Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
J Immunother Cancer ; 11(3)2023 03.
Article in English | MEDLINE | ID: mdl-36990508

ABSTRACT

BACKGROUND: High-risk human papillomavirus (HPV) is a primary cause of an increasing number of oropharyngeal squamous cell carcinomas (OPSCCs). The viral etiology of these cancers provides the opportunity for antigen-directed therapies that are restricted in scope compared with cancers without viral components. However, specific virally-encoded epitopes and their corresponding immune responses are not fully defined. METHODS: To understand the OPSCC immune landscape, we conducted a comprehensive single-cell analysis of HPV16+ and HPV33+ primary tumors and metastatic lymph nodes. We used single-cell analysis with encoded peptide-human leukocyte antigen (HLA) tetramers to analyze HPV16+ and HPV33+ OPSCC tumors, characterizing the ex vivo cellular responses to HPV-derived antigens presented in major Class I and Class II HLA alleles. RESULTS: We identified robust cytotoxic T-cell responses to HPV16 proteins E1 and E2 that were shared across multiple patients, particularly in HLA-A*01:01 and HLA-B*08:01. Responses to E2 were associated with loss of E2 expression in at least one tumor, indicating the functional capacity of these E2-recognizing T cells and many of these interactions validated in a functional assay. Conversely, cellular responses to E6 and E7 were limited in quantity and cytotoxic capacity, and tumor E6 and E7 expression persisted. CONCLUSIONS: These data highlight antigenicity beyond HPV16 E6 and E7 and nominate candidates for antigen-directed therapies.


Subject(s)
Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Humans , Human papillomavirus 16 , Tumor Microenvironment
2.
Sci Immunol ; 7(67): eabk3070, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34793243

ABSTRACT

Effective presentation of antigens by human leukocyte antigen (HLA) class I molecules to CD8+ T cells is required for viral elimination and generation of long-term immunological memory. In this study, we applied a single-cell, multiomic technology to generate a unified ex vivo characterization of the CD8+ T cell response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) across four major HLA class I alleles. We found that HLA genotype conditions key features of epitope specificity, TCRα/ß sequence diversity, and the utilization of pre-existing SARS-CoV-2-reactive memory T cell pools. Single-cell transcriptomics revealed functionally diverse T cell phenotypes of SARS-CoV-2-reactive T cells, associated with both disease stage and epitope specificity. Our results show that HLA variations notably influence the CD8+ T cell repertoire shape and utilization of immune recall upon SARS-CoV-2 infection.


Subject(s)
Alleles , CD8-Positive T-Lymphocytes/immunology , COVID-19 , Histocompatibility Antigens Class I/immunology , Memory T Cells/immunology , Receptors, Antigen, T-Cell, alpha-beta , SARS-CoV-2/immunology , COVID-19/genetics , COVID-19/immunology , Histocompatibility Antigens Class I/genetics , Humans , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , SARS-CoV-2/genetics
3.
Cell Rep ; 35(10): 109224, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34107252

ABSTRACT

Drug development is hampered by poor target selection. Phenotypic screens using neurons differentiated from patient stem cells offer the possibility to validate known and discover novel disease targets in an unbiased fashion. To identify targets for managing hyperexcitability, a pathological feature of amyotrophic lateral sclerosis (ALS), we design a multi-step screening funnel using patient-derived motor neurons. High-content live cell imaging is used to evaluate neuronal excitability, and from a screen against a chemogenomic library of 2,899 target-annotated compounds, 67 reduce the hyperexcitability of ALS motor neurons carrying the SOD1(A4V) mutation, without cytotoxicity. Bioinformatic deconvolution identifies 13 targets that modulate motor neuron excitability, including two known ALS excitability modulators, AMPA receptors and Kv7.2/3 ion channels, constituting target validation. We also identify D2 dopamine receptors as modulators of ALS motor neuron excitability. This screen demonstrates the power of human disease cell-based phenotypic screens for identifying clinically relevant targets for neurological disorders.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cell Differentiation , Humans , Phenotype
4.
Immunohorizons ; 4(6): 332-338, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32554437

ABSTRACT

Several human autoimmune diseases are characterized by increased expression of type 1 IFN-stimulated genes in both the peripheral blood and tissue. The contributions of different type I IFNs to this gene signature are uncertain as the type I IFN family consists of 13 alphas and one each of ß, ε, κ, and ω subtypes. We sought to investigate the contribution of various IFNs to IFN signaling in primary human cell types. We stimulated primary skin, muscle, kidney, and PBMCs from normal healthy human donors with various TLR ligands and measured the expression of type I IFN subtypes and activation of downstream signaling by quantitative PCR. We show that IFNB1 is the dominant type I IFN expressed upon TLR3 and TLR4 stimulation, and its expression profile is associated with subsequent MX1 transcription. Furthermore, using an IFN-ß-specific neutralizing Ab, we show that MX1 expression is inhibited in a dose-dependent manner, suggesting that IFN-ß is the primary driver of IFN-stimulated genes following TLR3 and TLR4 engagement. Stimulation with TLR7/8 and TLR9 ligands induced IFNB1 and IFNA subtypes and MX1 expression only in PBMCs and not in tissue resident cell types. Concordantly, IFN-ß neutralization had no effect on MX1 expression in PBMCs potentially because of the combination of IFNB1 and IFNA expression. Combined, these data highlight the potential role for IFN-ß in driving local inflammatory responses in clinically relevant human tissue types and opportunities to treat local inflammation by targeting IFN-ß.


Subject(s)
Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Myxovirus Resistance Proteins/metabolism , Cells, Cultured , Healthy Volunteers , Humans , Interferon-alpha/genetics , Interferon-beta/genetics , Ligands , Myxovirus Resistance Proteins/genetics , Signal Transduction/immunology , Toll-Like Receptor 3 , Toll-Like Receptor 4 , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Toll-Like Receptor 9
6.
J Biol Chem ; 293(9): 3039-3055, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29317497

ABSTRACT

Slit guidance ligand 2 (SLIT2) is a large, secreted protein that binds roundabout (ROBO) receptors on multiple cell types, including neurons and kidney podocytes. SLIT2-ROBO-mediated signaling regulates neuronal migration and ureteric bud (UB) outgrowth during kidney development as well as glomerular filtration in adult kidneys. Additionally, SLIT2 binds Gremlin, an antagonist of bone morphogenetic proteins (BMPs), and BMP-Gremlin signaling also regulates UB formation. However, direct cross-talk between the ROBO2-SLIT2 and BMP-Gremlin signaling pathways has not been established. Here, we report the discovery of negative feedback between the SLIT2 and BMP-Gremlin signaling pathways. We found that the SLIT2-Gremlin interaction inhibited both SLIT2-ROBO2 signaling in neurons and Gremlin antagonism of BMP activity in myoblasts and fibroblasts. Furthermore, BMP2 down-regulated SLIT2 expression and promoter activity through canonical BMP signaling. Gremlin treatment, BMP receptor inhibition, and SMAD family member 4 (SMAD4) knockdown rescued BMP-mediated repression of SLIT2. BMP2 treatment of nephron progenitor cells derived from human embryonic stem cells decreased SLIT2 expression, further suggesting an interaction between the BMP2-Gremlin and SLIT2 pathways in human kidney cells. In conclusion, our study has revealed direct negative cross-talk between two pathways, previously thought to be unassociated, that may regulate both kidney development and adult tissue maintenance.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Bone Morphogenetic Protein 2/pharmacology , Cell Movement/drug effects , Down-Regulation/drug effects , Feedback, Physiological/drug effects , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Promoter Regions, Genetic/genetics , Protein Domains , Signal Transduction/drug effects
7.
JCI Insight ; 1(19): e86934, 2016 Nov 17.
Article in English | MEDLINE | ID: mdl-27882344

ABSTRACT

The repulsive guidance cue SLIT2 and its receptor ROBO2 are required for kidney development and podocyte foot process structure, but the SLIT2/ROBO2 signaling mechanism regulating podocyte function is not known. Here we report that a potentially novel signaling pathway consisting of SLIT/ROBO Rho GTPase activating protein 1 (SRGAP1) and nonmuscle myosin IIA (NMIIA) regulates podocyte adhesion downstream of ROBO2. We found that the myosin II regulatory light chain (MRLC), a subunit of NMIIA, interacts directly with SRGAP1 and forms a complex with ROBO2/SRGAP1/NMIIA in the presence of SLIT2. Immunostaining demonstrated that SRGAP1 is a podocyte protein and is colocalized with ROBO2 on the basal surface of podocytes. In addition, SLIT2 stimulation inhibits NMIIA activity, decreases focal adhesion formation, and reduces podocyte attachment to collagen. In vivo studies further showed that podocyte-specific knockout of Robo2 protects mice from hypertension-induced podocyte detachment and albuminuria and also partially rescues the podocyte-loss phenotype in Myh9 knockout mice. Thus, we have identified SLIT2/ROBO2/SRGAP1/NMIIA as a potentially novel signaling pathway in kidney podocytes, which may play a role in regulating podocyte adhesion and attachment. Our findings also suggest that SLIT2/ROBO2 signaling might be a therapeutic target for kidney diseases associated with podocyte detachment and loss.


Subject(s)
GTPase-Activating Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nonmuscle Myosin Type IIA/metabolism , Podocytes/cytology , Receptors, Immunologic/metabolism , Signal Transduction , Animals , Cell Movement , Kidney , Mice , Mice, Knockout
8.
Sci Transl Med ; 8(353): 353ra112, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27559095

ABSTRACT

Thrombosis is a major cause of morbidity and mortality. Current antithrombotic drugs are not ideal in that they must balance prevention of thrombosis against bleeding risk. Inhibition of coagulation factor XI (FXI) may offer an improvement over existing antithrombotic strategies by preventing some forms of thrombosis with lower bleeding risk. To permit exploration of this hypothesis in humans, we generated and characterized a series of human immunoglobulin Gs (IgGs) that blocked FXIa active-site function but did not bind FXI zymogen or other coagulation proteases. The most potent of these IgGs, C24 and DEF, inhibited clotting in whole human blood and prevented FeCl3-induced carotid artery occlusion in FXI-deficient mice reconstituted with human FXI and in thread-induced venous thrombosis in rabbits at clinically relevant doses. At doses substantially higher than those required for inhibition of intravascular thrombus formation in these models, DEF did not increase cuticle bleeding in rabbits or cause spontaneous bleeding in macaques over a 2-week study. Anticipating the desirability of a reversal agent, we also generated a human IgG that rapidly reversed DEF activity ex vivo in human plasma and in vivo in rabbits. Thus, an active site-directed FXIa-specific antibody can block thrombosis in animal models and, together with the reversal agent, may facilitate exploration of the roles of FXIa in human disease.


Subject(s)
Factor XI/physiology , Factor XIa/antagonists & inhibitors , Factor XIa/immunology , Hemostasis/physiology , Immunoglobulin G/metabolism , Thrombosis/blood , Animals , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/metabolism , Antibody Specificity , Humans , In Vitro Techniques , Kinetics , Macaca fascicularis , Mice , Rabbits
9.
J Immunol ; 197(1): 42-50, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27226090

ABSTRACT

Type I IFNs play a critical role in the immune response to viral infection and may also drive autoimmunity through modulation of monocyte maturation and promotion of autoreactive lymphocyte survival. Recent demonstrations of type I IFN gene signatures in autoimmune diseases, including scleroderma, led us to investigate the pathological role of IFNs in a preclinical model of sclerodermatous graft-versus-host disease. Using a neutralizing Ab against the type I IFN receptor IFNAR1, we observed a marked reduction in dermal inflammation, vasculopathy, and fibrosis compared with that seen in the presence of intact IFNAR1 signaling. The ameliorative effects of IFNAR1 blockade were restricted to the skin and were highly associated with inhibition of chronic vascular injury responses and not due to the inhibition of the T or B cell alloresponse. Inhibition of IFNAR1 normalized the overexpression of IFN-inducible genes in graft-versus-host disease skin and markedly reduced dermal IFN-α levels. Depletion of plasmacytoid dendritic cells, a major cellular source of type I IFNs, did not reduce the severity of fibrosis or type I IFN gene signature in the skin. Taken together, these studies demonstrate an important role for type I IFN in skin fibrosis, and they provide a rationale for IFNAR1 inhibition in scleroderma.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Inflammation/immunology , Interferon-alpha/metabolism , Scleroderma, Systemic/immunology , Skin/pathology , Vascular Diseases/immunology , Animals , Antibodies, Blocking/administration & dosage , Autoantibodies/blood , Disease Models, Animal , Female , Fibrosis , Gene Expression Regulation , Humans , Interferon-alpha/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Interferon alpha-beta/immunology , Signal Transduction
10.
Arthritis Rheumatol ; 68(2): 473-83, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26414805

ABSTRACT

OBJECTIVE: Systemic sclerosis (SSc) is a fibrotic disease characterized by an obliterative vasculopathy with thrombosis and impairment of the coagulation-fibrinolysis balance. Plasminogen activator inhibitor 1 (PAI-1) is the major inhibitor of profibrinolytic plasminogen activators (PAs). This study was undertaken to evaluate the contribution of PAI-1 to SSc pathology in the skin. METHODS: PAI-1 was evaluated in skin from patients with diffuse SSc (dSSc) and those with limited SSc (lSSc) by immunohistochemistry. The contribution of PAI-1 to SSc pathology was tested in vivo in murine graft-versus-host disease (GVHD) and bleomycin models of progressive skin fibrosis and in vitro in dermal human microvascular endothelial cells (HMVECs) using a monoclonal antibody that selectively prevents the binding of PAI-1 to PA. RESULTS: Skin from patients with dSSc and those with lSSc showed increased PAI-1 levels in the epidermis and microvessel endothelium. PAI-1 neutralization in the GVHD model led to a dramatic, dose-dependent improvement in clinical skin score, concomitant with vasculopathy resolution, including a reduction in fibrinolysis regulators and vascular injury markers, as well as reduced inflammation. Resolution of vasculopathy and inflammation was associated with resolution of skin fibrosis, as assessed by reduction in collagen content and expression of key profibrotic mediators, including transforming growth factor ß1 and tissue inhibitor of metalloproteinases 1. Similar to the GVHD model, PAI-1 neutralization reduced dermal inflammation and fibrosis in the bleomycin model. PAI-1 neutralization stimulated plasmin-mediated metalloproteinase 1 activation in dermal HMVECs. CONCLUSION: Our findings indicate that neutralization of the antifibrinolytic function of PAI-1 resolves skin fibrosis by limiting the extent of initial vascular injury and connective tissue inflammation. These data suggest that PAI-1 represents an important checkpoint in disease pathology in human SSc.


Subject(s)
Endothelial Cells/metabolism , Graft vs Host Disease/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Scleroderma, Diffuse/metabolism , Scleroderma, Limited/metabolism , Skin/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bleomycin/toxicity , Case-Control Studies , Disease Models, Animal , Endothelial Cells/drug effects , Fibrosis/chemically induced , Fibrosis/metabolism , Humans , Immunohistochemistry , Matrix Metalloproteinase 1/metabolism , Mice , Plasminogen Activators/metabolism , Real-Time Polymerase Chain Reaction , Skin/drug effects , Skin/pathology
11.
J Biol Chem ; 291(5): 2435-43, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26631728

ABSTRACT

The Wnt family of secreted glycolipoproteins plays pivotal roles in development and human diseases. Tiki family proteins were identified as novel Wnt inhibitors that act by cleaving the Wnt amino-terminal region to inactivate specific Wnt ligands. Tiki represents a new metalloprotease family that is dependent on Mn(2+)/Co(2+) but lacks known metalloprotease motifs. The Tiki extracellular domain shares homology with bacterial TraB/PrgY proteins, known for their roles in the inhibition of mating pheromones. The TIKI/TraB fold is predicted to be distantly related to structures of additional bacterial proteins and may use a core ß-sheet within an α+ß-fold to coordinate conserved residues for catalysis. In this study, using assays for Wnt3a cleavage and signaling inhibition, we performed mutagenesis analyses of human TIKI2 to examine the structural prediction and identify the active site residues. We also established an in vitro assay for TIKI2 protease activity using FRET peptide substrates derived from the cleavage motifs of Wnt3a and Xenopus wnt8 (Xwnt8). We further identified two pairs of potential disulfide bonds that reside outside the ß-sheet catalytic core but likely assist the folding of the TIKI domain. Finally, we systematically analyzed TIKI2 cleavage of the 19 human WNT proteins, of which we identified 10 as potential TIKI2 substrates, revealing the hydrophobic nature of Tiki cleavage sites. Our study provides insights into the Tiki family of proteases and its Wnt substrates.


Subject(s)
Gene Expression Regulation, Enzymologic , Metalloendopeptidases/chemistry , Wnt Proteins/chemistry , Amino Acid Motifs , Animals , Catalytic Domain , Cysteine/chemistry , Disulfides/chemistry , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Luciferases/metabolism , Membrane Proteins/chemistry , Metalloproteases/chemistry , Mutagenesis, Site-Directed , Peptides/chemistry , Pheromones, Human/metabolism , Protein Folding , Protein Structure, Secondary , Signal Transduction , Wnt3A Protein/chemistry , Xenopus
12.
Nat Struct Mol Biol ; 22(12): 983-90, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26551075

ABSTRACT

Aberrant activation of the EGF receptor (EGFR) contributes to many human cancers by activating the Ras-MAPK pathway and other pathways. EGFR signaling is augmented by Src-family kinases, but the mechanism is poorly understood. Here, we show that human EGFR preferentially phosphorylates peptide substrates that are primed by a prior phosphorylation. Using peptides based on the sequence of the adaptor protein Shc1, we show that Src mediates the priming phosphorylation, thus promoting subsequent phosphorylation by EGFR. Importantly, the doubly phosphorylated Shc1 peptide binds more tightly than singly phosphorylated peptide to the Ras activator Grb2; this binding is a key step in activating the Ras-MAPK pathway. Finally, a crystal structure of EGFR in complex with a primed Shc1 peptide reveals the structural basis for EGFR substrate specificity. These results provide a molecular explanation for the integration of Src and EGFR signaling with downstream effectors such as Ras.


Subject(s)
ErbB Receptors/drug effects , ErbB Receptors/metabolism , Peptides/metabolism , Phosphotyrosine/metabolism , Shc Signaling Adaptor Proteins/metabolism , Crystallography, X-Ray , ErbB Receptors/chemistry , GRB2 Adaptor Protein/metabolism , Humans , Phosphorylation , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Sensitivity and Specificity , Shc Signaling Adaptor Proteins/chemistry , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Substrate Specificity
13.
Mol Cancer Ther ; 14(8): 1868-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26089370

ABSTRACT

Antibody-drug conjugates (ADC) represent a promising therapeutic modality for managing cancer. Here, we report a novel humanized ADC that targets the tetraspanin-like protein TM4SF1. TM4SF1 is highly expressed on the plasma membranes of many human cancer cells and also on the endothelial cells lining tumor blood vessels. TM4SF1 is internalized upon interaction with antibodies. We hypothesized that an ADC against TM4SF1 would inhibit cancer growth directly by killing cancer cells and indirectly by attacking the tumor vasculature. We generated a humanized anti-human TM4SF1 monoclonal antibody, v1.10, and armed it with an auristatin cytotoxic agent LP2 (chemical name mc-3377). v1.10-LP2 selectively killed cultured human tumor cell lines and human endothelial cells that express TM4SF1. Acting as a single agent, v1.10-LP2 induced complete regression of several TM4SF1-expressing tumor xenografts in nude mice, including non-small cell lung cancer and pancreas, prostate, and colon cancers. As v1.10 did not react with mouse TM4SF1, it could not target the mouse tumor vasculature. Therefore, we generated a surrogate anti-mouse TM4SF1 antibody, 2A7A, and conjugated it to LP2. At 3 mpk, 2A7A-LP2 regressed several tumor xenografts without noticeable toxicity. Combination therapy with v1.10-LP2 and 2A7A-LP2 together was more effective than either ADC alone. These data provide proof-of-concept that TM4SF1-targeting ADCs have potential as anticancer agents with dual action against tumor cells and the tumor vasculature. Such agents could offer exceptional therapeutic value and warrant further investigation. Mol Cancer Ther; 14(8); 1868-76. ©2015 AACR.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Angiogenesis Inhibitors/toxicity , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antineoplastic Agents/toxicity , Cell Line, Tumor , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression , Humans , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic , Rabbits , Tissue Distribution , Xenograft Model Antitumor Assays
14.
PLoS One ; 9(3): e92608, 2014.
Article in English | MEDLINE | ID: mdl-24658703

ABSTRACT

Brown adipose tissue (BAT) plays a pivotal role in promoting energy expenditure by the virtue of uncoupling protein-1 (UCP-1) that differentiates BAT from its energy storing white adipose tissue (WAT) counterpart. The clinical implication of "classical" BAT (originates from Myf5 positive myoblastic lineage) or the "beige" fat (originates through trans-differentiation of WAT) activation in improving metabolic parameters is now becoming apparent. However, the inducers and endogenous molecular determinants that govern the lineage commitment and differentiation of classical BAT remain obscure. We report here that in the absence of any forced gene expression, stimulation with bone morphogenetic protein 6 (BMP6) induces brown fat differentiation from skeletal muscle precursor cells of murine and human origins. Through a comprehensive transcriptional profiling approach, we have discovered that two days of BMP6 stimulation in C2C12 myoblast cells is sufficient to induce genes characteristic of brown preadipocytes. This developmental switch is modulated in part by newly identified regulators, Optineurin (Optn) and Cyclooxygenase-2 (Cox2). Furthermore, pathway analyses using the Causal Reasoning Engine (CRE) identified additional potential causal drivers of this BMP6 induced commitment switch. Subsequent analyses to decipher key pathway that facilitates terminal differentiation of these BMP6 primed cells identified a key role for Insulin Like Growth Factor-1 Receptor (IGF-1R). Collectively these data highlight a therapeutically innovative role for BMP6 by providing a means to enhance the amount of myogenic lineage derived brown fat.


Subject(s)
Adipose Tissue, Brown/metabolism , Bone Morphogenetic Protein 6/metabolism , Myoblasts/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Line , Cluster Analysis , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Fatty Acids/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Ion Channels/genetics , Ion Channels/metabolism , Membrane Transport Proteins , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Phenotype , Receptor, IGF Type 1 , Signal Transduction , Transcription Factor TFIIIA/genetics , Transcription Factor TFIIIA/metabolism , Uncoupling Protein 1
15.
PLoS One ; 9(2): e88714, 2014.
Article in English | MEDLINE | ID: mdl-24551140

ABSTRACT

Allergic asthma is a chronic immune-inflammatory disease of the airways. Despite aeroallergen exposure being universal, allergic asthma affects only a fraction of individuals. This is likely related, at least in part, to the extent of allergen exposure. Regarding house dust mite (HDM), we previously identified the threshold required to elicit allergic responses in BALB/c mice. Here, we investigated the impact of an initial immune perturbation on the response to sub-threshold HDM exposure. We show that transient GM-CSF expression in the lung facilitated robust eosinophilic inflammation, long-lasting antigen-specific Th2 responses, mucus production and airway hyperresponsiveness. This was associated with increased IL-33 levels and activated CD11b(+) DCs expressing OX40L. GM-CSF-driven allergic responses were significantly blunted in IL-33-deficient mice. IL-33 was localized on alveolar type II cells and in vitro stimulation of human epithelial cells with GM-CSF enhanced intracellular IL-33 independently of IL-1α. Likewise, GM-CSF administration in vivo resulted in increased levels of IL-33 but not IL-1α. These findings suggest that exposures to environmental agents associated with GM-CSF production, including airway infections and pollutants, may decrease the threshold of allergen responsiveness and, hence, increase the susceptibility to develop allergic asthma through a GM-CSF/IL-33/OX40L pathway.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hypersensitivity/immunology , Hypersensitivity/parasitology , Interleukins/metabolism , Lung/immunology , Lung/parasitology , Pyroglyphidae/immunology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/pathology , Animals , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunity/drug effects , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Interleukin-1alpha/metabolism , Interleukin-33 , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Mice, Inbred BALB C , Models, Immunological , Pyroglyphidae/drug effects , Time Factors
16.
J Exp Med ; 210(11): 2447-63, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24081950

ABSTRACT

Recognition of DNA and RNA molecules derived from pathogens or self-antigen is one way the mammalian immune system senses infection and tissue damage. Activation of immune signaling receptors by nucleic acids is controlled by limiting the access of DNA and RNA to intracellular receptors, but the mechanisms by which endosome-resident receptors encounter nucleic acids from the extracellular space are largely undefined. In this study, we show that the receptor for advanced glycation end-products (RAGE) promoted DNA uptake into endosomes and lowered the immune recognition threshold for the activation of Toll-like receptor 9, the principal DNA-recognizing transmembrane signaling receptor. Structural analysis of RAGE-DNA complexes indicated that DNA interacted with dimers of the outermost RAGE extracellular domains, and could induce formation of higher-order receptor complexes. Furthermore, mice deficient in RAGE were unable to mount a typical inflammatory response to DNA in the lung, indicating that RAGE is important for the detection of nucleic acids in vivo.


Subject(s)
DNA/metabolism , Pneumonia/metabolism , Pneumonia/pathology , Receptors, Immunologic/metabolism , Animals , Base Sequence , Cell Membrane/metabolism , Crystallography, X-Ray , DNA/chemistry , Endocytosis , Endosomes/metabolism , HEK293 Cells , HeLa Cells , Humans , Ligands , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , NF-kappa B/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Receptor for Advanced Glycation End Products , Receptors, Immunologic/chemistry , Static Electricity , Toll-Like Receptor 9/metabolism
17.
J Allergy Clin Immunol ; 131(1): 187-200.e1-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23006545

ABSTRACT

BACKGROUND: Allergen exposure at lung and gut mucosae can lead to aberrant T(H)2 immunity and allergic disease. The epithelium-associated cytokines thymic stromal lymphopoietin (TSLP), IL-25, and IL-33 are suggested to be important for the initiation of these responses. OBJECTIVE: We sought to investigate the contributions of TSLP, IL-25, and IL-33 in the development of allergic disease to the common allergens house dust mite (HDM) or peanut. METHODS: Neutralizing antibodies or mice deficient in TSLP, IL-25, or IL-33 signaling were exposed to HDM intranasally or peanut intragastrically, and immune inflammatory and physiologic responses were evaluated. In vitro assays were performed to examine specific dendritic cell (DC) functions. RESULTS: We showed that experimental HDM-induced allergic asthma and food allergy and anaphylaxis to peanut were associated with TSLP production but developed independently of TSLP, likely because these allergens functionally mimicked TSLP inhibition of IL-12 production and induction of OX40 ligand (OX40L) on DCs. Blockade of OX40L significantly lessened allergic responses to HDM or peanut. Although IL-25 and IL-33 induced OX40L on DCs in vitro, only IL-33 signaling was necessary for intact allergic immunity, likely because of its superior ability to induce DC OX40L and expand innate lymphoid cells in vivo. CONCLUSION: These data identify a nonredundant, IL-33-driven mechanism initiating T(H)2 responses to the clinically relevant allergens HDM and peanut. Our findings, along with those in infectious and transgenic/surrogate allergen systems, favor a paradigm whereby multiple molecular pathways can initiate T(H)2 immunity, which has implications for the conceptualization and manipulation of these responses in health and disease.


Subject(s)
Allergens/immunology , Arachis/immunology , Hypersensitivity/immunology , Interleukins/immunology , Pyroglyphidae/immunology , Thymus Gland/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gastrointestinal Tract/immunology , Humans , Hypersensitivity/metabolism , Interleukin-33 , Interleukin-4/immunology , Interleukin-4/metabolism , Lung/immunology , Lung/metabolism , Mice , OX40 Ligand/immunology , OX40 Ligand/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction , Stromal Cells/immunology , Stromal Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Thymus Gland/cytology
18.
Immunity ; 37(6): 986-997, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23219390

ABSTRACT

Toll-like receptor-9 (TLR9) is largely responsible for discriminating self from pathogenic DNA. However, association of host DNA with autoantibodies activates TLR9, inducing the pathogenic secretion of type I interferons (IFNs) from plasmacytoid dendritic cells (pDCs). Here, we found that in response to DNA-containing immune complexes (DNA-IC), but not to soluble ligands, IFN-α production depended upon the convergence of the phagocytic and autophagic pathways, a process called microtubule-associated protein 1A/1B-light chain 3 (LC3)-associated phagocytosis (LAP). LAP was required for TLR9 trafficking into a specialized interferon signaling compartment by a mechanism that involved autophagy-related proteins, but not the conventional autophagic preinitiation complex, or adaptor protein-3 (AP-3). Our findings unveil a new role for nonconventional autophagy in inflammation and provide one mechanism by which anti-DNA autoantibodies, such as those found in several autoimmune disorders, bypass the controls that normally restrict the apportionment of pathogenic DNA and TLR9 to the interferon signaling compartment.


Subject(s)
Antigen-Antibody Complex/immunology , Autophagy/immunology , DNA/immunology , Interferon Type I/biosynthesis , Animals , Humans , Immunoglobulin G/immunology , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Phagocytosis/immunology , Phagosomes/metabolism , Protein Transport , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
19.
J Clin Invest ; 122(11): 3965-76, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064362

ABSTRACT

Psoriasis is a chronic inflammatory disorder of the skin affecting approximately 2% of the world's population. Accumulating evidence has revealed that the IL-23/IL-17/IL-22 pathway is key for development of skin immunopathology. However, the role of keratinocytes and their crosstalk with immune cells at the onset of disease remains poorly understood. Here, we show that IL-36R-deficient (Il36r-/-) mice were protected from imiquimod-induced expansion of dermal IL-17-producing γδ T cells and psoriasiform dermatitis. Furthermore, IL-36R antagonist-deficient (Il36rn-/-) mice showed exacerbated pathology. TLR7 ligation on DCs induced IL-36-mediated crosstalk with keratinocytes and dermal mesenchymal cells that was crucial for control of the pathological IL-23/IL-17/IL-22 axis and disease development. Notably, mice lacking IL-23, IL-17, or IL-22 were less well protected from disease compared with Il36r-/- mice, indicating an additional distinct activity of IL-36 beyond induction of the pathological IL-23 axis. Moreover, while the absence of IL-1R1 prevented neutrophil infiltration, it did not protect from acanthosis and hyperkeratosis, demonstrating that neutrophils are dispensable for disease manifestation. These results highlight a central and unique IL-1-independent role for IL-36 in control of the IL-23/IL-17/IL-22 pathway and development of psoriasiform dermatitis.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Dermatitis/immunology , Interleukin-1/immunology , Keratinocytes/immunology , Psoriasis/immunology , Animals , Cell Communication/genetics , Dendritic Cells/metabolism , Dendritic Cells/pathology , Dermatitis/genetics , Dermatitis/metabolism , Dermatitis/pathology , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-23/genetics , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Psoriasis/genetics , Psoriasis/metabolism , Psoriasis/pathology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Interleukin-22
20.
Ann Rheum Dis ; 71(8): 1402-10, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22730375

ABSTRACT

BACKGROUND: Angiopoietin (Ang)-1 and Ang-2, and their shared receptor Tie2, are expressed in rheumatoid arthritis (RA) synovial tissue, but the cellular targets of Ang signalling and the relative contributions of Ang-1 and Ang-2 to arthritis are poorly understood. OBJECTIVES: To determine the cellular targets of Ang signalling in RA synovial tissue, and the effects of Ang-2 neutralisation in murine collagen-induced arthritis (CIA). METHODS: RA and psoriatic arthritis (PsA) synovial biopsies were examined for expression of Tie2 and activated phospho (p)-Tie2 by quantitative immunohistochemistry and immunofluorescent double staining. Human monocyte and macrophage Tie2 expression was determined by flow cytometry and quantitative PCR. Regulation of macrophage intracellular signalling pathways and gene expression were examined by immunoblotting and ELISA. CIA was assessed in mice treated with saline, control antibody, prednisolone or neutralising anti-Ang-2 antibody. RESULTS: Expression of synovial Tie2 and p-Tie2 was similar in RA and PsA. Tie2 activation in RA patient synovial tissue was predominantly localised in synovial macrophages and was expressed by human macrophage. Ang-1 and Ang-2 stimulated activation of multiple intracellular signalling pathways, and cooperated with tumour necrosis factor to induce macrophage interleukin 6 and macrophage inflammatory protein 1α production. Ang-2 selectively suppressed macrophage thrombospondin-2 production. Ang-2 neutralisation significantly decreased disease severity, synovial inflammation, neo-vascularisation and joint destruction in established CIA. CONCLUSIONS: The authors identify synovial macrophages as primary targets of Ang signalling in RA, and demonstrate that Ang-2 promotes the pro-inflammatory activation of human macrophages. Ang-2 makes requisite contributions to pathology in CIA, indicating that targeting Ang-2 may be of therapeutic benefit in the treatment of RA.


Subject(s)
Angiopoietin-2/pharmacology , Arthritis, Experimental/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Angiopoietin-1/metabolism , Angiopoietin-1/pharmacology , Angiopoietin-2/immunology , Angiopoietin-2/metabolism , Animals , Antibodies, Blocking/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Psoriatic/metabolism , Arthritis, Psoriatic/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Chemokine CCL3/biosynthesis , Gene Expression/drug effects , Humans , Interleukin-6/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred DBA , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Phosphorylation , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Signal Transduction/drug effects , Signal Transduction/genetics , Synovial Membrane/metabolism , Synovial Membrane/pathology , Thrombospondins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...