191 results on '"Akbari O"'
Search Results
102. Efficacy of Rhesus Theta-Defensin-1 in Experimental Models of Pseudomonas aeruginosa Lung Infection and Inflammation.
- Author
-
Bensman TJ, Jayne JG, Sun M, Kimura E, Meinert J, Wang JC, Schaal JB, Tran D, Rao AP, Akbari O, Selsted ME, and Beringer PM
- Subjects
- Adult, Animals, Cystic Fibrosis physiopathology, Cytokines metabolism, Disease Models, Animal, Epithelial Cells microbiology, Female, Humans, Inflammation, Leukocytes microbiology, Lung microbiology, Macaca mulatta, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Middle Aged, Neutrophils microbiology, Pseudomonas Infections immunology, Pseudomonas Infections microbiology, Pseudomonas aeruginosa immunology, Specific Pathogen-Free Organisms, Sputum microbiology, Anti-Bacterial Agents therapeutic use, Cystic Fibrosis complications, Defensins therapeutic use, Pseudomonas Infections drug therapy, Pseudomonas aeruginosa drug effects
- Abstract
Chronic airway infection and inflammation contribute to the progressive loss of lung function and shortened survival of patients with cystic fibrosis (CF). Rhesus theta defensin-1 (RTD-1) is a macrocyclic host defense peptide with antimicrobial and immunomodulatory activities. Combined with favorable preclinical safety and peptide stability data, RTD-1 warrants investigation to determine its therapeutic potential for treatment of CF lung disease. We sought to evaluate the therapeutic potential of RTD-1 for CF airway infection and inflammation using in vitro , ex vivo , and in vivo models. We evaluated RTD-1's effects on basal and Pseudomonas aeruginosa- induced inflammation in CF sputum leukocytes and CF bronchial epithelial cells. Peptide stability was evaluated by incubation with CF sputum. Airway pharmacokinetics, safety, and tolerance studies were performed in naive mice. Aerosolized RTD-1 treatment effects were assessed by analyzing lung bacterial burdens and airway inflammation using an established model of chronic P. aeruginosa endobronchial infection in CF (ΔF508) mice. RTD-1 directly reduces metalloprotease activity, as well as inflammatory cytokine secretion from CF airway leukocyte and bronchial epithelial cells. Intrapulmonary safety, tolerability, and stability data support the aerosol administration route. RTD-1 reduced the bacterial lung burden, airway neutrophils, and inflammatory cytokines in CF mice with chronic P. aeruginosa lung infection. Collectively, these studies support further development of RTD-1 for treatment of CF airway disease., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
103. Type two innate lymphoid cells: the Janus cells in health and disease.
- Author
-
Maazi H and Akbari O
- Subjects
- Animals, Biomarkers, Cell Plasticity genetics, Cell Plasticity immunology, Cytokines metabolism, Disease Susceptibility, Gene Expression Regulation, Humans, Immunity, Immunologic Memory, Immunomodulation, Lymphocyte Activation genetics, Lymphocyte Activation immunology, Phenotype, Immunity, Innate, Lymphocyte Subsets immunology, Lymphocyte Subsets metabolism, Th2 Cells immunology, Th2 Cells metabolism
- Abstract
Innate lymphoid cells are functionally diverse subsets of immune cells including the conventional natural killer cells, lymphoid tissue inducers, type 1, 2, and 3 with significant roles in immunity and pathogenesis of inflammatory diseases. Type 2 innate lymphoid cells (ILC2s) resemble type 2 helper (Th2) cells in cytokine production and contribute to anti-helminth immunity, maintaining mucosal tissue integrity, and adipose tissue browning. ILC2s play important roles in the pathogenesis of allergic diseases and asthma. Studying the pathways of activation and regulation of ILC2s are currently a priority for giving a better understanding of pathogenesis of diseases with immunological roots. Recently, our laboratory and others have shown several pathways of regulation of ILC2s by co-stimulatory molecules such as ICOS, regulatory T cells and by compounds such as nicotine. In this review, we summarize the current understanding of the mechanisms of activation and regulation of ILC2s and the role of these cells in health and disease., (© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
104. Type 2 innate lymphoid cell suppression by regulatory T cells attenuates airway hyperreactivity and requires inducible T-cell costimulator-inducible T-cell costimulator ligand interaction.
- Author
-
Rigas D, Lewis G, Aron JL, Wang B, Banie H, Sankaranarayanan I, Galle-Treger L, Maazi H, Lo R, Freeman GJ, Sharpe AH, Soroosh P, and Akbari O
- Subjects
- Animals, Bronchoalveolar Lavage Fluid immunology, Cells, Cultured, Coculture Techniques, Disease Models, Animal, Humans, Immunity, Innate, Mice, Inbred NOD, Mice, SCID, Mice, Transgenic, Asthma immunology, Cytokines immunology, Lymphocytes immunology
- Abstract
Background: Atopic diseases, including asthma, exacerbate type 2 immune responses and involve a number of immune cell types, including regulatory T (Treg) cells and the emerging type 2 innate lymphoid cells (ILC2s). Although ILC2s are potent producers of type 2 cytokines, the regulation of ILC2 activation and function is not well understood., Objective: In the present study, for the first time, we evaluate how Treg cells interact with pulmonary ILC2s and control their function., Methods: ILC2s and Treg cells were evaluated by using in vitro suppression assays, cell-contact assays, and gene expression panels. Also, human ILC2s and Treg cells were adoptively transferred into NOD SCID γC-deficient mice, which were given isotype or anti-inducible T-cell costimulator ligand (ICOSL) antibodies and then challenged with IL-33 and assessed for airway hyperreactivity., Results: We show that induced Treg cells, but not natural Treg cells, effectively suppress the production of the ILC2-driven proinflammatory cytokines IL-5 and IL-13 both in vitro and in vivo. Mechanistically, our data reveal the necessity of inducible T-cell costimulator (ICOS)-ICOS ligand cell contact for Treg cell-mediated ILC2 suppression alongside the suppressive cytokines TGF-β and IL-10. Using a translational approach, we then demonstrate that human induced Treg cells suppress syngeneic human ILC2s through ICOSL to control airway inflammation in a humanized ILC2 mouse model., Conclusion: These findings suggest that peripheral expansion of induced Treg cells can serve as a promising therapeutic target against ILC2-dependent asthma., (Copyright © 2016 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
105. Reply.
- Author
-
Maazi H, Suzuki Y, Jung J, and Akbari O
- Published
- 2017
- Full Text
- View/download PDF
106. Nicotinic acetylcholine receptor agonist attenuates ILC2-dependent airway hyperreactivity.
- Author
-
Galle-Treger L, Suzuki Y, Patel N, Sankaranarayanan I, Aron JL, Maazi H, Chen L, and Akbari O
- Subjects
- Animals, Asthma drug therapy, Asthma metabolism, Cells, Cultured, Cytokines metabolism, Disease Models, Animal, Humans, Mice, Inbred BALB C, Mice, Knockout, Respiratory Hypersensitivity metabolism, Lymphocytes metabolism, Nicotinic Agonists pharmacology, Respiratory Hypersensitivity drug therapy, alpha7 Nicotinic Acetylcholine Receptor metabolism
- Abstract
Allergic asthma is a complex and chronic inflammatory disorder that is associated with airway hyperreactivity (AHR) and driven by Th2 cytokine secretion. Type 2 innate lymphoid cells (ILC2s) produce large amounts of Th2 cytokines and contribute to the development of AHR. Here, we show that ILC2s express the α7-nicotinic acetylcholine receptor (α7nAChR), which is thought to have an anti-inflammatory role in several inflammatory diseases. We show that engagement of a specific agonist with α7nAChR on ILC2s reduces ILC2 effector function and represses ILC2-dependent AHR, while decreasing expression of ILC2 key transcription factor GATA-3 and critical inflammatory modulator NF-κB, and reducing phosphorylation of upstream kinase IKKα/β. Additionally, the specific α7nAChR agonist reduces cytokine production and AHR in a humanized ILC2 mouse model. Collectively, our data suggest that α7nAChR expressed by ILC2s is a potential therapeutic target for the treatment of ILC2-mediated asthma.
- Published
- 2016
- Full Text
- View/download PDF
107. Isoaspartylation appears to trigger small cell lung cancer-associated autoimmunity against neuronal protein ELAVL4.
- Author
-
Pulido MA, DerHartunian MK, Qin Z, Chung EM, Kang DS, Woodham AW, Tsou JA, Klooster R, Akbari O, Wang L, Kast WM, Liu SV, Verschuuren JJGM, Aswad DW, and Laird-Offringa IA
- Subjects
- Adult, Aged, Amino Acid Sequence, Animals, ELAV-Like Protein 4 genetics, ELAV-Like Protein 4 metabolism, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Male, Mice, Mice, Knockout, Middle Aged, Neurons metabolism, Rabbits, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma metabolism, Autoimmunity immunology, ELAV-Like Protein 4 immunology, Lung Neoplasms immunology, Neurons immunology, Small Cell Lung Carcinoma immunology
- Abstract
Autoantibodies against SCLC-associated neuronal antigen ELAVL4 (HuD) have been linked to smaller tumors and improved survival, but the antigenic epitope and mechanism of autoimmunity have never been solved. We report that recombinant human ELAVL4 protein incubated under physiological conditions acquires isoaspartylation, a type of immunogenic protein damage. Specifically, the N-terminal region of ELAVL4, previously implicated in SCLC-associated autoimmunity, undergoes isoaspartylation in vitro, is recognized by sera from anti-ELAVL4 positive SCLC patients and is highly immunogenic in subcutaneously injected mice and in vitro stimulated human lymphocytes. Our data suggest that isoaspartylated ELAVL4 is the trigger for the SCLC-associated anti-ELAVL4 autoimmune response., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
108. Innate lymphoid cells: a paradigm for low SSI in cleft lip repair.
- Author
-
Simmerman E, Qin X, Marshall B, Perry L, Cai L, Wang T, Yu J, Akbari O, and Baban B
- Subjects
- Biomarkers metabolism, Cleft Lip immunology, Cytokines metabolism, Female, Flow Cytometry, Humans, Infant, Lip metabolism, Lymphocytes metabolism, Male, Microscopy, Electron, Transmission, Mouth Mucosa metabolism, Surgical Wound Infection epidemiology, Cleft Lip surgery, Immunity, Innate, Lip immunology, Lymphocytes immunology, Mouth Mucosa immunology, Plastic Surgery Procedures, Surgical Wound Infection immunology
- Abstract
Background: Cleft lip and palate reconstructions demonstrate significantly lower surgical site infection rates compared with clean-contaminated cases, prompting investigation into the pathophysiology causing this discrepancy. Recent studies have identified a new group of innate lymphocytes called innate lymphoid cells (ILCs), located in barrier surfaces of the skin, airways, and intestine. Our objectives were to explore for the first time the presence of ILCs in the vermillion of neonates and young children undergoing cleft lip reconstruction and characterize their composition by measuring the three classes of ILCs., Materials and Methods: Lip tissue samples were collected from 13 subjects undergoing vermillion resection during cleft lip reconstructive surgery. Preparative, transmission electron microscopy, and analytical flow cytometry were performed. The functionality of ILCs was tested in terms of their capacity to produce type 1 (IFN-γ/TNF-α), type 2 (IL-5/IL-13), and type 3 (IL-17/IL-22) cytokines. Data were analyzed using Student t test or the analysis of variance to establish significance (P < 0.05) among groups for all other data., Results: All three classes of ILCs were detected and visualized in the tissue samples. In all samples, the level of ILC2 subset was significantly higher than the other two ILC subsets (P < 0.01), followed by the ILC1 subset, which was present in significantly higher levels than the ILC3 subset (P < 0.05)., Conclusions: Our data place ILCs for the first time in the interface of oral mucosal immunity, tissue microenvironment, and homeostasis during and after tissue development, possibly explaining lower infection rates in cleft lip or palate reconstructions., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
109. IκB Kinase ε Is an NFATc1 Kinase that Inhibits T Cell Immune Response.
- Author
-
Zhang J, Feng H, Zhao J, Feldman ER, Chen SY, Yuan W, Huang C, Akbari O, Tibbetts SA, and Feng P
- Subjects
- Adoptive Transfer, Animals, CD8-Positive T-Lymphocytes enzymology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Cricetinae, Herpesviridae Infections enzymology, Herpesviridae Infections immunology, Lymphocyte Activation, Melanoma, Experimental enzymology, Melanoma, Experimental immunology, Mesocricetus, Mice, Knockout, Neoplasm Transplantation, Phosphorylation, Protein Processing, Post-Translational, T-Lymphocytes immunology, Virus Latency, I-kappa B Kinase physiology, Immunity, Cellular, NFATC Transcription Factors metabolism, T-Lymphocytes metabolism
- Abstract
Activation of nuclear factor of activated T cells (NFAT) is crucial for immune responses. IKKε is an IκB kinase (IKK)-related kinase, and the function of IKKε remains obscure in T cells, despite its abundant expression. We report that IKKε inhibits NFAT activation and T cell responses by promoting NFATc1 phosphorylation. During T cell activation, IKKε was transiently activated to phosphorylate NFATc1. Loss of IKKε elevated T cell antitumor and antiviral immunity and, therefore, reduced tumor development and persistent viral infection. IKKε was activated in CD8(+) T cells of mice bearing melanoma or persistently infected with a model herpesvirus. These results collectively show that IKKε promotes NFATc1 phosphorylation and inhibits T cell responses, identifying IKKε as a crucial negative regulator of T cell activation and a potential target for immunotherapy., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
110. Lower omental t-regulatory cell count is associated with higher fasting glucose and lower β-cell function in adults with obesity.
- Author
-
Gyllenhammer LE, Lam J, Alderete TL, Allayee H, Akbari O, Katkhouda N, and Goran MI
- Subjects
- Adipose Tissue metabolism, Adult, Blood Glucose analysis, Fasting, Female, Humans, Insulin Resistance, Male, Middle Aged, Tumor Necrosis Factor-alpha metabolism, Diabetes Mellitus, Type 2 metabolism, Obesity metabolism, Omentum metabolism, T-Lymphocytes, Regulatory metabolism
- Abstract
Objective: T-lymphocytes are potential initiators and regulators of adipose tissue (AT) inflammation, but there is limited human data on omental AT. The aim of this study was to assess the relationship between T cells, particularly Foxp3+ regulatory T (Treg) cells, in human subcutaneous (subQ) and omental AT and type 2 diabetes risk., Methods: SubQ and deep subQ (DsubQ) abdominal and omental AT biopsies were collected from 44 patients (body mass index, BMI ≥25) undergoing elective abdominal surgery. Flow cytometry was used to quantify CD4+ T cell (T effector and Treg) and macrophages (M1 and M2), and systemic inflammation was measured in fasting blood., Results: Tregs were significantly lower in omental versus subQ and DsubQ AT, and M1 cell counts were significantly higher in the omental and DsubQ depot relative to the subQ. Only omental AT Tregs were negatively associated with fasting glucose and MCP-1 and positively associated with homeostasis model assessment (HOMA)-β. M1 and M2 cell counts across multiple depots had significant relationships with HOMA-insulin resistance, tumor necrosis factor-α, insulin, and HOMA-β. All relationships were consistent across ethnicities., Conclusions: Tregs were significantly lower in omental versus both subQ adipose depots. Fewer omental Tregs may have metabolic implications based on depot-specific relationships with higher fasting glucose and lower β-cell function., (© 2016 The Obesity Society.)
- Published
- 2016
- Full Text
- View/download PDF
111. Maternal-Derived Hepatitis B Virus e Antigen Alters Macrophage Function in Offspring to Drive Viral Persistence after Vertical Transmission.
- Author
-
Tian Y, Kuo CF, Akbari O, and Ou JH
- Subjects
- Animals, Animals, Genetically Modified, B7-H1 Antigen genetics, CD8-Positive T-Lymphocytes virology, Female, Gene Expression Regulation, Hepatitis B transmission, Hepatitis B e Antigens immunology, Humans, Lymphocyte Activation, Macrophages virology, Mice, Mice, Inbred C57BL, Pregnancy, Viral Load, B7-H1 Antigen metabolism, CD8-Positive T-Lymphocytes immunology, Hepatitis B immunology, Hepatitis B virus physiology, Infectious Disease Transmission, Vertical, Macrophages immunology, Prenatal Exposure Delayed Effects immunology
- Abstract
In contrast to horizontal transmission of hepatitis B virus (HBV) between adults, which often leads to self-limited acute infection, vertical transmission of HBV from mother to child often leads to chronic infection. However, the mechanisms linking vertical transmission with chronic infection are not known. We developed a mouse model to study the effect of maternal HBV infection on HBV persistence in offspring and found that HBV carried by the mother impaired CD8(+) T cell responses to HBV in her offspring, resulting in HBV persistence. This impairment of CD8(+) T cell responses was mediated by hepatic macrophages, which were predisposed by maternal HBV e antigen (HBeAg) to support HBV persistence by upregulation of inhibitory ligand PD-L1 and altered polarization upon restimulation with HBeAg. Depletion of hepatic macrophages led to CD8(+) T cell activation and HBV clearance in the offspring, raising the possibility of targeting macrophages to treat chronic HBV patients., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
112. Lack of autophagy induces steroid-resistant airway inflammation.
- Author
-
Suzuki Y, Maazi H, Sankaranarayanan I, Lam J, Khoo B, Soroosh P, Barbers RG, James Ou JH, Jung JU, and Akbari O
- Subjects
- Animals, Anti-Inflammatory Agents therapeutic use, Autophagy-Related Protein 5 genetics, Bronchoalveolar Lavage Fluid cytology, Bronchoalveolar Lavage Fluid immunology, Cell Count, Cytokines immunology, Female, Lung cytology, Lung immunology, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Pyroglyphidae immunology, Asthma drug therapy, Asthma immunology, Autophagy, Dexamethasone therapeutic use, Drug Resistance
- Abstract
Background: Neutrophilic corticosteroid-resistant asthma accounts for a significant proportion of asthma; however, little is known about the mechanisms that underlie the pathogenesis of the disease., Objective: We sought to address the role of autophagy in lung inflammation and the pathogenesis of corticosteroid-resistant neutrophilic asthma., Methods: We developed CD11c-specific autophagy-related gene 5 (Atg5)(-/-) mice and used several murine models to investigate the role of autophagy in asthmatic patients., Results: For the first time, we found that deletion of the Atg5 gene specifically in CD11c(+) cells, which leads to impairment of the autophagy pathway, causes unprovoked spontaneous airway hyperreactivity and severe neutrophilic lung inflammation in mice. We found that severe lung inflammation impairs the autophagy pathway, particularly in pulmonary CD11c(+) cells in wild-type mice. We further found that adoptive transfer of Atg5(-/-), but not wild-type, bone marrow-derived dendritic cells augments lung inflammation with increased IL-17A levels in the lungs. Our data indicate that neutrophilic asthma in Atg5(-/-) mice is glucocorticoid resistant and IL-17A dependent., Conclusion: Our results suggest that lack of autophagy in pulmonary CD11c(+) cells induces neutrophilic airway inflammation and hyperreactivity., (Copyright © 2015 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
113. Impact of Hepatitis C Virus on the Circulating Levels of IL-7 in HIV-1 Coinfected Women.
- Author
-
Kerzerho J, McIlvaine EJ, Anthony P, Mack WJ, Wang CH, Frederick T, Operskalski E, Chen Z, Al-Harthi L, Landay A, Young MA, Tien PC, Augenbraun M, Strickler HD, Akbari O, Golub ET, Sharp GB, and Kovacs A
- Subjects
- Adult, Antiretroviral Therapy, Highly Active, CD4-Positive T-Lymphocytes immunology, Cross-Sectional Studies, Female, HIV Infections complications, HIV Infections drug therapy, Hepatitis C complications, Humans, Middle Aged, Natural Killer T-Cells, Prospective Studies, Viremia, Coinfection, HIV Infections immunology, HIV-1 immunology, Hepacivirus immunology, Hepatitis C immunology, Interleukin-7 blood
- Abstract
Objectives: Hepatitis C virus (HCV) infection causes an alteration in T-cell maturation and activation in patients coinfected with human immunodeficiency virus (HIV). Because interleukin 7 (IL-7) is a major cytokine controlling T-cell homeostasis, we analyzed the potential influence of HCV coinfection on circulating IL-7 levels in HIV-infected women before and after highly active antiretroviral therapy (HAART)., Design and Methods: This prospective study included 56 HIV monoinfected, 55 HIV/HCV coinfected without HCV viremia, 132 HIV/HCV coinfected with HCV viremia, and 61 HIV/HCV-uninfected women for whom plasma levels of IL-7 were determined by enzyme-linked immunosorbent assay at 1 or more follow-up visits before and after HAART. Cross-sectional analyses of the associations between plasma IL-7 levels and HCV infection, demographic, clinical, and immunologic characteristics were evaluated using univariate and multivariate linear regression models before and after HAART., Results: In multivariate models, IL-7 levels were significantly higher in coinfected HCV viremic women than in HIV monoinfected women (multiplicative effect = 1.48; 95% confidence interval: 1.01 to 2.16; P = 0.04) before HAART, but were similar between these two groups among women after HAART. In addition to HCV viremia, higher IL-7 levels were associated with older age (P = 0.02), lower CD4(+) T-cell count (P = 0.0007), and higher natural killer T-cell count (P = 0.02) in women before HAART. Among HAART-treated women, only lower CD4(+) T-cell count was significantly associated with IL-7 level (P = 0.006)., Conclusions: Our data demonstrate that in HIV-infected women, circulating levels of IL-7 are strongly associated with CD4 T-cell depletion both before and after HAART. Our data also demonstrate that HCV viremia increases circulating IL-7 levels before HAART but not after HAART in coinfected women. This suggests that the effect of HCV on lymphopenia is abrogated by HAART.
- Published
- 2016
- Full Text
- View/download PDF
114. ICOS regulates ILC2s in asthma.
- Author
-
Maazi H and Akbari O
- Subjects
- Animals, Asthma metabolism, Asthma physiopathology, Cytokines immunology, Cytokines metabolism, Humans, Immune System immunology, Immune System metabolism, Inducible T-Cell Co-Stimulator Protein metabolism, Inflammation Mediators immunology, Inflammation Mediators metabolism, Lung metabolism, Lung physiopathology, Signal Transduction, Asthma immunology, Immunity, Innate, Inducible T-Cell Co-Stimulator Protein immunology, Lung immunology
- Published
- 2015
- Full Text
- View/download PDF
115. A Subset of CD8αβ+ Invariant NKT Cells in a Humanized Mouse Model.
- Author
-
Wen X, Kim S, Xiong R, Li M, Lawrenczyk A, Huang X, Chen SY, Rao P, Besra GS, Dellabona P, Casorati G, Porcelli SA, Akbari O, Exley MA, and Yuan W
- Subjects
- Animals, Antigens, CD1d genetics, Antigens, CD1d metabolism, Cytotoxicity, Immunologic, Humans, Immunologic Memory, Immunophenotyping, Mice, Mice, Knockout, Models, Animal, Natural Killer T-Cells immunology, Phenotype, Protein Binding, Receptors, Antigen, T-Cell metabolism, Receptors, Antigen, T-Cell, alpha-beta metabolism, T-Lymphocyte Subsets immunology, Transcription, Genetic, CD8 Antigens metabolism, Natural Killer T-Cells metabolism, T-Lymphocyte Subsets metabolism
- Abstract
Invariant NKT (iNKT) cells are unconventional innate-like T cells demonstrating potent antitumor function in conventional mouse models. However, the iNKT cell ligands have had limited efficacy in human antitumor clinical trials, mostly due to the profound differences in the properties and compositions of iNKT cells between the two species, including the presence of a CD8(+) subset of iNKT cells only in humans. To build reliable in vivo models for studying human iNKT cells, we recently developed the first humanized mouse model (hCD1d-KI) with human CD1d knocked in. To further humanize the mouse model, we now introduced the human invariant NKT TCRα-chain (Vα24Jα18) into the hCD1d-knockin mice. Similar to humans, this humanized mouse model developed a subset of CD8αβ(+) iNKT cells among other human-like iNKT subsets. The presence of the CD8αβ(+) iNKT cells in the thymus suggests that these cells developed in the thymus. In the periphery, these NKT cells showed a strong Th1-biased cytokine response and potent cytotoxicity for syngeneic tumor cells upon activation, as do human CD8αβ(+) iNKT cells. The low binding avidity of iNKT TCRs to the human CD1d/lipid complex and high prevalence of Vβ7 TCRβ among the CD8(+) iNKT cells strongly point to a low avidity-based developmental program for these iNKT cells, which included the suppression of Th-POK and upregulation of eomesodermin transcriptional factors. Our establishment of this extensively humanized mouse model phenotypically and functionally reflecting the human CD1d/iNKT TCR system will greatly facilitate the future design and optimization of iNKT cell-based immunotherapies., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
116. Batf3 deficiency is not critical for the generation of CD8α⁺ dendritic cells.
- Author
-
Mott KR, Maazi H, Allen SJ, Zandian M, Matundan H, Ghiasi YN, Sharifi BG, Underhill D, Akbari O, and Ghiasi H
- Subjects
- Animals, CD8 Antigens genetics, Disease Models, Animal, Gene Knockout Techniques, Herpes Simplex genetics, Herpes Simplex immunology, Herpes Simplex metabolism, Herpesvirus 1, Human immunology, Immunohistochemistry, Immunophenotyping, Mice, Mice, Knockout, Phenotype, Rabbits, Basic-Leucine Zipper Transcription Factors deficiency, CD8 Antigens metabolism, Dendritic Cells immunology, Dendritic Cells metabolism, Repressor Proteins deficiency
- Abstract
Recently, we have reported that CD8α(+) DCs, rather than CD8(+) T cells, are involved in the establishment and maintenance of HSV-1 latency in the trigeminal ganglia (TG) of ocularly infected mice. In the current study, we investigated whether similar results can be obtained using Batf3(-/-) mice that previously were reported to lack CD8α(+) DCs. However, our results demonstrate that Batf3(-/-) mice, without any known infection, express CD8α(+) DCs. Consequently, due to the presence of CD8α(+) DCs, no differences were detected in the level of HSV-1 latency between Batf3(-/-) mice compared with wild type control mice., (Copyright © 2014 Elsevier GmbH. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
117. ICOS:ICOS-ligand interaction is required for type 2 innate lymphoid cell function, homeostasis, and induction of airway hyperreactivity.
- Author
-
Maazi H, Patel N, Sankaranarayanan I, Suzuki Y, Rigas D, Soroosh P, Freeman GJ, Sharpe AH, and Akbari O
- Subjects
- Animals, Asthma genetics, Asthma pathology, Female, Gene Expression Regulation, Homeostasis, Humans, Immunity, Innate, Inducible T-Cell Co-Stimulator Ligand genetics, Inducible T-Cell Co-Stimulator Protein genetics, Interleukin-13 genetics, Interleukin-13 immunology, Interleukin-2 genetics, Interleukin-2 immunology, Interleukin-33, Interleukin-5 genetics, Interleukin-5 immunology, Interleukins genetics, Interleukins immunology, Lymphocytes pathology, Mice, Transgenic, Respiratory System immunology, Respiratory System pathology, STAT5 Transcription Factor genetics, STAT5 Transcription Factor immunology, Signal Transduction, Asthma immunology, Inducible T-Cell Co-Stimulator Ligand immunology, Inducible T-Cell Co-Stimulator Protein immunology, Lymphocytes immunology
- Abstract
Allergic asthma is caused by Th2-cell-type cytokines in response to allergen exposure. Type 2 innate lymphoid cells (ILC2s) are a newly identified subset of immune cells that, along with Th2 cells, contribute to the pathogenesis of asthma by producing copious amounts of IL-5 and IL-13, which cause eosinophilia and airway hyperreactivity (AHR), a cardinal feature of asthma. ILC2s express ICOS, a T cell costimulatory molecule with a currently unknown function. Here we showed that a lack of ICOS on murine ILC2s and blocking the ICOS:ICOS-ligand interaction in human ILC2s reduced AHR and lung inflammation. ILC2s expressed both ICOS and ICOS-ligand, and the ICOS:ICOS-ligand interaction promoted cytokine production and survival in ILC2s through STAT5 signaling. Thus, ICOS:ICOS-ligand signaling pathway is critically involved in ILC2 function and homeostasis., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
118. Malnutrition Markers and Serum Ghrelin Levels in Hemodialysis Patients.
- Author
-
Montazerifar F, Karajibani M, Gorgij F, and Akbari O
- Abstract
Objective. The aim of study was to investigate the changes levels of serum ghrelin in HD patients and its relationship to some malnutrition markers compared with healthy controls. Methods. Forty-five patients on hemodialysis and forty healthy controls were studied. Biochemical parameters and serum ghrelin levels were measured. Both daily dietary intakes and body mass index (BMI) assessments were performed for evaluation of nutritional status. Results. Ghrelin concentrations were significantly reduced in patients undergoing hemodialysis when compared to healthy controls (5 ± 0.68 (1.1-18.5) pg/mL versus 7.8 ± 0.84 (2.4-18.3) pg/mL; P = 0.004). BMI and serum albumin in HD patients were markedly reduced compared to controls. The patients with an insufficient intake of energy and protein demonstrated slightly lower levels of serum ghrelin. A negative correlation between serum ghrelin concentration with age (r = -0.34, P = 0.02), BUN (r = -0.26, P < 0.01), and serum creatinine (r = -0.27, P < 0.01) was observed in HD patients. Conclusions. The findings suggest that decreased ghrelin levels in HD patients might be associated with anorexia. Further studies are needed to determine changes in serum ghrelin levels during dialysis and to clarify whether the decrease in ghrelin levels contributes to the malnutrition that is common in these patients.
- Published
- 2014
- Full Text
- View/download PDF
119. Inclusion of CD80 in HSV targets the recombinant virus to PD-L1 on DCs and allows productive infection and robust immune responses.
- Author
-
Mott KR, Allen SJ, Zandian M, Akbari O, Hamrah P, Maazi H, Wechsler SL, Sharpe AH, Freeman GJ, and Ghiasi H
- Subjects
- Adjuvants, Immunologic metabolism, Animals, B7-1 Antigen metabolism, B7-H1 Antigen metabolism, Blotting, Southern, DNA Primers genetics, Dendritic Cells metabolism, Flow Cytometry, Fluorescent Antibody Technique, Genetic Engineering, Herpes Simplex genetics, MicroRNAs genetics, Rabbits, Real-Time Polymerase Chain Reaction, Adjuvants, Immunologic genetics, B7-1 Antigen genetics, Herpes Simplex immunology, Herpesvirus 1, Human genetics, Recombination, Genetic genetics, Viral Vaccines genetics
- Abstract
CD80 plays a critical role in stimulation of T cells and subsequent control of infection. To investigate the effect of CD80 on HSV-1 infection, we constructed a recombinant HSV-1 virus that expresses two copies of the CD80 gene in place of the latency associated transcript (LAT). This mutant virus (HSV-CD80) expressed high levels of CD80 and had similar virus replication kinetics as control viruses in rabbit skin cells. In contrast to parental virus, this CD80 expressing recombinant virus replicated efficiently in immature dendritic cells (DCs). Additionally, the susceptibility of immature DCs to HSV-CD80 infection was mediated by CD80 binding to PD-L1 on DCs. This interaction also contributed to a significant increase in T cell activation. Taken together, these results suggest that inclusion of CD80 as a vaccine adjuvant may promote increased vaccine efficacy by enhancing the immune response directly and also indirectly by targeting to DC.
- Published
- 2014
- Full Text
- View/download PDF
120. Programmed cell death ligand 2 regulates TH9 differentiation and induction of chronic airway hyperreactivity.
- Author
-
Kerzerho J, Maazi H, Speak AO, Szely N, Lombardi V, Khoo B, Geryak S, Lam J, Soroosh P, Van Snick J, and Akbari O
- Subjects
- Adaptive Immunity, Allergens immunology, Animals, Antibodies immunology, Aspergillus fumigatus immunology, Bronchial Hyperreactivity immunology, Bronchial Hyperreactivity metabolism, Bronchial Hyperreactivity pathology, Cell Differentiation immunology, Chronic Disease, Disease Models, Animal, Female, Gene Expression Regulation, Interleukin-1alpha immunology, Lung metabolism, Lung pathology, Lymphocyte Count, Mice, Mice, Inbred BALB C, Programmed Cell Death 1 Ligand 2 Protein immunology, Severity of Illness Index, T-Lymphocyte Subsets pathology, Transforming Growth Factor beta immunology, Bronchial Hyperreactivity genetics, Interleukin-9 immunology, Lung immunology, Programmed Cell Death 1 Ligand 2 Protein genetics, T-Lymphocyte Subsets immunology
- Abstract
Background: Asthma is defined as a chronic inflammatory disease of the airways; however, the underlying physiologic and immunologic processes are not fully understood., Objective: The aim of this study was to determine whether TH9 cells develop in vivo in a model of chronic airway hyperreactivity (AHR) and what factors control this development., Method: We have developed a novel chronic allergen exposure model using the clinically relevant antigen Aspergillus fumigatus to determine the time kinetics of TH9 development in vivo., Results: TH9 cells were detectable in the lungs after chronic allergen exposure. The number of TH9 cells directly correlated with the severity of AHR, and anti-IL-9 treatment decreased airway inflammation. Moreover, we have identified programmed cell death ligand (PD-L) 2 as a negative regulator of TH9 cell differentiation. Lack of PD-L2 was associated with significantly increased TGF-β and IL-1α levels in the lungs, enhanced pulmonary TH9 differentiation, and higher morbidity in the sensitized mice., Conclusion: Our findings suggest that PD-L2 plays a pivotal role in the regulation of TH9 cell development in chronic AHR, providing novel strategies for modulating adaptive immunity during chronic allergic responses., (Copyright © 2012 American Academy of Allergy, Asthma & Immunology. Published by Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
121. Sex-specific perinatal nicotine-induced asthma in rat offspring.
- Author
-
Liu J, Naeem E, Tian J, Lombardi V, Kwong K, Akbari O, Torday JS, and Rehan VK
- Subjects
- Airway Resistance drug effects, Animals, Animals, Newborn, Asthma pathology, Asthma physiopathology, Disease Models, Animal, Female, Lung Compliance drug effects, Male, Muscle, Smooth drug effects, Muscle, Smooth pathology, Nicotine administration & dosage, PPAR gamma metabolism, Pregnancy, Rats, Rats, Sprague-Dawley, Rosiglitazone, Sex Characteristics, Smoking adverse effects, Thiazolidinediones administration & dosage, Trachea drug effects, Trachea pathology, Asthma etiology, Nicotine toxicity
- Abstract
Recently, we have suggested that down-regulation of homeostatic mesenchymal peroxisome proliferator-activated receptor γ signaling after in utero nicotine exposure might contribute to asthma. Here, we have exploited an in vivo rat model of asthma to determine if the effects of perinatal nicotine exposure on offspring pulmonary function and mesenchymal markers of airway contractility in both tracheal and lung parenchymal tissue are sex specific, and whether the protection afforded by the peroxisome proliferator-activated receptor γ agonist, rosiglitazone (RGZ), against the perinatal nicotine-induced effect on offspring lung is also sex specific. Pregnant rat dams received placebo, nicotine, or nicotine plus RGZ daily from Embryonic Day 6 until Postnatal Day 21, at which time lung resistance, compliance, tracheal contractility, and the expression of structural and functional mesenchymal markers of pulmonary contractility were determined. Compared with control animals, perinatal nicotine exposure caused a significant increase in airway resistance and a decrease in airway compliance after a methacholine challenge in both male and female offspring, with more pronounced changes in the males. In contrast to this, the effects of perinatal nicotine exposure on acetylcholine-induced tracheal constriction, along with the expression of its mesenchymal markers, were observed exclusively in the male offspring. Concomitant treatment with RGZ normalized the nicotine-induced alterations in pulmonary function in both sexes, as well as the male-specific effects on acetylcholine-induced tracheal constriction, along with the affected mesenchymal markers. These data suggest that perinatal nicotine exposure causes sex-specific perinatal cigarette smoke exposure-induced asthma, providing a powerful phenotypic model for unequivocally determining the underlying nature of the cell molecular mechanism for this disease.
- Published
- 2013
- Full Text
- View/download PDF
122. Lack of PD-L1 expression by iNKT cells improves the course of influenza A infection.
- Author
-
Maazi H, Singh AK, Speak AO, Lombardi V, Lam J, Khoo B, Inn KS, Sharpe AH, Jung JU, and Akbari O
- Subjects
- Animals, Cytokines metabolism, Dendritic Cells cytology, Dendritic Cells immunology, Dendritic Cells virology, Female, Gene Knockout Techniques, Lung immunology, Lung metabolism, Lung virology, Mice, Mice, Inbred BALB C, Natural Killer T-Cells cytology, Natural Killer T-Cells immunology, Orthomyxoviridae Infections metabolism, Programmed Cell Death 1 Ligand 2 Protein deficiency, Programmed Cell Death 1 Ligand 2 Protein genetics, B7-H1 Antigen deficiency, B7-H1 Antigen genetics, Gene Expression Regulation immunology, Influenza A Virus, H1N1 Subtype physiology, Natural Killer T-Cells metabolism, Natural Killer T-Cells virology, Orthomyxoviridae Infections immunology
- Abstract
There is evidence indicating that invariant Natural Killer T (iNKT) cells play an important role in defense against influenza A virus (IAV). However, the effect of inhibitory receptor, programmed death-1 (PD-1), and its ligands, programmed death ligand (PD-L) 1 and 2 on iNKT cells in protection against IAV remains to be elucidated. Here we investigated the effects of these co-stimulatory molecules on iNKT cells in the response to influenza. We discovered that compare to the wild type, PD-L1 deficient mice show reduced sensitivity to IAV infection as evident by reduced weight loss, decreased pulmonary inflammation and cellular infiltration. In contrast, PD-L2 deficient mice showed augmented weight loss, pulmonary inflammation and cellular infiltration compare to the wild type mice after influenza infection. Adoptive transfer of iNKT cells from wild type, PD-L1 or PD-L2 deficient mice into iNKT cell deficient mice recapitulated these findings. Interestingly, in our transfer system PD-L1(-/-)-derived iNKT cells produced high levels of interferon-gamma whereas PD-L2(-/-)-derived iNKT cells produced high amounts of interleukin-4 and 13 suggesting a role for these cytokines in sensitivity to influenza. We identified that PD-L1 negatively regulates the frequency of iNKT cell subsets in the lungs of IAV infected mice. Altogether, these results demonstrate that lack of PD-L1 expression by iNKT cells reduces the sensitivity to IAV and that the presence of PD-L2 is important for dampening the deleterious inflammatory responses after IAV infection. Our findings potentially have clinical implications for developing new therapies for influenza.
- Published
- 2013
- Full Text
- View/download PDF
123. Structural and functional characterization of a novel nonglycosidic type I NKT agonist with immunomodulatory properties.
- Author
-
Kerzerho J, Yu ED, Barra CM, Alari-Pahissa E, Girardi E, Harrak Y, Lauzurica P, Llebaria A, Zajonc DM, Akbari O, and Castaño AR
- Subjects
- Animals, Bronchial Hyperreactivity drug therapy, Bronchial Hyperreactivity immunology, Bronchial Hyperreactivity pathology, Cells, Cultured, Crystallography, X-Ray, Cyclitols agonists, Cyclitols pharmacology, Disease Models, Animal, Female, Galactosylceramides agonists, Immunologic Factors classification, Lymphocyte Activation drug effects, Melanoma, Experimental drug therapy, Melanoma, Experimental immunology, Melanoma, Experimental pathology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Natural Killer T-Cells pathology, Cyclitols chemistry, Galactosylceramides chemistry, Galactosylceramides pharmacology, Immunologic Factors chemistry, Immunologic Factors pharmacology, Natural Killer T-Cells drug effects, Natural Killer T-Cells immunology, Quantitative Structure-Activity Relationship
- Abstract
Activation of type I NKT (iNKT) cells by CD1d-presented agonists is a potent immunotherapeutic tool. α-Galactosylceramide (α-GalCer) is the prototypic agonist, but its excessive potency with simultaneous production of both pro- and anti-inflammatory cytokines hampers its potential therapeutic use. In search for novel agonists, we have analyzed the structure and function of HS44, a synthetic aminocyclitolic ceramide analog designed to avoid unrestrained iNKT cell activation. HS44 is a weaker agonist compared with α-GalCer in vitro, although in vivo it induces robust IFN-γ production, and highly reduced but still functional Th2 response. The characteristic cytokine storm produced upon α-GalCer activation was not induced. Consequently, HS44 induced a very efficient iNKT cell-dependent antitumoral response in B16 animal model. In addition, intranasal administration showed the capacity to induce lung inflammation and airway hyperreactivity, a cardinal asthma feature. Thus, HS44 is able to elicit functional Th1 or Th2 responses. Structural studies show that HS44 binds to CD1d with the same conformation as α-GalCer. The TCR binds to HS44 similarly as α-GalCer, but forms less contacts, thus explaining its weaker TCR affinity and, consequently, its weaker recognition by iNKT cells. The ability of this compound to activate an efficient, but not massive, tailored functional immune response makes it an attractive reagent for immune manipulation.
- Published
- 2012
- Full Text
- View/download PDF
124. Effects of systemic versus local administration of corticosteroids on mucosal tolerance.
- Author
-
Kerzerho J, Wunsch D, Szely N, Meyer HA, Lurz L, Röse L, Wahn U, Akbari O, and Stock P
- Subjects
- Allergens immunology, Animals, Asthma drug therapy, Asthma immunology, Disease Models, Animal, Female, Humans, Mice, Mice, Inbred BALB C, Adrenal Cortex Hormones pharmacology, Cell Proliferation drug effects, Immune Tolerance drug effects, Immunity, Mucosal drug effects, T-Lymphocytes, Regulatory immunology, Th2 Cells immunology
- Abstract
Respiratory exposure to allergen induces T cell tolerance and protection against the development of airway hyperactivity in animal models of asthma. Whereas systemic administration of dexamethasone during the delivery of respiratory Ag has been suggested to prevent the development of mucosal tolerance, the effects of local administration of corticosteroids, first-line treatment for patients with bronchial asthma, on mucosal tolerance remain unknown. To analyze the effects of systemic versus local administration of different types of corticosteroids on the development of mucosal tolerance, mice were exposed to respiratory allergen to induce mucosal tolerance with or without systemic or intranasal application of different doses of dexamethasone or prednisolone. After the induction of mucosal tolerance, proliferation of T cells was inhibited in tolerized mice, whereas systemic applications of corticosteroids restored T cell proliferation and secretion of Th2 cytokines. In contrast, inhaled corticosteroids showed no effect on both T cell proliferation and cytokine secretion. In addition, mice systemically treated with corticosteroids showed an increased airway hyperactivity with a significant lung inflammation, but also an increased T effector cells/regulatory T cells ratio in the second lymphoid organs when compared with mice that receive corticosteroids by inhalation. These results demonstrate that local administration of corticosteroids has no effect on the development of immune tolerance in contrast to systemically applied corticosteroids. Furthermore, although different concentrations of corticosteroids are administered to patients, our results demonstrated that the route of administration rather than the doses affects the effect of corticosteroids on respiratory tolerance induction. Considering the broad application of corticosteroids in patients with allergic disease and asthma, the route of administration of steroid substances seems crucial in terms of treatment and potential side effects. These findings may help elucidate the apparently contradicting results of corticosteroid treatment in allergic diseases.
- Published
- 2012
- Full Text
- View/download PDF
125. Epithelium-specific deletion of TGF-β receptor type II protects mice from bleomycin-induced pulmonary fibrosis.
- Author
-
Li M, Krishnaveni MS, Li C, Zhou B, Xing Y, Banfalvi A, Li A, Lombardi V, Akbari O, Borok Z, and Minoo P
- Subjects
- Animals, Cell Differentiation, Disease Models, Animal, Epithelium immunology, Epithelium metabolism, Epithelium pathology, Female, Humans, Lung drug effects, Lung immunology, Lung metabolism, Lung pathology, Male, Mice, Mice, Knockout, Mice, Transgenic, Pregnancy, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Pulmonary Fibrosis immunology, Pulmonary Fibrosis pathology, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta genetics, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction, Transforming Growth Factor beta metabolism, Bleomycin toxicity, Protein Serine-Threonine Kinases deficiency, Pulmonary Fibrosis chemically induced, Pulmonary Fibrosis prevention & control, Receptors, Transforming Growth Factor beta deficiency
- Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibroproliferative pulmonary disorder for which there are currently no treatments. Although the etiology of IPF is unknown, dysregulated TGF-β signaling has been implicated in its pathogenesis. Recent studies also suggest a central role for abnormal epithelial repair. In this study, we sought to elucidate the function of epithelial TGF-β signaling via TGF-β receptor II (TβRII) and its contribution to fibrosis by generating mice in which TβRII was specifically inactivated in mouse lung epithelium. These mice, which are referred to herein as TβRIINkx2.1-cre mice, were used to determine the impact of TβRII inactivation on (a) embryonic lung morphogenesis in vivo; and (b) the epithelial cell response to TGF-β signaling in vitro and in a bleomycin-induced, TGF-β-mediated mouse model of pulmonary fibrosis. Although postnatally viable with no discernible abnormalities in lung morphogenesis and epithelial cell differentiation, TβRIINkx2.1-cre mice developed emphysema, suggesting a requirement for epithelial TβRII in alveolar homeostasis. Absence of TβRII increased phosphorylation of Smad2 and decreased, but did not entirely block, phosphorylation of Smad3 in response to endogenous/physiologic TGF-β. However, TβRIINkx2.1-cre mice exhibited increased survival and resistance to bleomycin-induced pulmonary fibrosis. To our knowledge, these findings are the first to demonstrate a specific role for TGF-β signaling in the lung epithelium in the pathogenesis of pulmonary fibrosis.
- Published
- 2011
- Full Text
- View/download PDF
126. The chromosomal association/dissociation of the chromatin insulator protein Cp190 of Drosophila melanogaster is mediated by the BTB/POZ domain and two acidic regions.
- Author
-
Oliver D, Sheehan B, South H, Akbari O, and Pai CY
- Subjects
- Animals, CCCTC-Binding Factor, Chromatin genetics, Chromosomes, Insect genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Drosophila Proteins genetics, Drosophila melanogaster chemistry, Drosophila melanogaster genetics, Eye Proteins genetics, Eye Proteins metabolism, Microtubule-Associated Proteins genetics, Nuclear Proteins genetics, Protein Binding, Protein Structure, Tertiary, Repressor Proteins genetics, Repressor Proteins metabolism, Chromatin metabolism, Chromosomes, Insect metabolism, Drosophila Proteins chemistry, Drosophila Proteins metabolism, Drosophila melanogaster metabolism, Insulator Elements, Microtubule-Associated Proteins chemistry, Microtubule-Associated Proteins metabolism, Nuclear Proteins chemistry, Nuclear Proteins metabolism
- Abstract
Background: Chromatin insulators or boundary elements are a class of functional elements in the eukaryotic genome. They regulate gene transcription by interfering with promoter-enhancer communication. The Cp190 protein of Drosophila melanogaster is essential to the function of at least three-types of chromatin insulator complexes organized by Su(Hw), CTCF and BEAF32., Results: We mapped functional regions of Cp190 in vivo and identified three domains that are essential for the insulator function and for the viability of flies: the BTB/POZ domain, an aspartic acid-rich (D-rich) region and a C-terminal glutamic acid-rich (E-rich) region. Other domains including the centrosomal targeting domain and the zinc fingers are dispensable. The N-terminal CP190BTB-D fragment containing the BTB/POZ domain and the D-rich region is sufficient to mediate association with all three types of insulator complexes. The fragment however is not sufficient for insulator activity or viability. The Cp190 and CP190BTB-D are regulated differently in cells treated with heat-shock. The Cp190 dissociated from chromosomes during heat-shock, indicating that dissociation of Cp190 with chromosomes can be regulated. In contrast, the CP190BTB-D fragment didn't dissociate from chromosomes in the same heat-shocked condition, suggesting that the deleted C-terminal regions have a role in regulating the dissociation of Cp190 with chromosomes., Conclusions: The N-terminal fragment of Cp190 containing the BTB/POZ domain and the D-rich region mediates association of Cp190 with all three types of insulator complexes and that the E-rich region of Cp190 is required for dissociation of Cp190 from chromosomes during heat-shock. The heat-shock-induced dissociation is strong evidence indicating that dissociation of the essential insulator protein Cp190 from chromosomes is regulated. Our results provide a mechanism through which activities of an insulator can be modulated by internal and external cues.
- Published
- 2010
- Full Text
- View/download PDF
127. TIM-4, a receptor for phosphatidylserine, controls adaptive immunity by regulating the removal of antigen-specific T cells.
- Author
-
Albacker LA, Karisola P, Chang YJ, Umetsu SE, Zhou M, Akbari O, Kobayashi N, Baumgarth N, Freeman GJ, Umetsu DT, and DeKruyff RH
- Subjects
- 3T3 Cells, Adoptive Transfer, Animals, Epitopes, T-Lymphocyte administration & dosage, Female, Lymphocyte Count, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Ovalbumin administration & dosage, Ovalbumin immunology, Phosphatidylserines biosynthesis, Rats, Rats, Inbred Lew, Receptors, Cell Surface biosynthesis, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets transplantation, Adaptive Immunity, Epitopes, T-Lymphocyte immunology, Membrane Proteins physiology, Phagocytosis immunology, Phosphatidylserines metabolism, Receptors, Cell Surface physiology, T-Lymphocyte Subsets immunology
- Abstract
Adaptive immunity is characterized by the expansion of an Ag-specific T cell population following Ag exposure. The precise mechanisms, however, that control the expansion and subsequent contraction in the number of Ag-specific T cells are not fully understood. We show that T cell/transmembrane, Ig, and mucin (TIM)-4, a receptor for phosphatidylserine, a marker of apoptotic cells, regulates adaptive immunity in part by mediating the removal of Ag-specific T cells during the contraction phase of the response. During Ag immunization or during infection with influenza A virus, blockade of TIM-4 on APCs increased the expansion of Ag-specific T cells, resulting in an increase in secondary immune responses. Conversely, overexpression of TIM-4 on APCs in transgenic mice reduced the number of Ag-specific T cells that remained after immunization, resulting in reduced secondary T cell responses. There was no change in the total number of cell divisions that T cells completed, no change in the per cell proliferative capacity of the remaining Ag-specific T cells, and no increase in the development of Ag-specific regulatory T cells in TIM-4 transgenic mice. Thus, TIM-4-expressing cells regulate adaptive immunity by mediating the removal of phosphatidylserine-expressing apoptotic, Ag-specific T cells, thereby controlling the number of Ag-specific T cells that remain after the clearance of Ag or infection.
- Published
- 2010
- Full Text
- View/download PDF
128. A CD1d-dependent antagonist inhibits the activation of invariant NKT cells and prevents development of allergen-induced airway hyperreactivity.
- Author
-
Lombardi V, Stock P, Singh AK, Kerzerho J, Yang W, Sullivan BA, Li X, Shiratsuchi T, Hnatiuk NE, Howell AR, Yu KO, Porcelli SA, Tsuji M, Kronenberg M, Wilson SB, and Akbari O
- Subjects
- Allergens administration & dosage, Animals, Antigens, CD1d metabolism, Binding, Competitive immunology, Cell Line, Disease Models, Animal, Female, Galactosylceramides administration & dosage, Galactosylceramides antagonists & inhibitors, Humans, Immunosuppressive Agents antagonists & inhibitors, Lymphocyte Activation drug effects, Mice, Mice, Inbred BALB C, Ovalbumin administration & dosage, Ovalbumin immunology, Phosphatidylethanolamines administration & dosage, Polyethylene Glycols administration & dosage, Allergens immunology, Antigens, CD1d physiology, Bronchial Hyperreactivity immunology, Bronchial Hyperreactivity prevention & control, Immunosuppressive Agents pharmacology, Lymphocyte Activation immunology, Natural Killer T-Cells immunology, Phosphatidylethanolamines pharmacology, Polyethylene Glycols pharmacology
- Abstract
The prevalence of asthma continues to increase in westernized countries, and optimal treatment remains a significant therapeutic challenge. Recently, CD1d-restricted invariant NKT (iNKT) cells were found to play a critical role in the induction of airway hyperreactivity (AHR) in animal models and are associated with asthma in humans. To test whether iNKT cell-targeted therapy could be used to treat allergen-induced airway disease, mice were sensitized with OVA and treated with di-palmitoyl-phosphatidyl-ethanolamine polyethylene glycol (DPPE-PEG), a CD1d-binding lipid antagonist. A single dose of DPPE-PEG prevented the development of AHR and pulmonary infiltration of lymphocytes upon OVA challenge, but had no effect on the development of OVA-specific Th2 responses. In addition, DPPE-PEG completely prevented the development of AHR after administration of alpha-galactosylceramide (alpha-GalCer) intranasally. Furthermore, we demonstrate that DPPE-PEG acts as antagonist to alpha-GalCer and competes with alpha-GalCer for binding to CD1d. Finally, we show that DPPE-PEG completely inhibits the alpha-GalCer-induced phosphorylation of ERK tyrosine kinase in iNKT cells, suggesting that DPPE-PEG specifically blocks TCR signaling and thus activation of iNKT cells. Because iNKT cells play a critical role in the development of AHR, the inhibition of iNKT activation by DPPE-PEG suggests a novel approach to treat iNKT cell-mediated diseases such as asthma.
- Published
- 2010
- Full Text
- View/download PDF
129. The role of costimulatory molecules in allergic disease and asthma.
- Author
-
Lombardi V, Singh AK, and Akbari O
- Subjects
- Animals, Antigens, CD immunology, Antigens, CD metabolism, Antigens, Differentiation, T-Lymphocyte immunology, Antigens, Differentiation, T-Lymphocyte metabolism, Asthma metabolism, B7 Antigens, B7-1 Antigen immunology, B7-1 Antigen metabolism, B7-2 Antigen immunology, B7-2 Antigen metabolism, B7-H1 Antigen, Humans, Hypersensitivity metabolism, Inducible T-Cell Co-Stimulator Protein, Intercellular Signaling Peptides and Proteins immunology, Intercellular Signaling Peptides and Proteins metabolism, Programmed Cell Death 1 Ligand 2 Protein, Receptors, Immunologic immunology, Receptors, Immunologic metabolism, Receptors, Tumor Necrosis Factor immunology, Receptors, Tumor Necrosis Factor metabolism, Asthma immunology, Hypersensitivity immunology
- Abstract
The prevalence of allergic diseases has increased rapidly in recent years. It is well established that the deleterious allergic response is initiated by T-cell recognition of major histocompatibility class II-peptide complexes at the surface of antigen-presenting cells. While this first signal gives antigen specificity to the adaptive immune response, a second nonspecific costimulatory signal is required by T cells to become fully activated. This signal is provided by interactions between antigen-presenting cells and T cells through molecules borne at the surfaces of the two cell types. Depending on the type of molecules involved, this secondary signal can promote the development of an inflammatory allergic reaction or may favor immune regulation. Several molecules of the B7 family (CD80, CD86, PD-1, ICOS, CTLA-4) and tumor necrosis factor receptor family (OX40, CD30, 4-1BB, Fas, CD27, CD40) play an important role in delivering costimulatory signals in early and late phases of allergic response. Therefore, costimulatory molecules involved in promotion or prevention of allergic immune responses are potential targets for the development of novel therapeutic approaches. This review aims to recapitulate our current understanding of the relationship between allergic diseases and costimulatory molecules.
- Published
- 2010
- Full Text
- View/download PDF
130. Dendritic cell modulation as a new interventional approach for the treatment of asthma.
- Author
-
Lombardi V and Akbari O
- Subjects
- Animals, Asthma drug therapy, CD4-Positive T-Lymphocytes immunology, Cell Differentiation immunology, Drug Delivery Systems, Humans, Hypersensitivity drug therapy, Immune Tolerance, T-Lymphocytes, Helper-Inducer immunology, Asthma immunology, Dendritic Cells immunology, Hypersensitivity immunology
- Abstract
Dendritic cells (DCs) have a central role in the immune system, as they control the adaptive immune response and mediate both protective immunity and the maintenance of immune tolerance to self antigens. In this review, we will summarize the recent advances regarding the subsets of DCs and how they regulate the differentiation of naive CD4(+) T cells towards different populations of T helper cells. We will particularly describe the role of DCs in the development and regulation of allergic diseases and asthma, and discuss the capacity of DCs to induce proallergenic Th2 cells versus regulatory T cells. Undoubtedly, tolerogenic DCs play a crucial role in the induction of regulatory cells. Understanding the biology of these cells will help us design novel strategies to cure or prevent allergic diseases and asthma., (Copyright 2009 Prous Science, S.A.U. or its licensors. All rights reserved.)
- Published
- 2009
131. Natural killer T cells in the lungs of patients with asthma.
- Author
-
Matangkasombut P, Marigowda G, Ervine A, Idris L, Pichavant M, Kim HY, Yasumi T, Wilson SB, DeKruyff RH, Faul JL, Israel E, Akbari O, and Umetsu DT
- Subjects
- Adult, Allergens immunology, Bronchoalveolar Lavage Fluid cytology, Bronchoalveolar Lavage Fluid immunology, Female, Humans, Lung pathology, Male, Middle Aged, Natural Killer T-Cells metabolism, Asthma immunology, Lung immunology, Natural Killer T-Cells immunology
- Published
- 2009
- Full Text
- View/download PDF
132. Induction of airway hyperreactivity by IL-25 is dependent on a subset of invariant NKT cells expressing IL-17RB.
- Author
-
Stock P, Lombardi V, Kohlrautz V, and Akbari O
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Female, Mice, Asthma chemically induced, Asthma immunology, Interleukins pharmacology, Natural Killer T-Cells immunology, Receptors, Interleukin-17 immunology
- Abstract
IL-25 has been shown to induce Th2 responses and airway hyperreactivity (AHR) in mice, but the mechanism of action is not understood and it is unclear which cells mediate this disease. In this study we show that the receptor for IL-25, IL-17RB, is highly expressed on a subset of naive and activated CD4(+) invariant NKT (iNKT) cells, but not on activated T cells. IL-17RB(+) iNKT cells produced large amounts of Th2 cytokines that were substantially increased by IL-25 stimulation. Furthermore, IL-17RB(+) iNKT cells were capable of restoring AHR in iNKT cell-deficient mice, whereas IL-17RB(-) iNKT cells failed to reconstitute AHR and lung inflammation. Finally, IL-17RB(+) iNKT cells were detected in the lungs of wild-type mice, and induction of AHR by intranasal administration of IL-25 was significantly impaired in iNKT cell-deficient mice. Overall, our data suggest a critical role for iNKT cells in IL-25-mediated AHR. These results may lead to novel therapeutic approaches to target IL-17RB(+) iNKT cells for the treatment of allergic asthma.
- Published
- 2009
- Full Text
- View/download PDF
133. Activation of nonclassical CD1d-restricted NK T cells induces airway hyperreactivity in beta 2-microglobulin-deficient mice.
- Author
-
Koh YI, Kim HY, Meyer EH, Pichavant M, Akbari O, Yasumi T, Savage PB, DeKruyff RH, and Umetsu DT
- Subjects
- Animals, Antigens, CD1d genetics, Cells, Cultured, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Natural Killer T-Cells classification, Natural Killer T-Cells metabolism, Th2 Cells immunology, Antigens, CD1d immunology, Bronchial Hyperreactivity genetics, Bronchial Hyperreactivity immunology, Lymphocyte Activation genetics, Natural Killer T-Cells immunology, beta 2-Microglobulin deficiency, beta 2-Microglobulin genetics
- Abstract
Allergic asthma is characterized by Th2-driven eosinophilic airway inflammation and by a central feature called airway hyperreactivity (AHR), development of which requires the presence of classical type I invariant NK T (iNKT) cells. Allergen-induced AHR, however, develops in beta(2)-microglobulin (beta(2)m)(-/-) mice, which lack classical iNKT cells, suggesting that in some situations iNKT cells may be dispensable for the development of AHR. In contrast, our studies now suggest that a CD1d-restricted, NK1.1(+) noninvariant TCR NKT cell population is present in beta(2)m(-/-) mice and is responsible for the development of AHR but not for Th2 responses. Furthermore, treatment of beta(2)m(-/-) mice with anti-CD1d mAb or anti-NK1.1 mAb unexpectedly abolished allergen-induced AHR. The CD1-restricted NKT cells in these mice, which failed to respond to alpha-galactosylceramide and which therefore were not classical type I iNKT cells, appear to represent an NKT cell subset restricted by a beta(2)m-independent form of CD1d. These results indicate that, although classical type I iNKT cells are normally required for the development of AHR, under different circumstances other NKT cell subsets, including nonclassical NKT cells, may substitute for classical iNKT cells and induce AHR.
- Published
- 2008
- Full Text
- View/download PDF
134. Selective activation, expansion, and monitoring of human iNKT cells with a monoclonal antibody specific for the TCR alpha-chain CDR3 loop.
- Author
-
Exley MA, Hou R, Shaulov A, Tonti E, Dellabona P, Casorati G, Akbari O, Akman HO, Greenfield EA, Gumperz JE, Boyson JE, Balk SP, and Wilson SB
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibody Specificity immunology, Antigens, CD1 genetics, Antigens, CD1d, Bronchi chemistry, Bronchi cytology, Cell Proliferation drug effects, Galactosylceramides pharmacology, Humans, Interferon-gamma metabolism, Interleukin-4 metabolism, Killer Cells, Natural cytology, Killer Cells, Natural metabolism, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear immunology, Liver chemistry, Liver cytology, Lymphocyte Activation drug effects, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Peptides immunology, Phytohemagglutinins pharmacokinetics, Receptors, Antigen, T-Cell analysis, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, Spleen chemistry, Spleen cytology, Vaccination, Antibodies, Monoclonal immunology, Complementarity Determining Regions immunology, Killer Cells, Natural immunology, Lymphocyte Activation immunology, Monitoring, Immunologic methods, Receptors, Antigen, T-Cell, alpha-beta immunology
- Abstract
A significant fraction of CD1d-restricted T cells express an invariant T cell receptor (TCR) alpha-chain. These highly conserved invariant NKT (iNKT) populations are important regulators of a wide spectrum of immune responses. The ability to directly identify and manipulate iNKT cells is essential to understanding their function and to exploit their therapeutic potential. To this end, we sought monoclonal and polyclonal antibodies specific for iNKT cells by immunizing CD1d KO mice, which lack iNKT cells, with a cyclic peptide modeled after the TCRalpha CDR3 loop. One mAb (6B11) was specific for cloned and primary human but not rodent iNKT cells and the human invariant TCRalpha, as shown by transfection and reactivity with human invariant TCRalpha transgenic T cells ex vivo and in situ. 6B11 was utilized to identify, purify, and expand iNKT cells from an otherwise minor component of human peripheral blood lymphocytes and to specifically identify human iNKT cells in tissue. Thus, we report a novel and general strategy for the generation of mAb specific for the CDR3 loop encoded by the TCR of interest. Specifically, an anti-Valpha24Jalpha18 CDR3 loop clonotypic TCR mAb is available for the enumeration and therapeutic manipulation of human and non-human primate iNKT populations.
- Published
- 2008
- Full Text
- View/download PDF
135. ICOS/ICOSL interaction is required for CD4+ invariant NKT cell function and homeostatic survival.
- Author
-
Akbari O, Stock P, Meyer EH, Freeman GJ, Sharpe AH, Umetsu DT, and DeKruyff RH
- Subjects
- Adoptive Transfer, Animals, Antigens, Differentiation, T-Lymphocyte immunology, Apoptosis, Bronchial Hyperreactivity immunology, Bronchial Hyperreactivity metabolism, Bronchoalveolar Lavage Fluid immunology, CD4-Positive T-Lymphocytes metabolism, Cytokines immunology, Cytokines metabolism, Disease Models, Animal, Female, Inducible T-Cell Co-Stimulator Ligand, Inducible T-Cell Co-Stimulator Protein, Ligands, Mice, Mice, Inbred BALB C, Mice, Mutant Strains, Proteins immunology, T-Lymphocyte Subsets metabolism, Antigens, Differentiation, T-Lymphocyte metabolism, Asthma immunology, CD4-Positive T-Lymphocytes immunology, Proteins metabolism, T-Lymphocyte Subsets immunology
- Abstract
The development of airway hyperreactivity (AHR), a cardinal feature of asthma, requires the presence of invariant NKT (iNKT) cells. In a mouse model of asthma, we demonstrated that the induction of AHR required ICOS costimulation of iNKT cells. ICOS was highly expressed on both naive and activated iNKT cells, and expression of ICOS was greater on the CD4(+) iNKT than on CD4(-) iNKT cells. Furthermore, the number of CD4(+) iNKT cells was significantly lower in spleens and livers of ICOS(-/-) and ICOSL(-/-) mice, and the remaining iNKT cells in ICOS(-/-) mice were dysfunctional and failed to reconstitute AHR when adoptively transferred into iNKT cell-deficient Jalpha18(-/-) mice. In addition, direct activation of iNKT cells with alpha-GalCer, which induced AHR in wild-type mice, failed to induce AHR in ICOS(-/-) mice. The failure of ICOS(-/-) iNKT cells to induce AHR was due in part to an inability of the ICOS(-/-) iNKT cells to produce IL-4 and IL-13 on activation. Moreover, survival of wild-type iNKT cells transferred into ICOSL(-/-) mice was greatly reduced due to the induction of apoptosis. These results indicate that ICOS costimulation plays a major role in induction of AHR by iNKT cells and is required for CD4(+) iNKT cell function, homeostasis, and survival in the periphery.
- Published
- 2008
- Full Text
- View/download PDF
136. Recent advances in the role of NKT cells in allergic diseases and asthma.
- Author
-
Stock P and Akbari O
- Subjects
- Animals, Asthma physiopathology, Humans, Immunity, Cellular immunology, Inflammation immunology, Interleukin-13 immunology, Interleukin-4 immunology, Mice, Asthma immunology, Bronchial Hyperreactivity immunology, Killer Cells, Natural immunology
- Abstract
Asthma is the result of chronic airway inflammation that is dominated by the presence of eosinophils and CD4(+) T lymphocytes. CD4(+) T cells include several subsets and play a critical role in orchestrating the inflammation, predominantly by secreting interleukin-4 and interleukin-13. Recently, research identified a new subset of T cells, natural killer T (NKT) cells, which also express the CD4 marker. In contrast to conventional CD4(+) T cells, NKT cells do not respond to peptide antigens, but rather to glycolipids. In animal models of asthma, direct activation of NKT cells by glycolipids results in the secretion of extensive amounts of cytokines and triggers the development of airway hyperreactivity. Moreover, in patients with chronic asthma, NKT cells can be found in bronchoalveolar lavage fluids in significant amounts. These data strongly suggest that NKT cells play an important role in asthma pathogenesis.
- Published
- 2008
- Full Text
- View/download PDF
137. Glycolipid activation of invariant T cell receptor+ NK T cells is sufficient to induce airway hyperreactivity independent of conventional CD4+ T cells.
- Author
-
Meyer EH, Goya S, Akbari O, Berry GJ, Savage PB, Kronenberg M, Nakayama T, DeKruyff RH, and Umetsu DT
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, Eosinophilia immunology, Galactosylceramides immunology, Interleukin-13 genetics, Interleukin-13 physiology, Interleukin-4 genetics, Interleukin-4 physiology, Lung immunology, Lymphocyte Activation, Mice, Mice, Mutant Strains, Asthma immunology, Bronchial Hyperreactivity immunology, Glycolipids immunology, Killer Cells, Natural immunology, Receptors, Antigen, T-Cell analysis, T-Lymphocyte Subsets immunology
- Abstract
Asthma is an inflammatory lung disease, in which conventional CD4+ T cells producing IL-4/IL-13 appear to play an obligatory pathogenic role. Here we show, in a mouse model of asthma, that activation of pulmonary IL-4/IL-13 producing invariant TCR+ CD1d-restricted natural killer T (NKT) cells is sufficient for the development of airway hyperreactivity (AHR), a cardinal feature of asthma, in the absence of conventional CD4+ T cells and adaptive immunity. Respiratory administration of glycolipid antigens that specifically activate NKT cells (alpha-GalactosylCeramide and a Sphingomonas bacterial glycolipid) rapidly induced AHR and inflammation typically associated with protein allergen administration. Naïve MHC class II-deficient mice, which lack conventional CD4+ T but have NKT cells, showed exaggerated baseline AHR and, when challenged with alpha-GalactosylCeramide, demonstrated even greater AHR. These studies demonstrate an expanded role for NKT cells, in which NKT cells not only produce cytokines that influence adaptive immunity but also function as critical effector cells that can induce AHR. These results suggest that NKT cells responding to glycolipid antigens, as well as conventional CD4+ T cells responding to peptide antigens, may be synergistic in the induction of AHR, although in some cases, each may independently induce AHR.
- Published
- 2006
- Full Text
- View/download PDF
138. Respiratory tolerance is inhibited by the administration of corticosteroids.
- Author
-
Stock P, Akbari O, DeKruyff RH, and Umetsu DT
- Subjects
- Administration, Intranasal, Allergens administration & dosage, Allergens immunology, Animals, Cell Proliferation drug effects, Dendritic Cells drug effects, Dendritic Cells immunology, Dendritic Cells metabolism, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Interleukin-10 biosynthesis, Interleukin-10 immunology, Mice, Mice, Inbred BALB C, Ovalbumin administration & dosage, Ovalbumin immunology, Respiratory Hypersensitivity drug therapy, Reverse Transcriptase Polymerase Chain Reaction, T-Lymphocytes, Regulatory immunology, Th2 Cells drug effects, Th2 Cells immunology, Adrenal Cortex Hormones pharmacology, Dexamethasone pharmacology, Immune Tolerance drug effects, Respiratory Hypersensitivity immunology, T-Lymphocytes, Regulatory drug effects
- Abstract
Corticosteroids constitute the most effective current anti-inflammatory therapy for acute and chronic forms of allergic diseases and asthma. Corticosteroids are highly effective in inhibiting the effector function of Th2 cells, eosinophils, and epithelial cells. However, treatment with corticosteroids may also limit beneficial T cell responses, including respiratory tolerance and the development of regulatory T cells (T(Reg)), which actively suppress inflammation in allergic diseases. To examine this possibility, we investigated the effects of corticosteroid administration on the development of respiratory tolerance. Respiratory exposure to Ag-induced T cell tolerance and prevented the subsequent development of allergen-induced airway hyperreactivity. However, treatment with dexamethasone during the delivery of respiratory Ag prevented tolerance, such that allergen sensitization and severe airway hyperreactivity subsequently occurred. Treatment with dexamethasone during respiratory exposure to allergen eliminated the development of IL-10-secreting dendritic cells, which was required for the induction of IL-10-producing allergen-specific T(Reg) cells. Therefore, because allergen-specific T(Reg) cells normally develop to prevent allergic disease and asthma, our results suggest that treatment with corticosteroids, which limit the development of T(Reg) cells and tolerance to allergens, could enhance subsequent Th2 responses and aggravate the long-term course of allergic diseases and asthma.
- Published
- 2005
- Full Text
- View/download PDF
139. TIM-1 induces T cell activation and inhibits the development of peripheral tolerance.
- Author
-
Umetsu SE, Lee WL, McIntire JJ, Downey L, Sanjanwala B, Akbari O, Berry GJ, Nagumo H, Freeman GJ, Umetsu DT, and DeKruyff RH
- Subjects
- Animals, Antibodies administration & dosage, Antibodies immunology, Antibodies pharmacology, Bronchial Hyperreactivity immunology, Bronchial Hyperreactivity pathology, Bronchial Hyperreactivity therapy, Cell Proliferation drug effects, Cells, Cultured, Gene Expression Regulation, Hepatitis A Virus Cellular Receptor 1, Immune Tolerance drug effects, Immunotherapy, Lymphocyte Activation drug effects, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, Mice, Inbred BALB C, T-Lymphocytes cytology, T-Lymphocytes drug effects, Immune Tolerance immunology, Lymphocyte Activation immunology, Membrane Proteins metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
We have examined the function of TIM-1, encoded by a gene identified as an 'atopy susceptibility gene' (Havcr1*), and demonstrate here that TIM-1 is a molecule that costimulates T cell activation. TIM-1 was expressed on CD4(+) T cells after activation and its expression was sustained preferentially in T helper type 2 (T(H)2) but not T(H)1 cells. In vitro stimulation of CD4(+) T cells with a TIM-1-specific monoclonal antibody and T cell receptor ligation enhanced T cell proliferation; in T(H)2 cells, such costimulation greatly enhanced synthesis of interleukin 4 but not interferon-gamma. In vivo, the use of antibody to TIM-1 plus antigen substantially increased production of both interleukin 4 and interferon-gamma in unpolarized T cells, prevented the development of respiratory tolerance, and increased pulmonary inflammation. Our studies suggest that immunotherapies that regulate TIM-1 function may downmodulate allergic inflammatory diseases.
- Published
- 2005
- Full Text
- View/download PDF
140. Role of regulatory dendritic cells in allergy and asthma.
- Author
-
Akbari O and Umetsu DT
- Subjects
- Asthma prevention & control, Female, Humans, Hypersensitivity prevention & control, Hypersensitivity, Delayed immunology, Hypersensitivity, Delayed prevention & control, Hypersensitivity, Immediate immunology, Hypersensitivity, Immediate prevention & control, Immune Tolerance, Male, Sensitivity and Specificity, Th1 Cells physiology, Th2 Cells immunology, Asthma immunology, Dendritic Cells immunology, Hypersensitivity immunology, T-Lymphocyte Subsets immunology
- Abstract
Dendritic cells (DCs) are the most efficient inducers of all immune responses, and are capable of either inducing productive immunity or maintaining the state of tolerance to self antigens and allergens. In this review, we summarize the emerging literature on DCs, with emphasis on the regulatory function of DCs in allergy and asthma. In particular, we summarize recent data regarding the relationship between DC subsets and TH1, TH2, and regulatory T (TReg) cells. The diverse functions of DCs have been attributed to distinct lineages of DCs, which arise from common immature precursor cells that differentiate in response to specific maturation-inducing or local microenvironment conditions. These subsets of DCs induce different lineages of T cells, such as TH1, TH2, and TReg cells, including Th1Reg and Th2Reg cells, which regulate allergic diseases and asthma. Subsets of DCs regulate the induction of a variety of T-cell subtypes, which suppress the development of allergy and asthma, thus providing anti-inflammatory responses and protective immunity.
- Published
- 2005
- Full Text
- View/download PDF
141. Role of regulatory dendritic cells in allergy and asthma.
- Author
-
Akbari O and Umetsu DT
- Subjects
- Animals, Humans, T-Lymphocytes, Regulatory immunology, Th1 Cells immunology, Th2 Cells immunology, Asthma immunology, Dendritic Cells immunology, Respiratory Hypersensitivity immunology, T-Lymphocyte Subsets immunology
- Abstract
Purpose of Review: Dendritic cells are the most efficient inducers of all immune responses, and are capable of inducing either productive immunity or maintaining the state of tolerance to self-antigens and allergens. The present review summarizes the emerging literature on dendritic cells, with the emphasis on regulatory function of dendritic cells in allergy and asthma. In particular we summarize recent data regarding the relationship between dendritic cell subsets and Th1, Th2 and regulatory T (TReg) cells., Recent Findings: The diverse functions of dendritic cells have been attributed to distinct lineages of dendritic cells, which arise from common immature precursor cells that differentiate in response to specific maturation-inducing or local microenvironment conditions. These subsets induce different lineages of T cells such as Th1, Th2 and TReg cells, including Th1Reg and Th2Reg cells, which regulate allergic diseases and asthma., Summary: Subsets of dendritic cells regulate the induction of a variety of T-cell subtypes, which suppress the development of allergy and asthma, thus providing antiinflammatory responses and protective immunity.
- Published
- 2004
- Full Text
- View/download PDF
142. Induction of T helper type 1-like regulatory cells that express Foxp3 and protect against airway hyper-reactivity.
- Author
-
Stock P, Akbari O, Berry G, Freeman GJ, Dekruyff RH, and Umetsu DT
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte metabolism, CD8 Antigens immunology, DNA-Binding Proteins immunology, DNA-Binding Proteins metabolism, Dendritic Cells immunology, Dendritic Cells metabolism, Forkhead Transcription Factors, Inducible T-Cell Co-Stimulator Protein, Interferon-gamma metabolism, Interleukin-10 metabolism, Mice, T-Box Domain Proteins, Th1 Cells metabolism, Transcription Factors metabolism, DNA-Binding Proteins genetics, Respiratory System immunology, Th1 Cells immunology
- Abstract
The range of regulatory T cell (T(R) cell) types that control immune responses is poorly understood. We describe here a population of T(R) cells that developed in vivo from naive CD4(+)CD25(-) T cells during a T helper type 1 (T(H)1)-polarized response, distinct from CD25(+) T(R) cells. These antigen-specific T(R) cells were induced by CD8alpha(+) DCs, produced both interleukin 10 and interferon-gamma, and potently inhibited the development of airway hyper-reactivity. These T(R) cells expressed the transcription factors Foxp3 and T-bet, indicating that these T(R) cells are related to T(H)1 cells. Thus, adaptive T(R) cells are heterogeneous and comprise T(H)1-like T(R) cells as well as previously described T(H)2-like T(R) cells, which express Foxp3 and are induced during the development of respiratory tolerance by CD8alpha(-) DCs.
- Published
- 2004
- Full Text
- View/download PDF
143. CD8(+) T cells regulate immune responses in a murine model of allergen-induced sensitization and airway inflammation.
- Author
-
Stock P, Kallinich T, Akbari O, Quarcoo D, Gerhold K, Wahn U, Umetsu DT, and Hamelmann E
- Subjects
- Animals, Antibodies blood, Antibodies immunology, Cells, Cultured, Female, Hypersensitivity physiopathology, Inflammation immunology, Inflammation physiopathology, Lung physiopathology, Mice, Mice, Inbred BALB C, Allergens immunology, CD8-Positive T-Lymphocytes immunology, Disease Models, Animal, Hypersensitivity immunology, Lung immunology
- Abstract
The role of CD8(+) T cells in the development of allergic airway disease is controversial. On the one hand, CD8(+) T cells are known to inhibit the development of airway hyperreactivity (AHR) in murine models of asthma. In humans, IL-10-producing CD8(+) T cells were shown to act as regulatory cells, inhibiting both proliferation and cytokine secretion of T cells. On the other hand, CD8(+) T cells can promote IL-5-mediated eosinophilic airway inflammation and the development of AHR in animal models. To examine this, we investigated the role of CD8(+) T cells during the induction of allergen-induced AHR and demonstrated a protective effect of CD8(+) T cells. Depletion of CD8(+) T cells prior to the immunization led to increased Th2 responses and increased allergic airway disease. However, after development of AHR, CD8(+) T cells that infiltrated the lungs secreted high levels of IL-4, IL-5 and IL-10, but little IFN-gamma, whereas CD8(+) T cells in the peribronchial lymph nodes or spleen produced high levels of IFN-gamma, but little or no Th2 cytokines. These data demonstrate protective effects of CD8(+)T cells against the induction of immune responses and show a functional diversity of CD8(+) T cells in different compartments of sensitized mice.
- Published
- 2004
- Full Text
- View/download PDF
144. Regulatory T cells control the development of allergic disease and asthma.
- Author
-
Umetsu DT, Akbari O, and Dekruyff RH
- Subjects
- Asthma immunology, Cell Cycle Proteins, Hepatitis A immunology, Humans, Hypersensitivity immunology, Immune Tolerance, Intracellular Signaling Peptides and Proteins, Killer Cells, Natural immunology, Models, Immunological, T-Lymphocyte Subsets immunology, Th1 Cells immunology, Th2 Cells immunology, Transcription Factors immunology, Asthma etiology, Hypersensitivity etiology, T-Lymphocytes immunology
- Abstract
The role of T(H)2 cells in the pathogenesis of allergy and asthma has been well described. However, the immunologic mechanisms that downmodulate and protect against the development of these disorders are poorly characterized. A spectrum of CD4+ T cells, including T(H)1 cells, T(H)3 cells, regulatory T cells, CD25+ T cells, and natural killer T cells might play a critical role in regulating these diseases and are discussed in this review.
- Published
- 2003
145. Essential role of NKT cells producing IL-4 and IL-13 in the development of allergen-induced airway hyperreactivity.
- Author
-
Akbari O, Stock P, Meyer E, Kronenberg M, Sidobre S, Nakayama T, Taniguchi M, Grusby MJ, DeKruyff RH, and Umetsu DT
- Subjects
- Adoptive Transfer, Animals, Antigens, CD1 physiology, Antigens, CD1d, Mice, Mice, Inbred BALB C, Allergens immunology, Bronchial Hyperreactivity etiology, Interleukin-13 physiology, Interleukin-4 physiology, Killer Cells, Natural immunology
- Abstract
Using natural killer T (NKT) cell-deficient mice, we show here that allergen-induced airway hyperreactivity (AHR), a cardinal feature of asthma, does not develop in the absence of V(alpha)14i NKT cells. The failure of NKT cell-deficient mice to develop AHR is not due to an inability of these mice to produce type 2 T-helper (Th2) responses because NKT cell-deficient mice that are immunized subcutaneously at non-mucosal sites produce normal Th2-biased responses. The failure to develop AHR can be reversed by the adoptive transfer of tetramer-purified NKT cells producing interleukin (IL)-4 and IL-13 to Ja281(-/-) mice, which lack the invariant T-cell receptor (TCR) of NKT cells, or by the administration to Cd1d(-/-) mice of recombinant IL-13, which directly affects airway smooth muscle cells. Thus, pulmonary V(alpha)14i NKT cells crucially regulate the development of asthma and Th2-biased respiratory immunity against nominal exogenous antigens. Therapies that target V(alpha)14i NKT cells may be clinically effective in limiting the development of AHR and asthma.
- Published
- 2003
- Full Text
- View/download PDF
146. Mucosal tolerance and immunity: regulating the development of allergic disease and asthma.
- Author
-
Akbari O, Stock P, DeKruyff RH, and Umetsu DT
- Subjects
- Allergens immunology, Animals, Antigen Presentation immunology, Asthma etiology, Dendritic Cells immunology, Eosinophilia immunology, Humans, Hypersensitivity etiology, Immunity, Mucosal immunology, Interleukin-10 immunology, Respiratory Mucosa immunology, Th2 Cells immunology, Asthma immunology, Hypersensitivity immunology, Immune Tolerance immunology
- Abstract
Allergic diseases and asthma are characterized by eosinophilic inflammation induced by Th2 lymphocytes. However, the immunological events and the molecular and cellular mechanisms that protect against and regulate these pathological immune responses are poorly understood. In this review, we discuss the role of immunological tolerance, regulatory T cells, and dendritic cells (DCs) in these protective processes. In addition, we discuss factors like the maturation state of DCs, source and effect of Il-10 production as well as specific combinations of costimulatory molecules in the antigen-presenting cell: T cell synapse, which determine whether tolerance or immunity develops in the respiratory mucosa in response to inhaled allergen., (Copyright 2003 S. Karger AG, Basel)
- Published
- 2003
- Full Text
- View/download PDF
147. Antigen-specific regulatory T cells develop via the ICOS-ICOS-ligand pathway and inhibit allergen-induced airway hyperreactivity.
- Author
-
Akbari O, Freeman GJ, Meyer EH, Greenfield EA, Chang TT, Sharpe AH, Berry G, DeKruyff RH, and Umetsu DT
- Subjects
- Allergens, Animals, Antigens, Antigens, Differentiation, T-Lymphocyte immunology, Asthma immunology, Bronchoalveolar Lavage Fluid, Inducible T-Cell Co-Stimulator Ligand, Inducible T-Cell Co-Stimulator Protein, Interleukin-10 metabolism, Ligands, Mice, Mice, Inbred BALB C, Mice, Transgenic, Ovalbumin toxicity, Antigens, Differentiation, T-Lymphocyte metabolism, Bronchial Hyperreactivity immunology, Proteins metabolism, T-Lymphocytes immunology
- Abstract
Asthma is caused by T-helper cell 2 (Th2)-driven immune responses, but the immunological mechanisms that protect against asthma development are poorly understood. T-cell tolerance, induced by respiratory exposure to allergen, can inhibit the development of airway hyperreactivity (AHR), a cardinal feature of asthma, and we show here that regulatory T (T(R)) cells can mediate this protective effect. Mature pulmonary dendritic cells in the bronchial lymph nodes of mice exposed to respiratory allergen induced the development of T(R) cells, in a process that required T-cell costimulation via the inducible costimulator (ICOS-ICOS-ligand pathway. The T(R) cells produced IL-10, and had potent inhibitory activity; when adoptively transferred into sensitized mice, T(R) cells blocked the development of AHR. Both the development and the inhibitory function of regulatory cells were dependent on the presence of IL-10 and on ICOS-ICOS-ligand interactions. These studies demonstrate that T(R) cells and the ICOS-ICOS-ligand signaling pathway are critically involved in respiratory tolerance and in downregulating pulmonary inflammation in asthma.
- Published
- 2002
- Full Text
- View/download PDF
148. CD4 T-helper cells engineered to produce IL-10 prevent allergen-induced airway hyperreactivity and inflammation.
- Author
-
Oh JW, Seroogy CM, Meyer EH, Akbari O, Berry G, Fathman CG, Dekruyff RH, and Umetsu DT
- Subjects
- Animals, Asthma pathology, Asthma therapy, Bronchial Hyperreactivity immunology, Cell Line, Cytokines biosynthesis, Dose-Response Relationship, Drug, Genetic Therapy, Inflammation pathology, Inflammation prevention & control, Interleukin-10 antagonists & inhibitors, Interleukin-10 physiology, Methacholine Chloride pharmacology, Mice, Mice, Inbred BALB C, Mice, SCID, Ovalbumin immunology, Protein Engineering, Pulmonary Eosinophilia prevention & control, T-Lymphocytes, Helper-Inducer transplantation, Th2 Cells immunology, Transduction, Genetic, Allergens immunology, Asthma immunology, Bronchial Hyperreactivity prevention & control, Interleukin-10 genetics, T-Lymphocytes, Helper-Inducer immunology
- Abstract
Background: T(H)2 cells play a critical role in the pathogenesis of asthma, but the precise immunologic mechanisms that inhibit T(H)2 cell function in vivo are not well understood., Objective: The purpose of our studies was to determine whether T cells producing IL-10 regulate the development of asthma., Methods: We used gene therapy to generate ovalbumin-specific CD4 T-helper cells to express IL-10, and we examined their capacity to regulate allergen-induced airway hyperreactivity., Results: We demonstrated that the CD4 T-helper cells engineered to express IL-10 abolished airway hyperreactivity and airway eosinophilia in BALB/c mice sensitized and challenged with ovalbumin and in SCID mice reconstituted with ovalbumin-specific T(H)2 effector cells. The inhibitory effect of the IL-10-secreting T-helper cells was accompanied by the presence of increased quantities of IL-10 in the bronchoalveolar lavage fluid, was antigen-specific, and was reversed by neutralization of IL-10. Moreover, neutralization of IL-10 by administration of anti-IL-10 mAb in mice sensitized and challenged with ovalbumin seriously exacerbated airway hyperreactivity and airway inflammation., Conclusion: Our results demonstrate that T cells secreting IL-10 in the respiratory mucosa can indeed regulate T(H)2-induced airway hyperreactivity and inflammation, and they strongly suggest that IL-10 plays an important inhibitory role in allergic asthma.
- Published
- 2002
- Full Text
- View/download PDF
149. Asthma: an epidemic of dysregulated immunity.
- Author
-
Umetsu DT, McIntire JJ, Akbari O, Macaubas C, and DeKruyff RH
- Subjects
- Antigens, CD, Asthma epidemiology, Asthma pathology, Communicable Diseases immunology, Genetic Predisposition to Disease, Humans, Hygiene, Immune Tolerance immunology, Inducible T-Cell Co-Stimulator Ligand, Prevalence, Proteins immunology, Th2 Cells immunology, Th2 Cells microbiology, Th2 Cells virology, United States epidemiology, Asthma immunology
- Abstract
The remarkable increase in asthma prevalence that has occurred over the last two decades is thought to be caused by changes in the environment due to improved hygiene and fewer childhood infections. However, the specific infections that limit T helper type 2 (T(H)2)-biased inflammation and asthma are not fully known. Infectious organisms, including commensal bacteria in the gastrointestinal tract and hepatitis A virus, may normally induce the development of regulatory T (T(R)) cells and protective immunity that limit airway inflammation and promote tolerance to respiratory allergens. In the absence of such infections, T(H)2 cells--which are developmentally related to T(R) cells--develop instead and coordinate the development of asthmatic inflammation.
- Published
- 2002
- Full Text
- View/download PDF
150. Identification of Tapr (an airway hyperreactivity regulatory locus) and the linked Tim gene family.
- Author
-
McIntire JJ, Umetsu SE, Akbari O, Potter M, Kuchroo VK, Barsh GS, Freeman GJ, Umetsu DT, and DeKruyff RH
- Subjects
- Amino Acid Sequence, Animals, Asthma immunology, Bronchial Hyperreactivity immunology, Cells, Cultured, Chromosome Mapping, Chromosomes, Human, Pair 5, Cloning, Molecular, Genetic Predisposition to Disease, Hepatitis A Virus Cellular Receptor 1, Hepatitis A Virus Cellular Receptor 2, Humans, Interleukin-13 biosynthesis, Interleukin-4 biosynthesis, Membrane Glycoproteins genetics, Membrane Proteins physiology, Mice, Mice, Inbred DBA, Molecular Sequence Data, Multigene Family, Polymorphism, Genetic, Receptors, Virus genetics, Sequence Homology, Amino Acid, Asthma genetics, Bronchial Hyperreactivity genetics, Membrane Proteins genetics, T-Lymphocytes, Helper-Inducer immunology
- Abstract
To simplify the analysis of asthma susceptibility genes located at human chromosome 5q23-35, we examined congenic mice that differed at the homologous chromosomal segment. We identified a Mendelian trait encoded by T cell and Airway Phenotype Regulator (Tapr). Tapr is genetically distinct from known cytokine genes and controls the development of airway hyperreactivity and T cell production of interleukin 4 (IL-4) and IL-13. Positional cloning identified a gene family that encodes T cell membrane proteins (TIMs); major sequence variants of this gene family (Tim) completely cosegregated with Tapr. The human homolog of TIM-1 is the hepatitis A virus (HAV) receptor, which may explain the inverse relationship between HAV infection and the development of atopy.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.