Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Res Sq ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38853999

ABSTRACT

Human lung cancer carries high genetic alterations, expressing high tumor-specific neoantigens. Although orthotopic murine lung cancer models recapitulate many characteristics of human lung cancers, genetically engineered mouse models have fewer somatic mutations than human lung cancer, resulting in scarce immune cell infiltration and deficient immune responses. The endogenous mouse lung cancer model driven by Kras mutation and Trp53 deletion (KP model) has minimal immune infiltration because of a scarcity of neoantigens. Fine-tuning tumor antigenicity to trigger the appropriate level of antitumor immunity would be key to investigating immune responses against human lung cancer. We engineered the KP model to express antigens of OVA peptides (minOVA) as neoantigens along with ZsGreen, a traceable fluorescent conjugate. The KP model expressing minOVA exhibited stronger immunogenicity with higher immune cell infiltration comprised of CD8+ T cells and CD11c+ dendritic cells (DCs). Consequentially, the KP model expressing minOVA exhibits suppressed tumor growth compared to its origin. We further analyzed tumor-infiltrated DCs. The majority of ZsGreen conjugated with minOVA was observed in the conventional type 2 DCs (cDC2), where cDC1 has minimal. These data indicate that tumor immunogenicity regulates host immune responses, and tumor neoantigen is mostly recognized by cDC2 cells, which may play a critical role in initiating anti-tumor immune responses in an orthotopic murine lung cancer model.

2.
J Allergy Clin Immunol ; 152(5): 1141-1152.e2, 2023 11.
Article in English | MEDLINE | ID: mdl-37562753

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are heterogeneous, comprising multiple subsets with unique functional specifications. Our previous work has demonstrated that the specific conventional type 2 DC subset, CSF1R+cDC2s, plays a critical role in sensing aeroallergens. OBJECTIVE: It remains to be understood how CSF1R+cDC2s recognize inhaled allergens. We sought to elucidate the transcriptomic programs and receptor-ligand interactions essential for function of this subset in allergen sensitization. METHODS: We applied single-cell RNA sequencing to mouse lung DCs. Conventional DC-selective knockout mouse models were employed, and mice were subjected to inhaled allergen sensitization with multiple readouts of asthma pathology. Under the clinical arm of this work, human lung transcriptomic data were integrated with mouse data, and bronchoalveolar lavage (BAL) specimens were collected from subjects undergoing allergen provocation, with samples assayed for C1q. RESULTS: We found that C1q is selectively enriched in lung CSF1R+cDC2s, but not in other lung cDC2 or cDC1 subsets. Depletion of C1q in conventional DCs significantly attenuates allergen sensing and features of asthma. Additionally, we found that C1q binds directly to human dust mite allergen, and the C1q receptor CD91 (LRP1) is required for lung CSF1R+cDC2s to recognize the C1q-allergen complex and induce allergic lung inflammation. Lastly, C1q is enriched in human BAL samples following subsegmental allergen challenge, and human RNA sequencing data demonstrate close homology between lung IGSF21+DCs and mouse CSF1R+cDC2s. CONCLUSIONS: C1q is secreted from the CSF1R+cDC2 subset among conventional DCs. Our data indicate that the C1q-LRP1 axis represents a candidate for translational therapeutics in the prevention and suppression of allergic lung inflammation.


Subject(s)
Asthma , Pneumonia , Animals , Humans , Mice , Allergens/metabolism , Asthma/metabolism , Complement C1q/metabolism , Dendritic Cells , Mice, Knockout , Pneumonia/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Colony-Stimulating Factor/metabolism
3.
ACS Omega ; 8(23): 20505-20512, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37323402

ABSTRACT

The colony-stimulating factor-1 receptor (CSF1R) is a tyrosine-protein kinase that is a potential target for asthma therapeutics. We have applied a fragment-lead combination approach to identify small fragments that act synergistically with GW2580, a known inhibitor of CSF1R. Two fragment libraries were screened in combination with GW2580 by surface plasmon resonance (SPR). Binding affinity measurements confirmed that thirteen fragments bind specifically to the CSF1R, and a kinase activity assay further validated the inhibitory effect of these fragments. Several fragment compounds enhanced the inhibitory activity of the lead inhibitor. Computational solvent mapping, molecular docking, and modeling studies suggest that some of these fragments bind adjacent to the binding site of the lead inhibitor and further stabilize the inhibitor-bound state. Modeling results guided the computational fragment-linking approach to design potential next-generation compounds. The inhalability of these proposed compounds was predicted using quantitative structure-property relationships (QSPR) modeling based on an analysis of 71 drugs currently on the market. This work provides new insights into the development of inhalable small molecule therapeutics for asthma.

4.
Am J Respir Cell Mol Biol ; 69(1): 13-21, 2023 07.
Article in English | MEDLINE | ID: mdl-37017484

ABSTRACT

Asthma is a chronic inflammatory airway disease driven by various infiltrating immune cell types into the lung. Optical microscopy has been used to study immune infiltrates in asthmatic lungs. Confocal laser scanning microscopy (CLSM) identifies the phenotypes and locations of individual immune cells in lung tissue sections by employing high-magnification objectives and multiplex immunofluorescence staining. In contrast, light-sheet fluorescence microscopy (LSFM) can visualize the macroscopic and mesoscopic architecture of whole-mount lung tissues in three dimensions (3D) by adopting an optical tissue-clearing method. Despite each microscopy method producing image data with unique resolution from a tissue sample, CLSM and LSFM have not been applied together because of different tissue-preparation procedures. Here, we introduce a new approach combining LSFM and CLSM into a sequential imaging pipeline. We built a new optical tissue clearing workflow in which the immersion clearing agent can be switched from an organic solvent to an aqueous sugar solution for sequential 3D LSFM and CLSM of mouse lungs. This sequential combination microscopy offered quantitative 3D spatial analyses of the distribution of immune infiltrates in the same mouse asthmatic lung tissue at the organ, tissue, and cell levels. These results show that our method facilitates multiresolution 3D fluorescence microscopy as a new imaging approach providing comprehensive spatial information for a better understanding of inflammatory lung diseases.


Subject(s)
Asthma , Imaging, Three-Dimensional , Animals , Mice , Imaging, Three-Dimensional/methods , Microscopy, Fluorescence/methods , Lung/diagnostic imaging , Asthma/diagnostic imaging , Microscopy, Confocal/methods
5.
Am J Respir Crit Care Med ; 207(11): 1451-1463, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36790376

ABSTRACT

Rationale: The resolution of inflammation is an active process coordinated by mediators and immune cells to restore tissue homeostasis. However, the mechanisms for resolving eosinophilic allergic lung inflammation triggered by inhaled allergens have not been fully elucidated. Objectives: Our objectives were to investigate the cellular mechanism of tissue-resident macrophages involved in the resolution process of eosinophilic lung inflammation. Methods: For the study, we used the institutional review board-approved protocol for human subsegmental bronchoprovocation with allergen, mouse models for allergic lung inflammation, and novel transgenic mice, including a conditional CCL26 knockout. The samples were analyzed using mass cytometry, single-cell RNA sequencing, and biophysical and immunological analyses. Measurements and Main Results: We compared alveolar macrophage (AM) subsets in the BAL before and after allergen provocation. In response to provocation with inhaled allergens, the subsets of AMs are dynamically changed in humans and mice. In the steady state, the AM subset expressing CX3CR1 is a relatively small fraction in bronchoalveolar space and lung tissue but drastically increases after allergen challenges. This subset presents unique patterns of gene expression compared with classical AMs, expressing high C1q family genes. CX3CR1+ macrophages are activated by airway epithelial cell-derived CCL26 via a receptor-ligand interaction. The binding of CCL26 to the CX3CR1+ receptor induces CX3CR1+ macrophages to secrete C1q, subsequently facilitating the clearance of eosinophils. Furthermore, the depletion of CX3CR1 macrophages or CCL26 in airway epithelial cells delays the resolution of allergic lung inflammation displaying prolonged tissue eosinophilia. Conclusions: These findings indicate that the CCL26-CX3CR1 pathway is pivotal in resolving eosinophilic allergic lung inflammation.


Subject(s)
Alveolitis, Extrinsic Allergic , Hypersensitivity , Pneumonia , Pulmonary Eosinophilia , Humans , Mice , Animals , Complement C1q/metabolism , Lung/metabolism , Macrophages , Allergens , Inflammation/metabolism , Pneumonia/metabolism , Chemokine CCL26/metabolism
6.
Front Immunol ; 13: 943554, 2022.
Article in English | MEDLINE | ID: mdl-35958610

ABSTRACT

Asthma is phenotypically heterogeneous with several distinctive pathological mechanistic pathways. Previous studies indicate that neutrophilic asthma has a poor response to standard asthma treatments comprising inhaled corticosteroids. Therefore, it is important to identify critical factors that contribute to increased numbers of neutrophils in asthma patients whose symptoms are poorly controlled by conventional therapy. Leukocytes release chromatin fibers, referred to as extracellular traps (ETs) consisting of double-stranded (ds) DNA, histones, and granule contents. Excessive components of ETs contribute to the pathophysiology of asthma; however, it is unclear how ETs drive asthma phenotypes and whether they could be a potential therapeutic target. We employed a mouse model of severe asthma that recapitulates the intricate immune responses of neutrophilic and eosinophilic airway inflammation identified in patients with severe asthma. We used both a pharmacologic approach using miR-155 inhibitor-laden exosomes and genetic approaches using miR-155 knockout mice. Our data show that ETs are present in the bronchoalveolar lavage fluid of patients with mild asthma subjected to experimental subsegmental bronchoprovocation to an allergen and a severe asthma mouse model, which resembles the complex immune responses identified in severe human asthma. Furthermore, we show that miR-155 contributes to the extracellular release of dsDNA, which exacerbates allergic lung inflammation, and the inhibition of miR-155 results in therapeutic benefit in severe asthma mice. Our findings show that targeting dsDNA release represents an attractive therapeutic target for mitigating neutrophilic asthma phenotype, which is clinically refractory to standard care.


Subject(s)
Asthma , Eosinophilia , MicroRNAs , Pneumonia , Animals , Disease Models, Animal , Granulocytes , Humans , Mice , MicroRNAs/metabolism , Neutrophils , Pneumonia/drug therapy , Pneumonia/metabolism
7.
Nat Commun ; 12(1): 2736, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33980844

ABSTRACT

Endothelial barrier integrity is ensured by the stability of the adherens junction (AJ) complexes comprised of vascular endothelial (VE)-cadherin as well as accessory proteins such as ß-catenin and p120-catenin. Disruption of the endothelial barrier due to disassembly of AJs results in tissue edema and the influx of inflammatory cells. Using three-dimensional structured illumination microscopy, we observe that the mitochondrial protein Mitofusin-2 (Mfn2) co-localizes at the plasma membrane with VE-cadherin and ß-catenin in endothelial cells during homeostasis. Upon inflammatory stimulation, Mfn2 is sulfenylated, the Mfn2/ß-catenin complex disassociates from the AJs and Mfn2 accumulates in the nucleus where Mfn2 negatively regulates the transcriptional activity of ß-catenin. Endothelial-specific deletion of Mfn2 results in inflammatory activation, indicating an anti-inflammatory role of Mfn2 in vivo. Our results suggest that Mfn2 acts in a non-canonical manner to suppress the inflammatory response by stabilizing cell-cell adherens junctions and by binding to the transcriptional activator ß-catenin.


Subject(s)
Adherens Junctions/metabolism , GTP Phosphohydrolases/metabolism , Mitochondrial Proteins/metabolism , beta Catenin/metabolism , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Cell Line , Cell Membrane/metabolism , Female , Humans , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
8.
Int J Mol Sci ; 21(7)2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32244797

ABSTRACT

Human SNF5 and BAF155 constitute the core subunit of multi-protein SWI/SNF chromatin-remodeling complexes that are required for ATP-dependent nucleosome mobility and transcriptional control. Human SNF5 (hSNF5) utilizes its repeat 1 (RPT1) domain to associate with the SWIRM domain of BAF155. Here, we employed X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and various biophysical methods in order to investigate the detailed binding mechanism between hSNF5 and BAF155. Multi-angle light scattering data clearly indicate that hSNF5171-258 and BAF155SWIRM are both monomeric in solution and they form a heterodimer. NMR data and crystal structure of the hSNF5171-258/BAF155SWIRM complex further reveal a unique binding interface, which involves a coil-to-helix transition upon protein binding. The newly formed αN helix of hSNF5171-258 interacts with the ß2-α1 loop of hSNF5 via hydrogen bonds and it also displays a hydrophobic interaction with BAF155SWIRM. Therefore, the N-terminal region of hSNF5171-258 plays an important role in tumorigenesis and our data will provide a structural clue for the pathogenesis of Rhabdoid tumors and malignant melanomas that originate from mutations in the N-terminal loop region of hSNF5.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Mutation , Nucleosomes/genetics , SMARCB1 Protein/genetics , Transcription Factors/genetics , Binding Sites/genetics , Circular Dichroism , Crystallography, X-Ray , Gene Expression Regulation , Humans , Magnetic Resonance Spectroscopy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Nucleosomes/metabolism , Protein Binding , Rhabdoid Tumor/genetics , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , SMARCB1 Protein/chemistry , SMARCB1 Protein/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L921-L930, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32159972

ABSTRACT

The incidence of asthma has increased from 5.5% to near 8% of the population, which is a major health concern. The hallmarks of asthma include eosinophilic airway inflammation that is associated with chronic airway remodeling. Allergic airway inflammation is characterized by a complex interplay of resident and inflammatory cells. MicroRNAs (miRNAs) are small noncoding RNAs that function as posttranscriptional modulators of gene expression. However, the role of miRNAs, specifically miR-451, in the regulation of allergic airway inflammation is unexplored. Our previous findings showed that oxidant stress regulates miR-451 gene expression in macrophages during an inflammatory process. In this paper, we examined the role of miR-451 in regulating macrophage phenotype using an experimental poly-allergenic murine model of allergic airway inflammation. We found that miR-451 contributes to the allergic induction of CCL17 in the lung and plays a key role in proasthmatic macrophage activation. Remarkably, administration of a Sirtuin 2 (Sirt2) inhibitor diminished alternate macrophage activation and markedly abrogated triple-allergen [dust mite, ragweed, Aspergillus fumigatus (DRA)]-induced lung inflammation. These data demonstrate a role for miR-451 in modulating allergic inflammation by influencing allergen-mediated macrophages phenotype.


Subject(s)
Asthma/genetics , Macrophages, Alveolar/immunology , MicroRNAs/genetics , Pneumonia/genetics , Sirtuin 2/genetics , Allergens/administration & dosage , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Plant/administration & dosage , Aspergillus/chemistry , Aspergillus/immunology , Asthma/chemically induced , Asthma/pathology , Asthma/therapy , Chemokine CCL17/genetics , Chemokine CCL17/immunology , Disease Models, Animal , Fungi/chemistry , Fungi/immunology , Furans/pharmacology , Gene Expression Regulation , Lung/drug effects , Lung/immunology , Lung/pathology , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/immunology , Plant Extracts/administration & dosage , Pneumonia/chemically induced , Pneumonia/pathology , Pneumonia/therapy , Pyroglyphidae/chemistry , Pyroglyphidae/immunology , Quinolines/pharmacology , Signal Transduction , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/immunology
10.
Article in English | MEDLINE | ID: mdl-32004685

ABSTRACT

Lysophosphatidic acid (LPA) species are present in almost all organ systems and play diverse roles through its receptors. Asthma is an airway disease characterized by chronic allergic inflammation where various innate and adaptive immune cells participate in establishing Th2 immune response. Here, we will review the contribution of LPA and its receptors to the functions of immune cells that play a key role in establishing allergic airway inflammation and aggravation of allergic asthma.


Subject(s)
Asthma/immunology , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/immunology , Animals , Asthma/blood , Asthma/genetics , Asthma/pathology , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Gene Knock-In Techniques , Humans , Lung/blood supply , Lung/cytology , Lung/immunology , Lung/pathology , Lymph Nodes/blood supply , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Knockout , Phosphoric Diester Hydrolases/genetics , Receptors, Lysophosphatidic Acid/genetics , Th2 Cells/immunology , Th2 Cells/metabolism
11.
Allergy ; 75(2): 357-369, 2020 02.
Article in English | MEDLINE | ID: mdl-31385613

ABSTRACT

BACKGROUND: A new approach targeting aeroallergen sensing in the early events of mucosal immunity could have greater benefit. The CSF1-CSF1R pathway has a critical role in trafficking allergens to regional lymph nodes through activating dendritic cells. Intervention in this pathway could prevent allergen sensitization and subsequent Th2 allergic inflammation. OBJECTIVE: To examine the therapeutic effectiveness of CSF1 and CSF1R inhibition for blocking the dendritic cell function of sensing aeroallergens. METHODS: We adopted a model of chronic asthma induced by a panel of three naturally occurring allergens and novel delivery system of CSF1R inhibitor encapsulated nanoprobe. RESULTS: Selective depletion of CSF1 in airway epithelial cells abolished the production of allergen-reactive IgE, resulting in prevention of new asthma development as well as reversal of established allergic lung inflammation. CDPL-GW nanoprobe containing GW2580, a selective CSF1R inhibitor, showed favorable pharmacokinetics for inhalational treatment and intranasal insufflation delivery of CDPL-GW nanoprobe ameliorated asthma pathologies including allergen-specific serum IgE production, allergic lung and airway inflammation and airway hyper-responsiveness (AHR) with minimal pulmonary adverse reaction. CONCLUSION: The inhibition of the CSF1-CSF1R signaling pathway effectively suppresses sensitization to aeroallergens and consequent allergic lung inflammation in a murine model of chronic asthma. CSF1R inhibition is a promising new target for the treatment of allergic asthma.


Subject(s)
Anisoles/administration & dosage , Anisoles/pharmacology , Asthma/drug therapy , Drug Delivery Systems/methods , Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/metabolism , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Allergens/immunology , Allergens/pharmacology , Animals , Asthma/chemically induced , Disease Models, Animal , Female , Immunoglobulin E/biosynthesis , Macrophage Colony-Stimulating Factor/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nanostructures/administration & dosage , Quaternary Ammonium Compounds/administration & dosage , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Sulfonic Acids/administration & dosage , Treatment Outcome
12.
JCI Insight ; 4(4)2019 02 21.
Article in English | MEDLINE | ID: mdl-30668546

ABSTRACT

Allergic eosinophilic asthma is a chronic condition causing airway remodeling resulting in lung dysfunction. We observed that expression of sirtuin 2 (Sirt2), a histone deacetylase, regulates the recruitment of eosinophils after sensitization and challenge with a triple antigen: dust mite, ragweed, and Aspergillus fumigatus (DRA). Our data demonstrate that IL-4 regulates the expression of Sirt2 isoform 3/5. Pharmacological inhibition of Sirt2 by AGK2 resulted in diminished cellular recruitment, decreased CCL17/TARC, and reduced goblet cell hyperplasia. YM1 and Fizz1 expression was reduced in AGK2-treated, IL-4-stimulated lung macrophages in vitro as well as in lung macrophages from AGK2-DRA-challenged mice. Conversely, overexpression of Sirt2 resulted in increased cellular recruitment, CCL17 production, and goblet cell hyperplasia following DRA challenge. Sirt2 isoform 3/5 was upregulated in primary human alveolar macrophages following IL-4 and AGK2 treatment, which resulted in reduced CCL17 and markers of alternative activation. These gain-of-function and loss-of-function studies indicate that Sirt2 could be developed as a treatment for eosinophilic asthma.


Subject(s)
Asthma/immunology , Eosinophils/immunology , Interleukin-4/immunology , Sirtuin 2/immunology , Adoptive Transfer , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/diagnosis , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Chemokine CCL17/immunology , Chemokine CCL17/metabolism , Disease Models, Animal , Female , Furans/pharmacology , Goblet Cells/immunology , Goblet Cells/pathology , Humans , Interleukin-4/metabolism , Lung/cytology , Lung/immunology , Lung/pathology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/transplantation , Male , Mice , Mice, Knockout , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Quinolines/pharmacology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/genetics , Sirtuin 2/metabolism
13.
J Allergy Clin Immunol ; 143(2): 669-680.e12, 2019 02.
Article in English | MEDLINE | ID: mdl-29778505

ABSTRACT

BACKGROUND: Chemokine signaling through CCR3 is a key regulatory pathway for eosinophil recruitment into tissues associated with allergic inflammation and asthma. To date, none of the CCR3 antagonists have shown efficacy in clinical trials. One reason might be their unbiased mode of inhibition that prevents receptor internalization, leading to drug tolerance. OBJECTIVE: We sought to develop a novel peptide nanoparticle CCR3 inhibitor (R321) with a biased mode of inhibition that would block G protein signaling but enable or promote receptor internalization. METHODS: Self-assembly of R321 peptide into nanoparticles and peptide binding to CCR3 were analyzed by means of dynamic light scattering and nuclear magnetic resonance. Inhibitory activity on CCR3 signaling was assessed in vitro by using flow cytometry, confocal microscopy, and Western blot analysis in a CCR3+ eosinophil cell line and blood eosinophils. In vivo effects of R321 were assessed by using a triple-allergen mouse asthma model. RESULTS: R321 self-assembles into nanoparticles and binds directly to CCR3, altering receptor function. Half-maximal inhibitory concentration values for eotaxin-induced chemotaxis of blood eosinophils are in the low nanomolar range. R321 inhibits only the early phase of extracellular signal-regulated kinase 1/2 activation and not the late phase generally associated with ß-arrestin recruitment and receptor endocytosis, promoting CCR3 internalization and degradation. In vivo R321 effectively blocks eosinophil recruitment into the blood, lungs, and airways and prevents airway hyperresponsiveness in a mouse eosinophilic asthma model. CONCLUSIONS: R321 is a potent and selective antagonist of the CCR3 signaling cascade. Inhibition through a biased mode of antagonism might hold significant therapeutic promise by eluding the formation of drug tolerance.


Subject(s)
Eosinophils/immunology , Hypersensitivity/drug therapy , Lung/immunology , Nanoparticles/therapeutic use , Peptides/therapeutic use , Receptors, CCR3/antagonists & inhibitors , Respiratory Hypersensitivity/drug therapy , Allergens/immunology , Cell Line , Cell Movement , GTP-Binding Proteins/antagonists & inhibitors , Humans , Magnetic Resonance Spectroscopy , Protein Binding , Signal Transduction
14.
Immunity ; 49(2): 275-287.e5, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30054206

ABSTRACT

Airway epithelial cells (AECs) secrete innate immune cytokines that regulate adaptive immune effector cells. In allergen-sensitized humans and mice, the airway and alveolar microenvironment is enriched with colony stimulating factor-1 (CSF1) in response to allergen exposure. In this study we found that AEC-derived CSF1 had a critical role in the production of allergen reactive-IgE production. Furthermore, spatiotemporally secreted CSF1 regulated the recruitment of alveolar dendritic cells (DCs) and enhanced the migration of conventional DC2s (cDC2s) to the draining lymph node in an interferon regulatory factor 4 (IRF4)-dependent manner. CSF1 selectively upregulated the expression of the chemokine receptor CCR7 on the CSF1R+ cDC2, but not the cDC1, population in response to allergen stimuli. Our data describe the functional specification of CSF1-dependent DC subsets that link the innate and adaptive immune responses in T helper 2 (Th2) cell-mediated allergic lung inflammation.


Subject(s)
Allergens/immunology , Dendritic Cells/immunology , Macrophage Colony-Stimulating Factor/immunology , Receptors, CCR7/biosynthesis , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Animals , Cell Line , Cell Movement/immunology , Dendritic Cells/classification , Epithelial Cells/cytology , Epithelial Cells/immunology , Humans , Immunity, Innate/immunology , Immunoglobulin E/immunology , Interferon Regulatory Factors/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RAW 264.7 Cells , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Th2 Cells/immunology , Up-Regulation/immunology
15.
Oncotarget ; 9(12): 10606-10620, 2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29535830

ABSTRACT

Specific therapies targeting cellular and molecular events of sepsis induced Acute Lung Injury (ALI) pathogenesis are lacking. We have reported a pivotal role for Nuclear Factors of Activated T cells (NFATc3) in regulating macrophage phenotype during sepsis induced ALI and subsequent studies demonstrate that NFATc3 transcriptionally regulates macrophage CCR2 and TNFα gene expression. Mouse pulmonary microvascular endothelial cell monolayer maintained a tighter barrier function when co-cultured with LPS stimulated NFATc3 deficient macrophages whereas wild type macrophages caused leaky monolayer barrier. More importantly, NFATc3 deficient mice showed decreased neutrophilic lung inflammation, improved alveolar capillary barrier function, arterial oxygen saturation and survival benefit in lethal CLP sepsis mouse models. In addition, survival of wild type mice subjected to the lethal CLP sepsis was not improved with broad-spectrum antibiotics, whereas the survival of NFATc3 deficient mice was improved to 40-60% when treated with imipenem. Passive adoptive transfer of NFATc3 deficient macrophages conferred protection against LPS induced ALI in wild type mice. Furthermore, CP9-ZIZIT, a highly potent, cell-permeable peptide inhibitor of Calcineurin inhibited NFATc3 activation. CP9-ZIZIT effectively reduced sepsis induced inflammatory cytokines and pulmonary edema in mice. Thus, this study demonstrates that inhibition of NFATc3 activation by CP9-ZIZIT provides a potential therapeutic option for attenuating sepsis induced ALI/pulmonary edema.

17.
Oncotarget ; 7(14): 17532-46, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27007158

ABSTRACT

Inflammatory monocyte and tissue macrophages influence the initiation, progression, and resolution of type 2 immune responses, and alveolar macrophages are the most prevalent immune-effector cells in the lung. While we were characterizing the M1- or M2-like macrophages in type 2 allergic inflammation, we discovered that FoxO1 is highly expressed in alternatively activated macrophages. Although several studies have been focused on the fundamental role of FoxOs in hematopoietic and immune cells, the exact role that FoxO1 plays in allergic asthmatic inflammation in activated macrophages has not been investigated. Growing evidences indicate that FoxO1 acts as an upstream regulator of IRF4 and could have a role in a specific inflammatory phenotype of macrophages. Therefore, we hypothesized that IRF4 expression regulated by FoxO1 in alveolar macrophages is required for established type 2 immune mediates allergic lung inflammation. Our data indicate that targeted deletion of FoxO1 using FoxO1-selective inhibitor AS1842856 and genetic ablation of FoxO1 in macrophages significantly decreases IRF4 and various M2 macrophage-associated genes, suggesting a mechanism that involves FoxO1-IRF4 signaling in alveolar macrophages that works to polarize macrophages toward established type 2 immune responses. In response to the challenge of DRA (dust mite, ragweed, and Aspergillus) allergens, macrophage specific FoxO1 overexpression is associated with an accentuation of asthmatic lung inflammation, whereas pharmacologic inhibition of FoxO1 by AS1842856 attenuates the development of asthmatic lung inflammation. Thus, our study identifies a role for FoxO1-IRF4 signaling in the development of alternatively activated alveolar macrophages that contribute to type 2 allergic airway inflammation.


Subject(s)
Asthma/immunology , Forkhead Box Protein O1/immunology , Macrophages, Alveolar/immunology , Animals , Cell Polarity/immunology , Inflammation/immunology , Interferon Regulatory Factors/immunology , Mice , Mice, Knockout , Phenotype
18.
Biomark Med ; 10(2): 123-35, 2016.
Article in English | MEDLINE | ID: mdl-26808693

ABSTRACT

Lysophosphatidic acid (LPA), a lipid mediator in biological fluids and tissues, is generated mainly by autotaxin that hydrolyzes lysophosphatidylcholine to LPA and choline. Total LPA levels are increased in bronchoalveolar lavage fluid from asthmatic lung, and are strongly induced following subsegmental bronchoprovocation with allergen in subjects with allergic asthma. Polyunsaturated molecular species of LPA (C22:5 and C22:6) are selectively synthesized in the airways of asthma subjects following allergen challenge and in mouse models of allergic airway inflammation, having been identified and quantified by LC/MS/MS lipidomics. This review discusses current knowledge of LPA production in asthmatic lung and the potential utility of polyunsaturated LPA molecular species as novel biomarkers in bronchoalveolar lavage fluid and exhaled breath condensate of asthma subjects.


Subject(s)
Allergens/immunology , Asthma/diagnosis , Bronchoalveolar Lavage Fluid/chemistry , Inflammation/metabolism , Lung/metabolism , Lysophospholipids/metabolism , Adult , Airway Remodeling , Animals , Asthma/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Disease Models, Animal , Humans , Inflammation/physiopathology , Lung/physiopathology , Lysophosphatidylcholines/metabolism , Lysophospholipids/analysis , Mice , Phospholipases A1/metabolism , Phosphoric Diester Hydrolases/analysis , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism
19.
J Mol Cell Biol ; 7(6): 557-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26101328

ABSTRACT

The transcription factor PU.1 is involved in regulation of macrophage differentiation and maturation. However, the role of PU.1 in alternatively activated macrophage (AAM) and asthmatic inflammation has yet been investigated. Here we report that PU.1 serves as a critical regulator of AAM polarization and promotes the pathological progress of asthmatic airway inflammation. In response to the challenge of DRA (dust mite, ragweed, and Aspergillus) allergens, conditional PU.1-deficient (PU/ER(T)(+/-)) mice displayed attenuated allergic airway inflammation, including decreased alveolar eosinophil infiltration and reduced production of IgE, which were associated with decreased mucous glands and goblet cell hyperplasia. The reduced asthmatic inflammation in PU/ER(T)(+/-) mice was restored by adoptive transfer of IL-4-induced wild-type (WT) macrophages. Moreover, after treating PU/ER(T)(+/-) mice with tamoxifen to rescue PU.1 function, the allergic asthmatic inflammation was significantly restored. In vitro studies demonstrate that treatment of PU.1-deficient macrophages with IL-4 attenuated the expression of chitinase 3-like 3 (Ym-1) and resistin-like molecule alpha 1 (Fizz-1), two specific markers of AAM polarization. In addition, PU.1 expression in macrophages was inducible in response to IL-4 challenge, which was associated with phosphorylation of signal transducer and activator of transcription 6 (STAT6). Furthermore, DRA challenge in sensitized mice almost abrogated gene expression of Ym-1 and Fizz-1 in lung tissues of PU/ER(T)(+/-) mice compared with WT mice. These data, all together, indicate that PU.1 plays a critical role in AAM polarization and asthmatic inflammation.


Subject(s)
Asthma/immunology , Asthma/pathology , Macrophage Activation , Macrophages/immunology , Proto-Oncogene Proteins/immunology , Trans-Activators/immunology , Adoptive Transfer , Allergens/immunology , Animals , Chemotaxis, Leukocyte/immunology , Eosinophils/immunology , Hypersensitivity/immunology , Inflammation/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-4/immunology , Lectins/metabolism , Lung/immunology , Lung/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Pulmonary Alveoli/cytology , Th2 Cells/immunology , Trans-Activators/genetics , beta-N-Acetylhexosaminidases/metabolism
20.
J Leukoc Biol ; 97(2): 327-39, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25420919

ABSTRACT

Macrophages are a heterogeneous population of immune cells that are essential for the initiation and containment inflammation. There are 2 well-established populations of inflammatory macrophages: classically activated M1 and alternatively activated M2 macrophages. The FoxO family of transcription factors plays key roles in a number of cellular processes, including cell growth, metabolism, survival, and inflammation. In this study, we determined whether the expression of FoxO1 contributes polarization of macrophages toward the M2-like phenotype by enhancing IL-10 cytokine expression. We identified that FoxO1 is highly expressed in M-CSF-derived (M2-like) macrophage subsets, and this M2-like macrophages showed a preferential FoxO1 enrichment on the IL-10 promoter but not in GM-CSF-derived (M1-like) macrophages during classic activation by LPS treatment, which suggests that FoxO1 enhances IL-10 by binding directly to the IL-10 promoter, especially in BMMs. In addition, our data show that macrophages in the setting of hyperglycemia contribute to the macrophage-inflammatory phenotype through attenuation of the contribution of FoxO1 to activate IL-10 expression. Our data identify a novel role for FoxO1 in regulating IL-10 secretion during classic activation and highlight the potential for therapeutic interventions for chronic inflammatory conditions, such as atherosclerosis, diabetes, inflammatory bowel disease, and arthritis.


Subject(s)
Cell Differentiation/drug effects , Forkhead Transcription Factors/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hyperglycemia/immunology , Interleukin-10/immunology , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Humans , Hyperglycemia/genetics , Hyperglycemia/pathology , Interleukin-10/genetics , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/pathology , Male , Mice , Mice, Knockout , Mice, Obese
SELECTION OF CITATIONS
SEARCH DETAIL
...