among cells. Interestingly, effector T cells including cytotoxic T lymphocytes (CTLs) release death-inducing molecules of the TNF superfamily through exosomes contained in their cytotoxic granules. The present chapter provides basic protocols for purification of exosomes secreted by CTLs.


Subject(s)
Exosomes/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen-Presenting Cells/immunology , Apoptosis/immunology , Biological Transport , Cell Communication , Centrifugation, Density Gradient/methods , Cytoplasmic Granules/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/cytology
13.
Methods Mol Biol ; 1024: 19-40, 2013.
Article in English | MEDLINE | ID: mdl-23719940

ABSTRACT

Exosomes are extremely small (<100 nm) membrane vesicles, generated in the endocytic compartment that are released to the extracellular milieu by living cells. Although the biological function of exosomes in vivo remains unclear, they seem to function as mechanisms of cell-to-cell communication for horizontal transfer of proteins, antigens, prions, morphogens, mRNA, and noncoding regulatory RNAs, including microRNAs (miRNAs) (also known as exosome-shuttle miRNAs). Dendritic cells (DCs), the most potent professional antigen-presenting leukocytes of the immune system, release relatively high levels of exosomes and also interact with free exosomes present in the extracellular space. Therefore, DCs constitute a good model for the analysis of exosome-shuttle miRNAs and their horizontal propagation between cells. This chapter provides basic protocols for purification of exosomes released by mouse bone marrow-derived DCs, analysis of their miRNA content, and assessment of the function of exosome-shuttle miRNAs, once they are transferred to target/acceptor DCs.


Subject(s)
Bone Marrow Cells/chemistry , Dendritic Cells/chemistry , Exosomes/chemistry , MicroRNAs/isolation & purification , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Communication , Culture Media, Conditioned/chemistry , Dendritic Cells/cytology , Dendritic Cells/metabolism , Electrophoresis, Polyacrylamide Gel , Exosomes/genetics , Exosomes/ultrastructure , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron , RNA Transport , Ultracentrifugation
14.
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
15.
Arthritis Rheum ; 65(1): 236-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23001877

ABSTRACT

OBJECTIVE: Fibrosis is a major contributor to morbidity and mortality in systemic sclerosis (SSc). T cells are the predominant inflammatory infiltrate in affected tissue and are thought to produce cytokines that drive the synthesis of extracellular matrix (ECM) proteins by fibroblasts, resulting in excessive fibrosis. We have previously shown that aberrant interleukin-13 (IL-13) production by peripheral blood effector CD8+ T cells from SSc patients correlates with the extent of skin fibrosis. The present study was undertaken to investigate the role of IL-13 production by CD8+ T cells in dermal fibrosis, an early and specific manifestation of SSc. METHODS: ECM protein production by normal dermal fibroblasts cocultured with SSc CD8+ T cell supernatants was determined by quantitative polymerase chain reaction and Western blotting. Skin-homing receptor expression and IL-13 production by CD8+ T cells in the peripheral blood of SSc patients were measured by flow cytometry. IL-13+ and CD8+ cells in sclerotic skin were identified by immunohistochemistry. RESULTS: IL-13-producing circulating CD8+ T cells from patients with SSc expressed skin-homing receptors and induced a profibrotic phenotype in normal dermal fibroblasts, which was inhibited by an anti-IL-13 antibody. High numbers of CD8+ T cells and IL-13+ cells were found in the skin lesions of SSc patients, particularly during the early inflammatory phase of the disease. CONCLUSION: These findings show that IL-13-producing CD8+ T cells are directly involved in modulating dermal fibrosis in SSc. The demonstration that CD8+ T cells homing to the skin early in the course of SSc are associated with accumulation of IL-13 is an important mechanistic contribution to the understanding of the pathogenesis of dermal fibrosis in SSc and may represent a potential target for therapeutic intervention.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Fibroblasts/pathology , Interleukin-13/metabolism , Scleroderma, Systemic/pathology , Skin/pathology , Adolescent , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Child , Female , Fibrosis , Flow Cytometry , Humans , Immunohistochemistry , Male , Middle Aged , Scleroderma, Systemic/immunology , Skin/immunology , Young Adult
16.
J Invest Dermatol ; 133(2): 460-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22951718

ABSTRACT

Human beta-defensins (hBDs) are antimicrobial peptides that have an important role in innate immune responses at epithelial barriers such as the skin. However, the role that hBDs have in initiating cellular immune responses that contribute to antigen-specific adaptive immunity is not well understood. Here we show that one member of the hBD family, hBD3, can induce maturation and T-helper type 1 skewing function in human Langerhans cell-like dendritic cells (LC-DCs). Specifically, hBD3 potently induces phenotypic maturation of LC-DCs, including increased expression of CCR7, which mediates functional chemotactic responses to CCL19 and CCL21. hBD3-stimulated LC-DCs induce strong proliferation of and IFN-γ secretion by naive human T cells. hBD3 also induces phenotypic maturation of primary human skin-migratory DCs derived from human skin explants. These results suggest an important role for hBD3 in inducing DC activation, migration, and polarization. Thus, hBD3 contributes to the integration of innate and adaptive immune responses in the skin, and may be a useful adjuvant for skin immunization and an important factor in the pathophysiology of inflammatory skin diseases.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Immunization/methods , Langerhans Cells/immunology , beta-Defensins/immunology , Adjuvants, Immunologic/pharmacology , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Cell Movement/drug effects , Cell Movement/immunology , Cell Polarity/drug effects , Cell Polarity/immunology , Cell Proliferation/drug effects , Chemokine CCL19/immunology , Chemokine CCL19/metabolism , Chemokine CCL21/immunology , Chemokine CCL21/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/immunology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Immunophenotyping , Interferon-gamma/metabolism , Langerhans Cells/cytology , Langerhans Cells/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , beta-Defensins/metabolism , beta-Defensins/pharmacology
17.
J Immunol ; 189(9): 4621-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018455

ABSTRACT

Aberrant activation of macrophages in arterial walls by oxidized lipoproteins can lead to atherosclerosis. Oxidized lipoproteins convert macrophages to foam cells through lipid uptake and TLR signaling. To investigate the relative contributions of lipid uptake and TLR signaling in foam cell formation, we established an in vitro assay using liposomes of defined lipid compositions. We found that TLRs signaling through Toll/IL-1R domain-containing adapter inducing IFN-ß promoted foam cell formation by inducing both NF-κB signaling and type I IFN production, whereas TLRs that do not induce IFN, like TLR2, did not enhance foam cell formation. Addition of IFN-α to TLR2 activator promoted robust foam cell formation. TLR signaling further required peroxisome proliferator-activated receptor α, as inhibition of peroxisome proliferator-activated receptor α blocked foam cell formation. We then investigated the ability of endogenous microparticles (MP) to contribute to foam cell formation. We found that lipid-containing MP promoted foam cell formation, which was enhanced by TLR stimulation or IFN-α. These MP also stimulated foam cell formation in a human skin model. However, these MP suppressed TNF-α production and T cell activation, showing that foam cell formation can occur by immunosuppressive MP. Taken together, the data reveal novel signaling requirements for foam cell formation and suggest that uptake of distinct types of MP in the context of activation of multiple distinct TLR can induce foam cell formation.


Subject(s)
Cell Differentiation/immunology , Cell-Derived Microparticles/immunology , Foam Cells/immunology , Foam Cells/metabolism , Macrophages/immunology , Toll-Like Receptors/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Cells, Cultured , Foam Cells/pathology , HeLa Cells , Humans , Ligands , Lipid Metabolism/immunology , Macrophages/metabolism , Macrophages/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , Toll-Like Receptors/physiology
18.
PLoS One ; 7(8): e43049, 2012.
Article in English | MEDLINE | ID: mdl-22900087

ABSTRACT

Extracellular matrix deposition and tissue scarring characterize the process of fibrosis. Transforming growth factor beta (TGFß) and Insulin-like growth factor binding protein-3 (IGFBP-3) have been implicated in the pathogenesis of fibrosis in various tissues by inducing mesenchymal cell proliferation and extracellular matrix deposition. We identified Syndecan-2 (SDC2) as a gene induced by TGFß in an IGFBP-3-dependent manner. TGFß induction of SDC2 mRNA and protein required IGFBP-3. IGFBP-3 independently induced production of SDC2 in primary fibroblasts. Using an ex-vivo model of human skin in organ culture expressing IGFBP-3, we demonstrate that IGFBP-3 induces SDC2 ex vivo in human tissue. We also identified Mitogen-activated protein kinase-interacting kinase (Mknk2) as a gene induced by IGFBP-3. IGFBP-3 triggered Mknk2 phosphorylation resulting in its activation. Mknk2 independently induced SDC2 in human skin. Since IGFBP-3 is over-expressed in fibrotic tissues, we examined SDC2 levels in skin and lung tissues of patients with systemic sclerosis (SSc) and lung tissues of patients with idiopathic pulmonary fibrosis (IPF). SDC2 levels were increased in fibrotic dermal and lung tissues of patients with SSc and in lung tissues of patients with IPF. This is the first report describing elevated levels of SDC2 in fibrosis. Increased SDC2 expression is due, at least in part, to the activity of two pro-fibrotic factors, TGFß and IGFBP-3.


Subject(s)
Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Regulation , Insulin-Like Growth Factor Binding Protein 3/metabolism , Syndecan-2/genetics , Cells, Cultured , Enzyme Activation , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Skin/metabolism , Skin/pathology , Syndecan-2/metabolism , Time Factors , Transforming Growth Factor beta1/pharmacology
19.
Sci Transl Med ; 4(136): 136ra71, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22649092

ABSTRACT

Fibroproliferative disorders such as idiopathic pulmonary fibrosis and systemic sclerosis have no effective therapies and result in significant morbidity and mortality due to progressive organ fibrosis. We examined the effect of peptides derived from endostatin on existing fibrosis and fibrosis triggered by two potent mediators, transforming growth factor-ß (TGF-ß) and bleomycin, in human and mouse tissues in vitro, ex vivo, and in vivo. We identified one peptide, E4, with potent antifibrotic activity. E4 prevented TGF-ß-induced dermal fibrosis in vivo in a mouse model, ex vivo in human skin, and in bleomycin-induced dermal and pulmonary fibrosis in vivo, demonstrating that E4 exerts potent antifibrotic effects. In addition, E4 significantly reduced existing fibrosis in these preclinical models. E4 amelioration of fibrosis was accompanied by reduced cell apoptosis and lower levels of lysyl oxidase, an enzyme that cross-links collagen, and Egr-1 (early growth response gene-1), a transcription factor that mediates the effects of several fibrotic triggers. Our findings identify E4 as a peptide with potent antifibrotic activity and a possible therapeutic agent for organ fibrosis.


Subject(s)
Endostatins/chemistry , Fibrosis/drug therapy , Peptides/chemistry , Peptides/therapeutic use , Animals , Bleomycin/pharmacology , Fibrosis/chemically induced , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Skin/drug effects , Skin/metabolism , Skin/pathology , Transforming Growth Factor beta/pharmacology
20.
Blood ; 119(3): 756-66, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22031862

ABSTRACT

Dendritic cells (DCs) are the most potent APCs. Whereas immature DCs down-regulate T-cell responses to induce/maintain immunologic tolerance, mature DCs promote immunity. To amplify their functions, DCs communicate with neighboring DCs through soluble mediators, cell-to-cell contact, and vesicle exchange. Transfer of nanovesicles (< 100 nm) derived from the endocytic pathway (termed exosomes) represents a novel mechanism of DC-to-DC communication. The facts that exosomes contain exosome-shuttle miRNAs and DC functions can be regulated by exogenous miRNAs, suggest that DC-to-DC interactions could be mediated through exosome-shuttle miRNAs, a hypothesis that remains to be tested. Importantly, the mechanism of transfer of exosome-shuttle miRNAs from the exosome lumen to the cytosol of target cells is unknown. Here, we demonstrate that DCs release exosomes with different miRNAs depending on the maturation of the DCs. By visualizing spontaneous transfer of exosomes between DCs, we demonstrate that exosomes fused with the target DCs, the latter followed by release of the exosome content into the DC cytosol. Importantly, exosome-shuttle miRNAs are functional, because they repress target mRNAs of acceptor DCs. Our findings unveil a mechanism of transfer of exosome-shuttle miRNAs between DCs and its role as a means of communication and posttranscriptional regulation between DCs.


Subject(s)
Cell Communication , Dendritic Cells/metabolism , Endosomes/metabolism , Exosomes/genetics , MicroRNAs/physiology , Animals , Antigen Presentation , Biomarkers/metabolism , Cytosol/metabolism , Dendritic Cells/cytology , Exosomes/metabolism , Gene Expression Profiling , Membrane Fusion , Mice , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...