1
|
Doron I, Kusakabe T, Iliev ID. Immunoglobulins at the interface of the gut mycobiota and anti-fungal immunity. Semin Immunol 2023; 67:101757. [PMID: 37003056 PMCID: PMC10192079 DOI: 10.1016/j.smim.2023.101757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
The dynamic and complex community of microbes that colonizes the intestines is composed of bacteria, fungi, and viruses. At the mucosal surfaces, immunoglobulins play a key role in protection against bacterial and fungal pathogens, and their toxins. Secretory immunoglobulin A (sIgA) is the most abundantly produced antibody at the mucosal surfaces, while Immunoglobulin G (IgG) isotypes play a critical role in systemic protection. IgA and IgG antibodies with reactivity to commensal fungi play an important role in shaping the mycobiota and host antifungal immunity. In this article, we review the latest evidence that establishes a connection between commensal fungi and B cell-mediated antifungal immunity as an additional layer of protection against fungal infections and inflammation.
Collapse
Affiliation(s)
- Itai Doron
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA; The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | - Takato Kusakabe
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA; The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | - Iliyan D Iliev
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA; The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
2
|
Yi EJ, Kim YI, Song JH, Ko HJ, Chang SY. Intranasal immunization with curdlan induce Th17 responses and enhance protection against enterovirus 71. Vaccine 2023; 41:2243-2252. [PMID: 36863926 DOI: 10.1016/j.vaccine.2023.01.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/28/2022] [Accepted: 01/31/2023] [Indexed: 03/04/2023]
Abstract
Mucosal surfaces are in contact with the external environment and protect the body from infection by various microbes. To prevent infectious diseases at the first line of defense, the establishment of pathogen-specific mucosal immunity by mucosal vaccine delivery is needed. Curdlan, a 1,3-β-glucan has a strong immunostimulatory effect when delivered as a vaccine adjuvant. Here, we investigated whether intranasal administration of curdlan and antigen (Ag) could induce sufficient mucosal immune responses and protect against viral infections. Intranasal co-administration of curdlan and OVA increased OVA-specific IgG and IgA Abs in both serum and mucosal secretions. In addition, intranasal co-administration of curdlan and OVA induced the differentiation of OVA-specific Th1/Th17 cells in the draining lymph nodes. To investigate the protective immunity of curdlan against viral infection, intranasal co-administration of curdlan with recombinant VP1 of EV71 C4a was administered and showed enhanced protection against enterovirus 71 in a passive serum transfer model using neonatal hSCARB2 mice, although intranasal administration of VP1 plus curdlan increased VP1-specific helper T cells responses but not mucosal IgA. Next, Mongolian gerbils were intranasally immunized with curdlan plus VP1, and they had effective protection against EV71 C4a infection, while decreasing viral infection and tissue damage by inducing Th17 responses. These results indicated that intranasal curdlan with Ag improved Ag-specific protective immunity by enhancing mucosal IgA and Th17 against viral infection. Our results suggest that curdlan is an advantageous candidate as a mucosal adjuvant and delivery vehicle for the development of mucosal vaccines.
Collapse
Affiliation(s)
- Eun-Je Yi
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Young-In Kim
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea; AI-Superconvergence KIURI Translational Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Jae-Hyoung Song
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do 16499, Republic of Korea.
| |
Collapse
|
3
|
The Effect of Curcumin-Loaded Glucan Nanoparticles on Immune Cells: Size as a Critical Quality Attribute. Pharmaceutics 2023; 15:pharmaceutics15020623. [PMID: 36839945 PMCID: PMC9959491 DOI: 10.3390/pharmaceutics15020623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Curcumin is known for its multiple health benefits, largely due to its antioxidant and anti-inflammatory properties. It has been extensively studied as a therapeutic agent, however, it does not have good clinical efficacy due to its poor water solubility and bioavailability. Despite accepting the encapsulation of this compound in polymeric particles as one of the most promising strategies to increase its therapeutic value, these nanoparticles have fallen short of expectations due to a lack of assessment of their possible adverse effects on the immune system. Therefore, in this work, we report on a new method to encapsulate curcumin into glucan nanoparticles and their effects on cells of the immune system were evaluated. Two different-sized curcumin-loaded glucan NPs (GluCur 100 and GluCur 380) were produced, each with an encapsulation efficiency close to 100%, and were characterized regarding their size distribution, surface properties, and morphology. The results revealed the greatest hemolytic effect and cytotoxicity for the smallest particles (100 nm) tested in human PBMCs and RAW 264.7 cells. Although GluCur 380 NPs showed a weaker ROS production, they were able to inhibit the production of NO by macrophages. Furthermore, we found that the coagulation time was not affected by both sized-particles as well as platelet function. Additionally, both nanoparticles induced lymphocyte proliferation and TNF-α secretion by Mo-DCs. In conclusion, this report emphasizes the importance of the immunotoxicity assessment and how this is dependent on the intrinsic properties of nanomaterials, hopefully contributing to increasing the safety of nanomedicines.
Collapse
|
4
|
Gupta S, Agrawal A. Dendritic cells in inborn errors of immunity. Front Immunol 2023; 14:1080129. [PMID: 36756122 PMCID: PMC9899832 DOI: 10.3389/fimmu.2023.1080129] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) are crucial cells for initiating and maintaining immune response. They play critical role in homeostasis, inflammation, and autoimmunity. A number of molecules regulate their functions including synapse formation, migration, immunity, and induction of tolerance. A number of IEI are characterized by mutations in genes encoding several of these molecules resulting in immunodeficiency, inflammation, and autoimmunity in IEI. Currently, there are 465 Inborn errors of immunity (IEI) that have been grouped in 10 different categories. However, comprehensive studies of DCs have been reported in only few IEI. Here we have reviewed biology of DCs in IEI classified according to recently published IUIS classification. We have reviewed DCs in selected IEI in each group category and discussed in depth changes in DCs where significant data are available regarding role of DCs in clinical and immunological manifestations. These include severe immunodeficiency diseases, antibody deficiencies, combined immunodeficiency with associated and syndromic features, especially disorders of synapse formation, and disorders of immune regulation.
Collapse
Affiliation(s)
- Sudhir Gupta
- Division of Basic and Clinical Immunology, University of California, Irvine, CA, United States
| | | |
Collapse
|
5
|
The Effect of Novel Selenopolysaccharide Isolated from Lentinula edodes Mycelium on Human T Lymphocytes Activation, Proliferation, and Cytokines Synthesis. Biomolecules 2022; 12:biom12121900. [PMID: 36551328 PMCID: PMC9776057 DOI: 10.3390/biom12121900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Polysaccharides isolated from Lentinula edodes are bioactive compounds with immunomodulatory properties. In our previous studies from L. edodes mycelium, we have isolated a selenium(Se)-enriched fraction (named Se-Le-30), a mixture of linear 1,4-α-glucan and linear 1,3-β- and 1,6-β-glucans. In this study, we analyzed the effects of Se-Le-30 on the activation and proliferation of human T lymphocytes stimulated by anti-CD3 and anti-CD3/CD28 antibodies (Abs) and on the production of cytokines by peripheral blood mononuclear cells (PBMCs). Se-Le-30 had effects on T cell proliferation induced by Abs against CD3 and CD28. It significantly inhibited the proliferation of CD3-stimulated CD4+ and CD8+ T cells and enhanced the proliferation of CD4+ T cells stimulated with anti-CD3/CD28 Ab. Moreover, Se-Le-30 downregulated the number of CD3-stimulated CD4+CD69+ cells, CD4+CD25+ cells, as well as CD8+CD25+ cells, and upregulated the expression of CD25 marker on CD4+ and CD8+ T cells activated with anti-CD3/CD28 Abs. Furthermore, Se-Le-30 enhanced the synthesis of IFN-γ by the unstimulated and anti-CD3/CD28-stimulated PBMCs, inhibited synthesis of IL-2 and IL-4 by CD3-stimulated cells, and augmented the synthesis of IL-6 and IL-10 by unstimulated, CD3-stimulated, and CD3/CD28-stimulated PBMCs. Together, we demonstrated that Se-Le-30 exerts immunomodulatory effects on human T lymphocytes. These observations are of importance for the prospective use of Se-Le-30 in research or as a therapeutic compound.
Collapse
|
6
|
Schmidhuber S, Scheiblhofer S, Weiss R, Cserepes M, Tóvári J, Gadermaier G, Bezard E, De Giorgi F, Ichas F, Strunk D, Mandler M. A Novel C-Type Lectin Receptor-Targeted α-Synuclein-Based Parkinson Vaccine Induces Potent Immune Responses and Therapeutic Efficacy in Mice. Vaccines (Basel) 2022; 10:1432. [PMID: 36146508 PMCID: PMC9506002 DOI: 10.3390/vaccines10091432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
The progressive accumulation of misfolded α-synuclein (α-syn) in the brain is widely considered to be causal for the debilitating clinical manifestations of synucleinopathies including, most notably, Parkinson's disease (PD). Immunotherapies, both active and passive, against α-syn have been developed and are promising novel treatment strategies for such disorders. To increase the potency and specificity of PD vaccination, we created the 'Win the Skin Immune System Trick' (WISIT) vaccine platform designed to target skin-resident dendritic cells, inducing superior B and T cell responses. Of the six tested WISIT candidates, all elicited higher immune responses compared to conventional, aluminum adjuvanted peptide-carrier conjugate PD vaccines, in BALB/c mice. WISIT-induced antibodies displayed higher selectivity for α-syn aggregates than those induced by conventional vaccines. Additionally, antibodies induced by two selected candidates were shown to inhibit α-syn aggregation in a dose-dependent manner in vitro. To determine if α-syn fibril formation could also be inhibited in vivo, WISIT candidate type 1 (CW-type 1) was tested in an established synucleinopathy seeding model and demonstrated reduced propagation of synucleinopathy in vivo. Our studies provide proof-of-concept for the efficacy of the WISIT vaccine technology platform and support further preclinical and clinical development of this vaccine candidate.
Collapse
Affiliation(s)
- Sabine Schmidhuber
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
| | - Sandra Scheiblhofer
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Richard Weiss
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Mihály Cserepes
- KINETO Lab Ltd., Csillaghegyi út 19-21, H-1037 Budapest, Hungary
| | - József Tóvári
- KINETO Lab Ltd., Csillaghegyi út 19-21, H-1037 Budapest, Hungary
| | - Gabriele Gadermaier
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Erwan Bezard
- Motac Neuroscience, Alderley Park, Macclesfield SK10 4TF, UK
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francesca De Giorgi
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francois Ichas
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Dirk Strunk
- Cell Therapy Institute, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Markus Mandler
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
| |
Collapse
|
7
|
Progress towards the Elusive Mastitis Vaccines. Vaccines (Basel) 2022; 10:vaccines10020296. [PMID: 35214754 PMCID: PMC8876843 DOI: 10.3390/vaccines10020296] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 01/25/2023] Open
Abstract
Mastitis is a major problem in dairy farming. Vaccine prevention of mammary bacterial infections is of particular interest in helping to deal with this issue, all the more so as antibacterial drug inputs in dairy farms must be reduced. Unfortunately, the effectiveness of current vaccines is not satisfactory. In this review, we examine the possible reasons for the current shortcomings of mastitis vaccines. Some reasons stem from the peculiarities of the mammary gland immunobiology, others from the pathogens adapted to the mammary gland niche. Infection does not induce sterilizing protection, and recurrence is common. Efficacious vaccines will have to elicit immune mechanisms different from and more effective than those induced by infection. We propose focusing our research on a few points pertaining to either the current immune knowledge or vaccinology approaches to get out of the current deadlock. A possible solution is to focus on the contribution of cell-mediated immunity to udder protection based on the interactions of T cells with the mammary epithelium. On the vaccinology side, studies on the orientation of the immune response by adjuvants, the route of vaccine administration and the delivery systems are among the keys to success.
Collapse
|
8
|
Souza TN, Valdez AF, Rizzo J, Zamith-Miranda D, Guimarães AJ, Nosanchuk JD, Nimrichter L. Host cell membrane microdomains and fungal infection. Cell Microbiol 2021; 23:e13385. [PMID: 34392593 PMCID: PMC8664998 DOI: 10.1111/cmi.13385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/14/2021] [Accepted: 07/24/2021] [Indexed: 01/13/2023]
Abstract
Lipid microdomains or lipid rafts are dynamic and tightly ordered regions of the plasma membrane. In mammalian cells, they are enriched in cholesterol, glycosphingolipids, Glycosylphosphatidylinositol-anchored and signalling-related proteins. Several studies have suggested that mammalian pattern recognition receptors are concentrated or recruited to lipid domains during host-pathogen association to enhance the effectiveness of host effector processes. However, pathogens have also evolved strategies to exploit these domains to invade cells and survive. In fungal organisms, a complex cell wall network usually mediates the first contact with the host cells. This cell wall may contain virulence factors that interfere with the host membrane microdomains dynamics, potentially impacting the infection outcome. Indeed, the microdomain disruption can dampen fungus-host cell adhesion, phagocytosis and cellular immune responses. Here, we provide an overview of regulatory strategies employed by pathogenic fungi to engage with and potentially subvert the lipid microdomains of host cells. TAKE AWAY: Lipid microdomains are ordered regions of the plasma membrane enriched in cholesterol, glycosphingolipids (GSL), GPI-anchored and signalling-related proteins. Pathogen recognition by host immune cells can involve lipid microdomain participation. During this process, these domains can coalesce in larger complexes recruiting receptors and signalling proteins, significantly increasing their signalling abilities. The antifungal innate immune response is mediated by the engagement of pathogen-associated molecular patterns to pattern recognition receptors (PRRs) at the plasma membrane of innate immune cells. Lipid microdomains can concentrate or recruit PRRs during host cell-fungi association through a multi-interactive mechanism. This association can enhance the effectiveness of host effector processes. However, virulence factors at the fungal cell surface and extracellular vesicles can re-assembly these domains, compromising the downstream signalling and favouring the disease development. Lipid microdomains are therefore very attractive targets for novel drugs to combat fungal infections.
Collapse
Affiliation(s)
- Taiane N Souza
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F Valdez
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Rizzo
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, Paris, France
| | - Daniel Zamith-Miranda
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Allan Jefferson Guimarães
- Departamento de Microbiologia e Parasitologia-MIP, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Joshua D Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leonardo Nimrichter
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
The Adjuvants Polyphosphazene (PCEP) and a Combination of Curdlan Plus Leptin Promote a Th17-Type Immune Response to an Intramuscular Vaccine in Mice. Vaccines (Basel) 2021; 9:vaccines9050507. [PMID: 34069081 PMCID: PMC8156850 DOI: 10.3390/vaccines9050507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022] Open
Abstract
Our aim was to determine whether polyphosphazene (PCEP), Curdlan (β-glucan, a dectin-1 agonist), and Leptin could act as adjuvants to promote a Th17-type adaptive immune response in mice. Mice were vaccinated via the intramuscular route then boosted three weeks later with Ovalbumin plus: PCEP, Leptin, Curdlan, PCEP+Curdlan, Curdlan+Leptin, or saline. Mice vaccinated with OVA+PCEP and OVA+Curdlan+Leptin showed significantly higher frequency of antigen-specific CD4+ T cells secreting IL-17 relative to OVA-vaccinated mice. No formulation increased the frequency of CD4+ T cells secreting IL-4 or IFNγ. Since activation of innate immunity precedes the development of adaptive immunity, we wished to establish whether induction of Th17-type immunity could be predicted from in vitro experiments and/or from the local cytokine environment after immunization with adjuvants alone. Elevated IL-6 and TGFβ with reduced secretion of IL-12 is a cytokine milieu known to promote differentiation of Th17-type immunity. We injected the immunostimulants or saline buffer into murine thigh muscles and measured acute local cytokine production. PCEP induced significant production of IL-6 and reduced IL-12 production in muscle but it did not lead to elevated TGFβ production. Curdlan+Leptin injected into muscle induced significant production of TGFβ and IL-17 but not IL-6 or IL-12. We also stimulated splenocytes with media or PCEP, Leptin, Curdlan, PCEP+Curdlan, Curdlan+Leptin, PCEP+Leptin, and PCEP+Curdlan+Leptin and measured cytokine production. PCEP stimulation of splenocytes failed to induce significant production of IL-6, IL-12, TGFβ, or IL-17 and therefore ex vivo splenocyte stimulation failed to predict the increased frequency of Th17-type T cells in response to the vaccine. Curdlan-stimulated splenocytes produced Th1-type, inducing cytokine, IL-12. Curdlan+/-PCEP stimulated TGF-β production and Curdlan+Leptin+/- PCEP induced secretion of IL-17. We conclude that PCEP as well as Curdlan+Leptin are Th17-type vaccine adjuvants in mice but that cytokines produced in response to these adjuvants in muscle after injection or in ex vivo cultured splenocytes did not predict their role as a Th17-type adjuvant. Together, these data suggest that the cytokine environments induced by these immunostimulants did not predict induction of an antigen-specific Th17-type adaptive immune response. This is the first report of these adjuvants inducing a Th17-type adaptive immune response.
Collapse
|
10
|
Desamero MJM, Chung SH, Kakuta S. Insights on the Functional Role of Beta-Glucans in Fungal Immunity Using Receptor-Deficient Mouse Models. Int J Mol Sci 2021; 22:4778. [PMID: 33946381 PMCID: PMC8125483 DOI: 10.3390/ijms22094778] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022] Open
Abstract
Understanding the host anti-fungal immunity induced by beta-glucan has been one of the most challenging conundrums in the field of biomedical research. During the last couple of decades, insights on the role of beta-glucan in fungal disease progression, susceptibility, and resistance have been greatly augmented through the utility of various beta-glucan cognate receptor-deficient mouse models. Analysis of dectin-1 knockout mice has clarified the downstream signaling pathways and adaptive effector responses triggered by beta-glucan in anti-fungal immunity. On the other hand, assessment of CR3-deficient mice has elucidated the compelling action of beta-glucans in neutrophil-mediated fungal clearance, and the investigation of EphA2-deficient mice has highlighted its novel involvement in host sensing and defense to oral mucosal fungal infection. Based on these accounts, this review focuses on the recent discoveries made by these gene-targeted mice in beta-glucan research with particular emphasis on the multifaceted aspects of fungal immunity.
Collapse
Affiliation(s)
- Mark Joseph Maranan Desamero
- Laboratory of Biomedical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan;
- Department of Basic Veterinary Sciences, College of Veterinary Medicine, University of the Philippines Los Baños, Laguna 4031, Philippines
| | - Soo-Hyun Chung
- Division of Experimental Animal Immunology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan;
| | - Shigeru Kakuta
- Laboratory of Biomedical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan;
| |
Collapse
|
11
|
Bao M, Ehexige E, Xu J, Ganbold T, Han S, Baigude H. Oxidized curdlan activates dendritic cells and enhances antitumor immunity. Carbohydr Polym 2021; 264:117988. [PMID: 33910726 DOI: 10.1016/j.carbpol.2021.117988] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
Curdlan activates dendritic cells (DCs) and enhances DC-based antitumor immunity. However, hydrophobicity and heterogeneity of curdlan particulates hinder perfect binding of curdlan to dectin-1 receptor, resulting in the reduced activation of antigen presenting cells and limited antitumor effects. Herein, we synthesized partially oxidized curdlan derivative (β-1,3-polyglucuronic acid, denote PGA). PGA-45 polymer, the reaction product prepared from curdlan by oxidation with 4-acetamido-TEMPO/NaClO/NaClO2 systems under acid conditions for 45 min, activated DCs, induced the expression of co-stimulatory molecules and cytokines, and promoted allogenic T cell proliferation as well as the expression of IL-2. Mechanistically, PGA-45 polymer strongly enhanced phosphorylation of IKK-β and reduced the expression of phosphorylated Akt, suggesting that PGA-45 may activate multiple cell surface receptors such as TLR4 and dectin-1. Administration of tumor lysate pulsed DCs pre-treated with PGA-45 particles induced strong antitumor activity in B16F10 melanoma model. Our data suggest that PGA-45 have strong adjuvant effects for anti-cancer immunity and the design of PGA polymers may provide insights in the development of novel adjuvants for cancer immunotherapy.
Collapse
Affiliation(s)
- Mingming Bao
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia, 010020, PR China
| | - Ehexige Ehexige
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia, 010020, PR China
| | - Jing Xu
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia, 010020, PR China
| | - Tsogzolmaa Ganbold
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia, 010020, PR China
| | - Shuqin Han
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia, 010020, PR China.
| | - Huricha Baigude
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia, 010020, PR China.
| |
Collapse
|
12
|
Intranasal Immunization with Acellular Pertussis Vaccines Results in Long-Term Immunity to Bordetella pertussis in Mice. Infect Immun 2021; 89:IAI.00607-20. [PMID: 33318136 PMCID: PMC8097269 DOI: 10.1128/iai.00607-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Bordetella pertussis colonizes the respiratory mucosa of humans, inducing an immune response seeded in the respiratory tract. An individual, once convalescent, exhibits long-term immunity to the pathogen. Current acellular pertussis (aP) vaccines do not induce the long-term immune response observed after natural infection in humans. In this study, we evaluated the durability of protection from intranasal (i.n.) pertussis vaccines in mice. Mice that convalesced from B. pertussis infection served as a control group. Mice were immunized with a mock vaccine (phosphate-buffered saline [PBS]), aP only, or an aP base vaccine combined with one of the following adjuvants: alum, curdlan, or purified whole glucan particles (IRI-1501). We utilized two study designs: short term (challenged 35 days after priming vaccination) and long term (challenged 6 months after boost). The short-term study demonstrated that immunization with i.n. vaccine candidates decreased the bacterial burden in the respiratory tract, reduced markers of inflammation, and induced significant serum and lung antibody titers. In the long-term study, protection from bacterial challenge mirrored the results observed in the short-term challenge study. Immunization with pertussis antigens alone was surprisingly protective in both models; however, the alum and IRI-1501 adjuvants induced significant B. pertussis-specific IgG antibodies in both the serum and lung and increased numbers of anti-B. pertussis IgG-secreting plasma cells in the bone marrow. Our data indicate that humoral responses induced by the i.n. vaccines correlated with protection, suggesting that long-term antibody responses can be protective.
Collapse
|
13
|
Percentage of Myeloid Dendritic Cells in Peripheral Venous Blood Is Negatively Related to Incidence of Graves' Orbitopathy. Mediators Inflamm 2021; 2021:8896055. [PMID: 33574732 PMCID: PMC7857924 DOI: 10.1155/2021/8896055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 11/25/2022] Open
Abstract
The aim of the study was to evaluate the distribution of blood dendritic cells (DCs) in patients with Graves' orbitopathy (GO) and to assess the influence of methylprednisolone therapy on subsets of peripheral blood mononuclear cells (PBMCs). Peripheral blood DC subsets were analyzed by flow cytometry in patients with active GO (n = 17), inactive GO (n = 8), and Graves' disease (GD) without GO (n = 8) and controls (n = 15); additionally, in patients with active GO (n = 17), analyses were done at three time points, i.e., before methylprednisolone treatment and after 6 weeks and after 12 weeks of the treatment. Percentage of myeloid DCs (mDCs) in PBMC fraction was significantly lower in patients with both active and inactive GO, compared to patients with GD without GO and controls (p < 0.05). In addition, mDCs were also documented to be an independent factor negatively associated with GO, however without essential differences between active and inactive phases. On the other hand, we did not observe any changes in the percentage of DCs after methylprednisolone therapy (p > 0.05). In the present study, we have succeeded to firstly demonstrate—according to our knowledge—that blood mDCs are negatively related to GO incidence.
Collapse
|
14
|
Using Expanded Natural Killer Cells as Therapy for Invasive Aspergillosis. J Fungi (Basel) 2020; 6:jof6040231. [PMID: 33080826 PMCID: PMC7712362 DOI: 10.3390/jof6040231] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Invasive aspergillosis (IA) is a major opportunistic fungal infection in patients with haematological malignancies. Morbidity and mortality rates are high despite anti-fungal treatment, as the compromised status of immune system prevents the host from responding optimally to conventional therapy. This raises the consideration for immunotherapy as an adjunctive treatment. In this study, we evaluated the utility of expanded human NK cells as treatment against Aspergillus fumigatus infection in vitro and in vivo. The NK cells were expanded and activated by K562 cells genetically modified to express 4-1BB ligand and membrane-bound interleukin-15 (K562-41BBL-mbIL-15) as feeders. The efficacy of these cells was investigated in A. fumigatus killing assays in vitro and as adoptive cellular therapy in vivo. The expanded NK cells possessed potent killing activity at low effector-to-target ratio of 2:1. Fungicidal activity was morphotypal-dependent and most efficacious against A. fumigatus conidia. Fungicidal activity was mediated by dectin-1 receptors on the expanded NK cells leading to augmented release of perforin, resulting in enhanced direct cytolysis. In an immunocompromised mice pulmonary aspergillosis model, we showed that NK cell treatment significantly reduced fungal burden, hence demonstrating the translational potential of expanded NK cells as adjunctive therapy against IA in immunocompromised patients.
Collapse
|
15
|
Alobaid M, Richards SJ, Alexander M, Gibson M, Ghaemmaghami A. Developing immune-regulatory materials using immobilized monosaccharides with immune-instructive properties. Mater Today Bio 2020; 8:100080. [PMID: 33205040 PMCID: PMC7649522 DOI: 10.1016/j.mtbio.2020.100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 11/15/2022] Open
Abstract
New strategies for immune modulation have shown real promise in regenerative medicine as well as the fight against autoimmune diseases, allergies, and cancer. Dendritic cells (DCs) are gatekeepers of the immune system and their ability in shaping the adaptive immune responses makes DCs ideal targets for immune modulation. Carbohydrates are abundant in different biological systems and are known to modulate DC phenotype and function. However, how simple monosaccharides instruct DC function is less well understood. In this study, we used a combinatorial array of immobilized monosaccharides to investigate how they modulate DC phenotype and function and crucially the impact of such changes on downstream adaptive immune responses. Our data show that a selection of monosaccharides significantly suppress lipopolysaccharide-induced DC activation as evidenced by a reduction in CD40 expression, IL-12 production, and indoleamine 2,3-dioxygenase activity, while inducing a significant increase in IL-10 production. These changes are indicative of the induction of an anti-inflammatory or regulatory phenotype in DCs, which was further confirmed in DC-T cell co-cultures where DCs cultured on the 'regulatory' monosaccharide-coated surfaces were shown to induce naïve T cell polarization toward regulatory phenotype. Our data also highlighted a selection of monosaccharides that are able to promote mixed Treg and Th17 cell differentiation, a T cell phenotype expected to be highly immune suppressive. These data show the potential immunomodulatory effects of immobilized monosaccharides in priming DCs and skewing T cell differentiation toward an immune-regulatory phenotype. The ability to fine-tune immune responses using these simple carbohydrate combinations (e.g. as coatings for existing materials) can be utilized as novel tools for immune modulation with potential applications in regenerative medicine, implantable medical devices, and wound healing where reduction of inflammatory responses and maintaining immune homeostasis are desirable.
Collapse
Key Words
- (Gal1), 100% 1-amino-1-deoxy-β-d-galactose
- (Gal1–Gal2), 50% 1-amino-1-deoxy-β-d-galactose + 50% 2-amino-2-deoxy-β-d-galactose
- (Gal2), 100% 2-amino-2-deoxy-β-d-galactose
- (Gal2–Man1), 90% 2-amino-2-deoxy-β-d-galactose + 10% 1-amino-1-deoxy-β-d-mannose
- (Gal2–Man2), 2-amino-2-deoxy-β-d-galactose + 10% 2-amino-2-deoxy-β-d-mannose
- (Man1–Man2), 40% 1-amino-1-deoxy-β-d-mannose + 60% 2-amino-2-deoxy-β-d-mannose
- CLR, C-type lectin receptor
- Carbohydrates
- DC-SIGN, Dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin
- DCs, Dendritic cells
- Dendritic cells
- FBS, Fetal bovine serum
- Fucose
- Galactose
- IDO, Indoleamine 2,3-dioxygenase
- Immune modulation
- Immune-instructive materials
- LPS, Lipopolysaccharide
- MFI, Median fluorescence intensity
- MR, Mannose receptor
- MT, 1-methyl-DL-tryptophan
- Mannose
- PRR, Pattern recognition receptor
- Polymers
- T cells
Collapse
Affiliation(s)
- M.A. Alobaid
- Immunology & Immuno-Bioengineering, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - S.-J. Richards
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - M.R. Alexander
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - M.I. Gibson
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - A.M. Ghaemmaghami
- Immunology & Immuno-Bioengineering, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| |
Collapse
|
16
|
Geller A, Yan J. Could the Induction of Trained Immunity by β-Glucan Serve as a Defense Against COVID-19? Front Immunol 2020; 11:1782. [PMID: 32760409 PMCID: PMC7372085 DOI: 10.3389/fimmu.2020.01782] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/03/2020] [Indexed: 12/22/2022] Open
Abstract
As the SARS-CoV-2 virus wreaks havoc on the populations, health care infrastructures and economies of nations around the world, finding ways to protect health care workers and bolster immune responses in the general population while we await an effective vaccine will be the difference between life and death for many people. Recent studies show that innate immune populations may possess a form of memory, termed Trained Immunity (TRIM), where innate immune cells undergo metabolic, mitochondrial, and epigenetic reprogramming following exposure to an initial stimulus that results in a memory phenotype of enhanced immune responses when exposed to a secondary, heterologous, stimulus. Throughout the literature, it has been shown that the induction of TRIM using such inducers as the BCG vaccine and β-glucan can provide protection through altered immune responses against a range of viral infections. Here we hypothesize a potential role for β-glucan in decreasing worldwide morbidity and mortality due to COVID-19, and posit several ideas as to how TRIM may actually shape the observed epidemiological phenomena related to COVID-19. We also evaluate the potential effects of β-glucan in relation to the immune dysregulation and cytokine storm observed in COVID-19. Ultimately, we hypothesize that the use of oral β-glucan in a prophylactic setting could be an effective way to boost immune responses and abrogate symptoms in COVID-19, though clinical trials are necessary to confirm the efficacy of this treatment and to further examine differential effects of β-glucan's from various sources.
Collapse
Affiliation(s)
- Anne Geller
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, United States
- Immuno-Oncology Program, Division of Immunotherapy, Department of Surgery, The James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Jun Yan
- Immuno-Oncology Program, Division of Immunotherapy, Department of Surgery, The James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
17
|
Lee J, Zhang J, Chung YJ, Kim JH, Kook CM, González-Navajas JM, Herdman DS, Nürnberg B, Insel PA, Corr M, Mo JH, Tao A, Yasuda K, Rifkin IR, Broide DH, Sciammas R, Webster NJG, Raz E. Inhibition of IRF4 in dendritic cells by PRR-independent and -dependent signals inhibit Th2 and promote Th17 responses. eLife 2020; 9:e49416. [PMID: 32014112 PMCID: PMC7000221 DOI: 10.7554/elife.49416] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cyclic AMP (cAMP) is involved in many biological processes but little is known regarding its role in shaping immunity. Here we show that cAMP-PKA-CREB signaling (a pattern recognition receptor [PRR]-independent mechanism) regulates conventional type-2 Dendritic Cells (cDC2s) in mice and reprograms their Th17-inducing properties via repression of IRF4 and KLF4, transcription factors essential for cDC2-mediated Th2 induction. In mice, genetic loss of IRF4 phenocopies the effects of cAMP on Th17 induction and restoration of IRF4 prevents the cAMP effect. Moreover, curdlan, a PRR-dependent microbial product, activates CREB and represses IRF4 and KLF4, resulting in a pro-Th17 phenotype of cDC2s. These in vitro and in vivo results define a novel signaling pathway by which cDC2s display plasticity and provide a new molecular basis for the classification of novel cDC2 and cDC17 subsets. The findings also reveal that repressing IRF4 and KLF4 pathway can be harnessed for immuno-regulation.
Collapse
Affiliation(s)
- Jihyung Lee
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Junyan Zhang
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- The Second Affiliated Hospital of Guangzhou Medical University (GMU), The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhouChina
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
| | - Young-Jun Chung
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- Department of Otorhinolaryngology-Head and Neck SurgeryDankook University College of MedicineChungnamRepublic of Korea
| | - Jun Hwan Kim
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Chae Min Kook
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - José M González-Navajas
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
- Alicante Institute for Health and Biomedical Research (ISABIAL - FISABIO)AlicanteSpain
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd)Institute of Health Carlos IIIMadridSpain
| | - David S Herdman
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental TherapyUniversity of TübingenTübingenGermany
| | - Paul A Insel
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- Department of PharmacologyUniversity of California San DiegoSan DiegoUnited States
| | - Maripat Corr
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Ji-Hun Mo
- Department of Otorhinolaryngology-Head and Neck SurgeryDankook University College of MedicineChungnamRepublic of Korea
| | - Ailin Tao
- The Second Affiliated Hospital of Guangzhou Medical University (GMU), The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhouChina
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
| | - Kei Yasuda
- Boston University School of MedicineBostonUnited States
| | - Ian R Rifkin
- Boston University School of MedicineBostonUnited States
- VA Boston Healthcare SystemBostonUnited States
| | - David H Broide
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Roger Sciammas
- Center for Comparative MedicineUniversity of California, DavisDavisUnited States
| | - Nicholas JG Webster
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- VA San Diego Healthcare SystemSan DiegoUnited States
| | - Eyal Raz
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
| |
Collapse
|
18
|
Fujimoto K, Kawaguchi Y, Shimohigoshi M, Gotoh Y, Nakano Y, Usui Y, Hayashi T, Kimura Y, Uematsu M, Yamamoto T, Akeda Y, Rhee JH, Yuki Y, Ishii KJ, Crowe SE, Ernst PB, Kiyono H, Uematsu S. Antigen-Specific Mucosal Immunity Regulates Development of Intestinal Bacteria-Mediated Diseases. Gastroenterology 2019; 157:1530-1543.e4. [PMID: 31445037 DOI: 10.1053/j.gastro.2019.08.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/31/2019] [Accepted: 08/15/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Dysregulation of the microbiome has been associated with development of complex diseases, such as obesity and diabetes. However, no method has been developed to control disease-associated commensal microbes. We investigated whether immunization with microbial antigens, using CpG oligodeoxynucleotides and/or curdlan as adjuvants, induces systemic antigen-specific IgA and IgG production and affects development of diseases in mice. METHODS C57BL/6 mice were given intramuscular injections of antigens (ovalbumin, cholera toxin B-subunit, or pneumococcal surface protein A) combined with CpG oligodeoxynucleotides and/or curdlan. Blood and fecal samples were collected weekly and antigen-specific IgG and IgA titers were measured. Lymph nodes and spleens were collected and analyzed by enzyme-linked immunosorbent assay for antigen-specific splenic T-helper 1 cells, T-helper 17 cells, and memory B cells. Six weeks after primary immunization, mice were given a oral, nasal, or vaginal boost of ovalbumin; intestinal lamina propria, bronchial lavage, and vaginal swab samples were collected and antibodies and cytokines were measured. Some mice were also given oral cholera toxin or intranasal Streptococcus pneumoniae and the severity of diarrhea or pneumonia was analyzed. Gnotobiotic mice were gavaged with fecal material from obese individuals, which had a high abundance of Clostridium ramosum (a commensal microbe associated with obesity and diabetes), and were placed on a high-fat diet 2 weeks after immunization with C ramosum. Intestinal tissues were collected and analyzed by quantitative real-time polymerase chain reaction. RESULTS Serum and fecal samples from mice given injections of antigens in combination with CpG oligodeoxynucleotides and curdlan for 3 weeks contained antigen-specific IgA and IgG, and splenocytes produced interferon-gamma and interleukin 17A. Lamina propria, bronchial, and vaginal samples contained antigen-specific IgA after the ovalbumin boost. This immunization regimen prevented development of diarrhea after injection of cholera toxin, and inhibited lung colonization by S pneumoniae. In gnotobiotic mice colonized with C ramosum and placed on a high-fat diet, the mice that had been immunized with C ramosum became less obese than the nonimmunized mice. CONCLUSIONS Injection of mice with microbial antigens and adjuvant induces antigen-specific mucosal and systemic immune responses. Immunization with S pneumoniae antigen prevented lung infection by this bacteria, and immunization with C ramosum reduced obesity in mice colonized with this microbe and placed on a high-fat diet. This immunization approach might be used to protect against microbe-associated disorders of intestine.
Collapse
Affiliation(s)
- Kosuke Fujimoto
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan; Division of Innate Immune Regulation
| | - Yunosuke Kawaguchi
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan; Division of Innate Immune Regulation; Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaki Shimohigoshi
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan; Division of Innate Immune Regulation; Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshiyuki Gotoh
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan; Division of Mucosal Symbiosis, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiko Nakano
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan; Division of Innate Immune Regulation
| | - Yuki Usui
- Division of Systems Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tetsuya Hayashi
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan; Department of Hematology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasumasa Kimura
- Division of Systems Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Miho Uematsu
- Division of Mucosal Symbiosis, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takuya Yamamoto
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan; Laboratory of Vaccine Science, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yukihiro Akeda
- Division of Infection Control and Prevention, Osaka University Hospital, Osaka, Japan; Research Institute for Microbial Diseases, Osaka University, Osaka, Japan; Department of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Joon Haeng Rhee
- Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yoshikazu Yuki
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan; Laboratory of Vaccine Science, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sheila E Crowe
- Department of Medicine, University of California, San Diego, La Jolla, La Jolla, California
| | - Peter B Ernst
- Division of Gastroenterology, Department of Medicine, Chiba University-University of California, San Diego Center for Mucosal Immunology, Allergy and Vaccines, University of California, San Diego, La Jolla, California; Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California; Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, La Jolla, California
| | - Hiroshi Kiyono
- Division of Gastroenterology, Department of Medicine, Chiba University-University of California, San Diego Center for Mucosal Immunology, Allergy and Vaccines, University of California, San Diego, La Jolla, California; Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California; Department of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan; Division of Innate Immune Regulation; Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
19
|
Intranasal acellular pertussis vaccine provides mucosal immunity and protects mice from Bordetella pertussis. NPJ Vaccines 2019; 4:40. [PMID: 31602318 PMCID: PMC6776550 DOI: 10.1038/s41541-019-0136-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022] Open
Abstract
Current acellular pertussis vaccines fall short of optimal protection against the human respiratory pathogen Bordetella pertussis resulting in increased incidence of a previously controlled vaccine- preventable disease. Natural infection is known to induce a protective mucosal immunity. Therefore, in this study, we aimed to use acellular pertussis vaccines to recapitulate these mucosal immune responses. We utilized a murine immunization and challenge model to characterize the efficacy of intranasal immunization (IN) with DTaP vaccine or DTaP vaccine supplemented with curdlan, a known Th1/Th17 promoting adjuvant. Protection from IN delivered DTaP was compared to protection mediated by intraperitoneal injection of DTaP and whole-cell pertussis vaccines. We tracked fluorescently labeled DTaP after immunization and detected that DTaP localized preferentially in the lungs while DTaP with curdlan was predominantly in the nasal turbinates. IN immunization with DTaP, with or without curdlan adjuvant, resulted in anti-B. pertussis and anti-pertussis toxin IgG titers at the same level as intraperitoneally administered DTaP. IN immunization was able to protect against B. pertussis challenge and we observed decreased pulmonary pro-inflammatory cytokines, neutrophil infiltrates in the lung, and bacterial burden in the upper and lower respiratory tract at day 3 post challenge. Furthermore, IN immunization with DTaP triggered mucosal immune responses such as production of B. pertussis-specific IgA, and increased IL-17A. Together, the induction of a mucosal immune response and humoral antibody-mediated protection associated with an IN administered DTaP and curdlan adjuvant warrant further exploration as a pertussis vaccine candidate formulation.
Collapse
|
20
|
Ghoneum MH, Gimzewski JK, Ghoneum AD, Agrawal S. Potential role of MRN-100, an iron-based compound, in upregulating production of cytokine IL-10 in human dendritic cells to promote an anti-inflammatory response in vitro. Int J Immunopathol Pharmacol 2019; 33:2058738419844932. [PMID: 30994016 PMCID: PMC6472165 DOI: 10.1177/2058738419844932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The hydroferrate fluid MRN-100, an iron-based compound with potent antioxidant characteristics, was examined to identify its possible anti-inflammatory effects on human dendritic cells (DCs) in vitro. Human monocyte-derived DCs were treated with MRN-100 at two concentrations (50 and 100 μL/mL) for 24 h and then stimulated with or without lipopolysaccharides (LPS). The expression of DC maturation markers was assessed by flow cytometry and the production of cytokines was determined by enzyme-linked immunosorbent assay (ELISA). Functional assay was performed by co-culturing MRN-100-treated and untreated DCs with allogeneic naïve CD4+ T cells and assaying the T cells' cytokine production. Results show that treatment with MRN-100 significantly upregulated the co-stimulatory molecules CD80 and CD86 and increased human leukocyte antigen-DR (HLA-DR) though not significantly. MRN-100 treatment also significantly increased the production of the anti-inflammatory cytokine interleukin (IL)-10. On the other hand, MRN-100 significantly induced the secretion of pro-inflammatory cytokines such as IL-6 only at high concentrations. Furthermore, DCs pretreated with MRN-100 and either stimulated or not with LPS were able to prime CD4+ T cells to secrete significant amounts of IL-10 while inhibiting the secretion of pro-inflammatory cytokine tumor necrosis factor (TNF)-α. These results indicate that MRN-100 is a powerful anti-inflammatory agent that promotes the generation of an anti-inflammatory immune response in vitro. MRN-100 could be beneficial for treating patients with inflammatory diseases, including arthritis and type 1 diabetes, and its potential benefits should be examined in clinical trials.
Collapse
Affiliation(s)
- Mamdooh H Ghoneum
- 1 Department of Surgery, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - James K Gimzewski
- 2 Department of Chemistry & Biochemistry, UCLA, Los Angeles, CA, USA.,3 California NanoSystems Institute (CNSI), UCLA, Los Angeles, CA, USA
| | - Aya D Ghoneum
- 2 Department of Chemistry & Biochemistry, UCLA, Los Angeles, CA, USA
| | - Sudhanshu Agrawal
- 4 Division of Basic and Clinical Immunology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
21
|
Jaiswal N, Agrawal S, Agrawal A. High fructose-induced metabolic changes enhance inflammation in human dendritic cells. Clin Exp Immunol 2019; 197:237-249. [PMID: 30919933 DOI: 10.1111/cei.13299] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2019] [Indexed: 12/25/2022] Open
Abstract
Dendritic cells (DCs) are critical antigen-presenting cells which are the initiators and regulators of the immune response. Numerous studies support the idea that dietary sugars influence DC functions. Increased consumption of fructose has been thought to be the leading cause of metabolic disorders. Although evidence supports their association with immune dysfunction, the specific mechanisms are not well understood. Fructose is one of the main dietary sugars in our diet. Therefore, here we compared the effect of fructose and glucose on the functions of human DCs. High levels of D-fructose compared to D-glucose led to activation of DCs in vitro by promoting interleukin (IL)-6 and IL-1β production. Moreover, fructose exposed DCs also induced interferon (IFN)-γ secretion from T cells. Proinflammatory response of DCs in high fructose environment was found to be independent of the major known metabolic regulators or glycolytic control. Instead, DC activation on acute exposure to fructose was via activation of receptor for advanced glycation end product (RAGE) in response to increased accumulation of advanced glycation end products (AGE). However, chronic exposure of DCs to high fructose environment induced a shift towards glycolysis compared to glucose cultured DCs. Further investigations revealed that the AGEs formed by fructose induced increased levels of inflammatory cytokines in DCs compared to AGEs from glucose. In summary, understanding the link between metabolic changes and fructose-induced DC activation compared to glucose has broad implications for immune dysfunction associated with metabolic disorders.
Collapse
Affiliation(s)
- N Jaiswal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - S Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - A Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
22
|
Cachat J, Deffert C, Alessandrini M, Roux-Lombard P, Le Gouellec A, Stasia MJ, Hugues S, Krause KH. Altered Humoral Immune Responses and IgG Subtypes in NOX2-Deficient Mice and Patients: A Key Role for NOX2 in Antigen-Presenting Cells. Front Immunol 2018; 9:1555. [PMID: 30050527 PMCID: PMC6050363 DOI: 10.3389/fimmu.2018.01555] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/25/2018] [Indexed: 01/08/2023] Open
Abstract
Chronic granulomatous disease (CGD) is a primary immunodeficiency resulting from loss of function mutations in the reactive oxygen species generating phagocyte NADPH oxidase (NOX2). CGD patients are prone to infection, but also have an increased susceptibility to autoimmune diseases. The aim of this study was to investigate the role of NOX2 in the regulation of specific immunity. In both CGD patients and NOX2-deficient mice, we observed an alteration in the basal proportions of IgG subtypes. Upon immunization with curdlan-a dectin 1 agonist-NOX2-deficient mice showed increased production of IgG2c compared to controls, and restimulation of lymph node-derived cells led to increased production of IFNγ, but not IL-5, indicative hallmark of an enhanced Th1 response. T cell activation was increased in NOX2-deficient mice and a similar trend was observed in vitro when T cells were co-cultured with NOX2-deficient bone marrow-derived cells. In contrast, no difference in T cell activation was observed when NOX2-deficient T cells were co-cultured with wild-type BMDC. Following stimulation of NOX2-deficient dendritic cells (DCs), no difference in costimulatory molecules was observed, while there was an increase in the release of Th1-driving cytokines. In summary, both CGD patients and CGD mice have an altered IgG subtype distribution, which is associated with an increased IFNγ production. Thus, NOX2 within DCs appears to be an important regulator at the interface of innate and specific immunity, especially after activation of the dectin 1 pathway, limiting immune activation and the development of autoimmunity.
Collapse
Affiliation(s)
- Julien Cachat
- Department of Pathology and Immunology, Geneva University Hospitals (HUG) and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christine Deffert
- Division of Laboratory Medicine, Department of Genetic and Laboratory Medicine and Department of Medical Specialities, Geneva University Hospitals (HUG) and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marco Alessandrini
- Department of Pathology and Immunology, Geneva University Hospitals (HUG) and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pascale Roux-Lombard
- Division of Laboratory Medicine, Department of Genetic and Laboratory Medicine and Department of Medical Specialities, Geneva University Hospitals (HUG) and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Audrey Le Gouellec
- TheREx (Thérapeutique Recombinante Expérimentale), Laboratoire TIMC-IMAG, University Grenoble Alpes, CNRS, Grenoble, France.,Laboratoire BEP, Pôle Biologie, CHU Grenoble Alpes, Grenoble, France
| | - Marie-José Stasia
- TheREx (Thérapeutique Recombinante Expérimentale), Laboratoire TIMC-IMAG, University Grenoble Alpes, CNRS, Grenoble, France.,Laboratoire BEP, Pôle Biologie, CHU Grenoble Alpes, Grenoble, France
| | - Stéphanie Hugues
- Department of Pathology and Immunology, Geneva University Hospitals (HUG) and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Geneva University Hospitals (HUG) and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
23
|
Dexamethasone induced inhibition of Dectin-1 activation of antigen presenting cells is mediated via STAT-3 and NF-κB signaling pathways. Sci Rep 2017; 7:4522. [PMID: 28674449 PMCID: PMC5495798 DOI: 10.1038/s41598-017-04558-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 05/17/2017] [Indexed: 02/08/2023] Open
Abstract
Treatment of patients with glucocorticoids can result in an increased risk of infection with pathogens such as fungi. Dectin-1 is a member of the C-type lectin receptor superfamily and was shown to be one of the major receptors for fungal beta-glucans. Activation of Dectin-1 increases the production of cytokines and chemokines and T-cell stimulatory capacity of DC and mediates resolution of fungal infections. Here we show that antigen-presenting cells generated in the presence of dexamethasone (Dex-DC) have a reduced capacity to stimulate T-cell proliferation and decreased expression of costimulatory molecules, that can not be enhanced upon stimulation with Dectin-1 ligands. Stimulation of Dex-DC with beta-glucans induced a strong upregulation of Syk phosphorylation and increased secretion of IL-10, while the production of IL-12, IL-23 and TNF-alpha was reduced. Downstream of Syk stimulation of Dectin-1 on Dex-DC resulted in phosphorylation of STAT3 and reduced nuclear localization of transcription factors involved in DC activation and function.
Collapse
|
24
|
Haas T, Heidegger S, Wintges A, Bscheider M, Bek S, Fischer JC, Eisenkolb G, Schmickl M, Spoerl S, Peschel C, Poeck H, Ruland J. Card9 controls Dectin-1-induced T-cell cytotoxicity and tumor growth in mice. Eur J Immunol 2017; 47:872-879. [PMID: 28295265 PMCID: PMC5434796 DOI: 10.1002/eji.201646775] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/25/2017] [Accepted: 03/07/2017] [Indexed: 02/03/2023]
Abstract
Activation of the C‐type lectin receptor Dectin‐1 by β‐glucans triggers multiple signals within DCs that result in activation of innate immunity. While these mechanisms can potently prime CD8+ cytotoxic T‐cell (CTL) responses without additional adjuvants, the Dectin‐1 effector pathways that control CTL induction remain unclear. Here we demonstrate that Dectin‐1‐induced CTL cross‐priming in mice does not require inflammasome activation but strictly depends on the adapter protein Card9 in vitro. In vivo, Dectin‐1‐mediated Card9 activation after vaccination drives both expansion and activation of Ag‐specific CTLs, resulting in long‐lasting CTL responses that are sufficient to protect mice from tumor challenge. This Dectin‐1‐induced antitumor immune response was independent of NK cell function and completely abrogated in Card9‐deficient mice. Thus, our results demonstrate that Dectin‐1‐triggered Card9 signaling but not inflammasome activation can potently cross‐prime Ag‐specific CTLs, suggesting that this pathway would be a candidate for immunotherapy and vaccine development.
Collapse
Affiliation(s)
- Tobias Haas
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Simon Heidegger
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexander Wintges
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Michael Bscheider
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sarah Bek
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Julius C Fischer
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gabriel Eisenkolb
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Martina Schmickl
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Silvia Spoerl
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Peschel
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Hendrik Poeck
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| |
Collapse
|
25
|
Whiteson K, Agrawal S, Agrawal A. Differential responses of human dendritic cells to metabolites from the oral/airway microbiome. Clin Exp Immunol 2017; 188:371-379. [PMID: 28194750 DOI: 10.1111/cei.12943] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2017] [Indexed: 12/22/2022] Open
Abstract
Small molecule metabolites that are produced or altered by host-associated microbial communities are emerging as significant immune response modifiers. However, there is a key gap in our knowledge of how oral microbial metabolites affect the immune response. Here, we examined the effects of metabolites from five bacterial strains found commonly in the oral/airway microbial communities of humans. The five strains, each isolated from cystic fibrosis patient sputum, were Pseudomonas aeruginosa FLR01 non-mucoid (P1) and FLR02 mucoid (P2) forms, Streptococcus pneumoniae (Sp), S. salivarius (Ss) and Rothia mucilaginosa (Rm). The effect of bacterial metabolites on dendritic cell (DC) activation, T cell priming and cytokine secretion was determined by exposing DCs to bacterial supernatants and individual metabolites of interest. Supernatants from P1 and P2 induced high levels of tumour necrosis factor (TNF)-α, interleukin (IL)-12 and IL-6 from DCs and primed T cells to secrete interferon (IFN)-γ, IL-22 compared to supernatants from Sp, Ss and Rm. Investigations into the composition of supernatants using gas chromatography-mass spectroscopy (GC-MS) revealed signature metabolites for each of the strains. Supernatants from P1 and P2 contained high levels of putrescine and glucose, while Sp and Ss contained high levels of 2,3-butanediol. The individual metabolites replicated the results of whole supernatants, although the magnitudes of their effects were reduced significantly. Altogether, our data demonstrate for the first time that the signature metabolites produced by different bacteria have different effects on DC functions. The identification of signature metabolites and their effects on the host immune system can provide mechanistic insights into diseases and may also be developed as biomarkers.
Collapse
Affiliation(s)
- K Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - S Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - A Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
26
|
Hassanzadeh-Kiabi N, Yáñez A, Dang I, Martins GA, Underhill DM, Goodridge HS. Autocrine Type I IFN Signaling in Dendritic Cells Stimulated with Fungal β-Glucans or Lipopolysaccharide Promotes CD8 T Cell Activation. THE JOURNAL OF IMMUNOLOGY 2016; 198:375-382. [PMID: 27872213 DOI: 10.4049/jimmunol.1601143] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/21/2016] [Indexed: 12/24/2022]
Abstract
Type I IFNs are key mediators of immune defense against viruses and bacteria. Type I IFNs were also previously implicated in protection against fungal infection, but their roles in antifungal immunity have not been thoroughly investigated. A recent study demonstrated that bacterial and fungal β-glucans stimulate IFN-β production by dendritic cells (DCs) following detection by the Dectin-1 receptor, but the effects of β-glucan-induced type I IFNs have not been defined. We investigated whether type I IFNs regulate CD8 T cell activation by fungal β-glucan particle-stimulated DCs. We demonstrate that β-glucan-stimulated DCs induce CD8 T cell proliferation, activation marker (CD44 and CD69) expression, and production of IFN-γ, IL-2, and granzyme B. Moreover, we show that type I IFNs support robust CD8 T cell activation (proliferation and IFN-γ and granzyme B production) by β-glucan-stimulated DCs in vitro and in vivo due to autocrine effects on the DCs. Specifically, type I IFNs promote Ag presentation on MHC I molecules, CD86 and CD40 expression, and the production of IL-12 p70, IL-2, IL-6, and TNF-α by β-glucan-stimulated DCs. We also demonstrate a role for autocrine type I IFN signaling in bacterial LPS-induced DC maturation, although, in the context of LPS stimulation, this mechanism is not so critical for CD8 T cell activation (promotes IFN-γ production but not proliferation or granzyme B production). This study provides insight into the mechanisms underlying CD8 T cell activation during infection, which may be useful in the rational design of vaccines directed against pathogens and tumors.
Collapse
Affiliation(s)
- Nargess Hassanzadeh-Kiabi
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048.,Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Alberto Yáñez
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048.,Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ivy Dang
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048.,Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Gislâine A Martins
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048.,F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - David M Underhill
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048.,F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Helen S Goodridge
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; .,Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048.,F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and.,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
27
|
Shakya AK, Chowdhury MYE, Tao W, Gill HS. Mucosal vaccine delivery: Current state and a pediatric perspective. J Control Release 2016; 240:394-413. [PMID: 26860287 PMCID: PMC5381653 DOI: 10.1016/j.jconrel.2016.02.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/21/2016] [Accepted: 02/05/2016] [Indexed: 12/30/2022]
Abstract
Most childhood infections occur via the mucosal surfaces, however, parenterally delivered vaccines are unable to induce protective immunity at these surfaces. In contrast, delivery of vaccines via the mucosal routes can allow antigens to interact with the mucosa-associated lymphoid tissue (MALT) to induce both mucosal and systemic immunity. The induced mucosal immunity can neutralize the pathogen on the mucosal surface before it can cause infection. In addition to reinforcing the defense at mucosal surfaces, mucosal vaccination is also expected to be needle-free, which can eliminate pain and the fear of vaccination. Thus, mucosal vaccination is highly appealing, especially for the pediatric population. However, vaccine delivery across mucosal surfaces is challenging because of the different barriers that naturally exist at the various mucosal surfaces to keep the pathogens out. There have been significant developments in delivery systems for mucosal vaccination. In this review we provide an introduction to the MALT, highlight barriers to vaccine delivery at different mucosal surfaces, discuss different approaches that have been investigated for vaccine delivery across mucosal surfaces, and conclude with an assessment of perspectives for mucosal vaccination in the context of the pediatric population.
Collapse
Affiliation(s)
| | | | - Wenqian Tao
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
28
|
Hang L, Blum AM, Kumar S, Urban JF, Mitreva M, Geary TG, Jardim A, Stevenson MM, Lowell CA, Weinstock JV. Downregulation of the Syk Signaling Pathway in Intestinal Dendritic Cells Is Sufficient To Induce Dendritic Cells That Inhibit Colitis. THE JOURNAL OF IMMUNOLOGY 2016; 197:2948-57. [PMID: 27559049 DOI: 10.4049/jimmunol.1600063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/14/2016] [Indexed: 12/21/2022]
Abstract
Helminthic infections modulate host immunity and may protect people in less-developed countries from developing immunological diseases. In a murine colitis model, the helminth Heligmosomoides polygyrus bakeri prevents colitis via induction of regulatory dendritic cells (DCs). The mechanism driving the development of these regulatory DCs is unexplored. There is decreased expression of the intracellular signaling pathway spleen tyrosine kinase (Syk) in intestinal DCs from H. polygyrus bakeri-infected mice. To explore the importance of this observation, it was shown that intestinal DCs from DC-specific Syk(-/-) mice were powerful inhibitors of murine colitis, suggesting that loss of Syk was sufficient to convert these cells into their regulatory phenotype. DCs sense gut flora and damaged epithelium via expression of C-type lectin receptors, many of which signal through the Syk signaling pathway. It was observed that gut DCs express mRNA encoding for C-type lectin (CLEC) 7A, CLEC9A, CLEC12A, and CLEC4N. H. polygyrus bakeri infection downmodulated CLEC mRNA expression in these cells. Focusing on CLEC7A, which encodes for the dectin-1 receptor, flow analysis showed that H. polygyrus bakeri decreases dectin-1 expression on the intestinal DC subsets that drive Th1/Th17 development. DCs become unresponsive to the dectin-1 agonist curdlan and fail to phosphorylate Syk after agonist stimulation. Soluble worm products can block CLEC7A and Syk mRNA expression in gut DCs from uninfected mice after a brief in vitro exposure. Thus, downmodulation of Syk expression and phosphorylation in intestinal DCs could be important mechanisms through which helminths induce regulatory DCs that limit colitis.
Collapse
Affiliation(s)
- Long Hang
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111
| | - Arthur M Blum
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111
| | - Sangeeta Kumar
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111
| | - Joseph F Urban
- Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705
| | - Makedonka Mitreva
- Genome Institute, Washington University School of Medicine, St. Louis, MO 63108
| | - Timothy G Geary
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec H9X 3V9, Canada
| | - Armando Jardim
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec H9X 3V9, Canada
| | - Mary M Stevenson
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada; Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada; and
| | - Clifford A Lowell
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143
| | - Joel V Weinstock
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111;
| |
Collapse
|
29
|
Agrawal S, Ganguly S, Hajian P, Cao JN, Agrawal A. PDGF upregulates CLEC-2 to induce T regulatory cells. Oncotarget 2016; 6:28621-32. [PMID: 26416420 PMCID: PMC4745681 DOI: 10.18632/oncotarget.5765] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/12/2015] [Indexed: 01/01/2023] Open
Abstract
The effect of platelet derived growth factor (PDGF) on immune cells is not elucidated. Here, we demonstrate PDGF inhibited the maturation of human DCs and induced IL-10 secretion. Culture of PDGF-DCs with T cells induced the polarization of T cells towards FoxP3 expressing T regulatory cells that secreted IL-10. Gene expression studies revealed that PDGF induced the expression of C-type lectin like receptor member 2, (CLEC-2) receptor on DCs. Furthermore, DCs transfected with CLEC-2 induced T regulatory cells in DC-T cell co-culture. CLEC-2 is naturally expressed on platelets. Therefore, to confirm whether CLEC-2 is responsible for inducing the T regulatory cells, T cells were cultured with either CLEC-2 expressing platelets or soluble CLEC-2. Both conditions resulted in the induction of regulatory T cells. The generation of T regulatory cells was probably due to the binding of CLEC-2 with its ligand podoplanin on T cells, since crosslinking of podoplanin on the T cells also resulted in the induction of T regulatory cells. These data demonstrate that PDGF upregulates the expression of CLEC-2 on cells to induce T regulatory cells.
Collapse
Affiliation(s)
- Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Sreerupa Ganguly
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Pega Hajian
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Jia-Ning Cao
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
30
|
Agrawal S, Agrawal A, Said HM. Biotin deficiency enhances the inflammatory response of human dendritic cells. Am J Physiol Cell Physiol 2016; 311:C386-91. [PMID: 27413170 DOI: 10.1152/ajpcell.00141.2016] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/12/2016] [Indexed: 12/24/2022]
Abstract
The water-soluble biotin (vitamin B7) is indispensable for normal human health. The vitamin acts as a cofactor for five carboxylases that are critical for fatty acid, glucose, and amino acid metabolism. Biotin deficiency is associated with various diseases, and mice deficient in this vitamin display enhanced inflammation. Previous studies have shown that biotin affects the functions of adaptive immune T and NK cells, but its effect(s) on innate immune cells is not known. Because of that and because vitamins such as vitamins A and D have a profound effect on dendritic cell (DC) function, we investigated the effect of biotin levels on the functions of human monocyte-derived DCs. Culture of DCs in a biotin-deficient medium (BDM) and subsequent activation with LPS resulted in enhanced secretion of the proinflammatory cytokines TNF-α, IL-12p40, IL-23, and IL-1β compared with LPS-activated DCs cultured in biotin-sufficient (control) and biotin-oversupplemented media. Furthermore, LPS-activated DCs cultured in BDM displayed a significantly higher induction of IFN-γ and IL-17 indicating Th1/Th17 bias in T cells compared with cells maintained in biotin control or biotin-oversupplemented media. Investigations into the mechanisms suggested that impaired activation of AMP kinase in DCs cultured in BDM may be responsible for the observed increase in inflammatory responses. In summary, these results demonstrate for the first time that biotin deficiency enhances the inflammatory responses of DCs. This may therefore be one of the mechanism(s) that mediates the observed inflammation that occurs in biotin deficiency.
Collapse
Affiliation(s)
- Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, California
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, California;
| | - Hamid M Said
- Department of Medicine, Department of Physiology and Biophysics, University of California, Irvine, California; and Veterans Affairs Medical Center-151, Long Beach, California
| |
Collapse
|
31
|
Ghoneum M, Felo N, Agrawal S, Agrawal A. A novel kefir product (PFT) activates dendritic cells to induce CD4+T and CD8+T cell responses in vitro. Int J Immunopathol Pharmacol 2015; 28:488-96. [PMID: 26384392 DOI: 10.1177/0394632015599447] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Lactobacilli have been widely studied for their probiotic effects and have been reported to function as antiviral and anticancer agents. However, the underlying mechanisms via immune modulation are poorly understood. PFT is a freeze dried compound of Lactobacillus kefiri P-IF with a unique composition and functionality. In this study, we examined the potential stimulatory effects of two concentrations (50 µg and 100 µg/mL) of PFT on human monocyte-derived dendritic cell (DC) function in vitro. Results showed that PFT upregulated the expression of DC surface co-stimulatory and maturation markers CD80, CD86, and HLADR in a concentration dependent manner. PFT at 100 µg/mL markedly increased the secretion of IL-6, IL-10, TNF-α, and IL-1β by DCs. This concentration of PFT also stimulated the production of antiviral cytokines, IFN-α and IFN-λ(IL29) in DCs. Additionally, PFT at 100 µg/mL activated moDCs prime CD4(+)T cells and significantly increased the levels of IL-10, IFN-γ, and TNF-α by 1.7, four, three-fold, respectively. Furthermore PFT-stimulated DCs were also effective in enhancing the cytotoxic potential of CD8(+)T cells via the induction of Granzyme-B and upregulation of CD107a, and CD103 expression, a marker of resident/regulatory CD8(+)T cells. These data suggest that PFT functions as a natural adjuvant for DC activation and thus may be used in DC-based vaccine strategies against viral infections and cancer.
Collapse
Affiliation(s)
- Mamdooh Ghoneum
- Department of Otolaryngology, Charles Drew University of Medicine and Science, 1621 E. 120th Street, Los Angeles, CA 90059, USA
| | - Nouran Felo
- Department of Otolaryngology, Charles Drew University of Medicine and Science, 1621 E. 120th Street, Los Angeles, CA 90059, USA
| | - Sudhanshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California Irvine, Irvine, CA 92697
| | - Anshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California Irvine, Irvine, CA 92697
| |
Collapse
|
32
|
Rainard P, Cunha P, Ledresseur M, Staub C, Touzé JL, Kempf F, Gilbert FB, Foucras G. Antigen-Specific Mammary Inflammation Depends on the Production of IL-17A and IFN-γ by Bovine CD4+ T Lymphocytes. PLoS One 2015; 10:e0137755. [PMID: 26375594 PMCID: PMC4573518 DOI: 10.1371/journal.pone.0137755] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/20/2015] [Indexed: 12/03/2022] Open
Abstract
Intramammary infusion of the antigen used to sensitize cows by the systemic route induces a local inflammation associated with neutrophil recruitment. We hypothesize that this form of delayed type hypersensitivity, which may occur naturally during infections or could be induced intentionally by vaccination, can impact the outcome of mammary gland infections. We immunized cows with ovalbumin to identify immunological correlates of antigen-specific mammary inflammation. Intraluminal injection of ovalbumin induced a mastitis characterized by a prompt tissue reaction (increase in teat wall thickness) and an intense influx of leukocytes into milk of 10 responder cows out of 14 immunized animals. The magnitude of the local inflammatory reaction, assessed through milk leukocytosis, correlated with antibody titers, skin thickness test, and production of IL-17A and IFN-γ in a whole-blood antigen stimulation assay (WBA). The production of these two cytokines significantly correlated with the magnitude of the milk leukocytosis following the ovalbumin intramammary challenge. The IL-17A and IFN-γ production in the WBA was dependent on the presence of CD4+ cells in blood samples. In vitro stimulation of peripheral blood lymphocytes with ovalbumin followed by stimulation with PMA/ionomycin allowed the identification by flow cytometry of CD4+ T cells producing either IL-17A, IFN-γ, or both cytokines. The results indicate that the antigen-specific WBA, and specifically IL-17A and IFN-γ production by circulating CD4+ cells, can be used as a predictor of mammary hypersensitivity to protein antigens. This prompts further studies aiming at determining how Th17 and/or Th1 lymphocytes modulate the immune response of the mammary gland to infection.
Collapse
Affiliation(s)
- Pascal Rainard
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
- * E-mail:
| | - Patricia Cunha
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Marion Ledresseur
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Christophe Staub
- UE1297, Unité Expérimentale de Physiologie Animale, UEPAO, INRA, Nouzilly, France
| | - Jean-Luc Touzé
- Université François Rabelais de Tours, Tours, France
- UMR0085, Physiologie de la Reproduction et du Comportement, INRA, Nouzilly, France
| | - Florent Kempf
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Florence B. Gilbert
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Gilles Foucras
- INP, ENVT, Université de Toulouse, Toulouse, France
- UMR1225, Interactions Hôte Agents Pathogènes, INRA, Toulouse, France
| |
Collapse
|
33
|
Jiang N, Zhao G, Lin J, Hu L, Che C, Li C, Wang Q, Xu Q, Peng X. Indoleamine 2,3-Dioxygenase Is Involved in the Inflammation Response of Corneal Epithelial Cells to Aspergillus fumigatus Infections. PLoS One 2015; 10:e0137423. [PMID: 26361229 PMCID: PMC4567309 DOI: 10.1371/journal.pone.0137423] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 08/17/2015] [Indexed: 01/09/2023] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO), which is mainly expressed in activated dendritic cells, is known as a regulator of immune responses. However, the role of IDO in immune responses against fungal corneal infection has not been investigated. To evaluate the regulatory mechanisms of IDO in fungal inflammation, we resorted to human corneal epithelial cells (HCECs), known as the first barrier of cornea against pathogenic microorganisms. We found that IDO was significantly up-regulated in corneal epithelium infected with Aspergillus fumigatus (A. fumigatus) and HCECs incubated with spores of A. fumigatus. Furthermore, IDO inhibitor (1-methyltryptophan, 1-MT) enhanced inflammatory cytokines IL-1β and IL-6 expression which were up-regulated by A. fumigatus spores infection. Dectin-1, as one of the important C-type lectin receptors, can identify β-glucan, and mediate fungal innate immune responses. In the present study, pre-treatment with curdlan, a Dectin-1 agonist, further enhanced IDO expression compared with A. fumigatus stimulation. While laminarin, the Dectin-1 specific inhibitor, partially inhibited IDO expression stimulated by A. fumigatus. Further studies demonstrated inhibition of IDO activity amplified the expressions of inflammatory cytokines IL-1β and IL-6 induced by activation of Dectin-1. These results suggested that IDO was involved in the immune responses of fungal keratitis. The activation of Dectin-1 may contribute to A. fumigatus spores-induced up-regulation of IDO.
Collapse
MESH Headings
- Animals
- Aspergillus fumigatus/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Epithelium, Corneal/metabolism
- Epithelium, Corneal/microbiology
- Epithelium, Corneal/pathology
- Female
- Gene Expression
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Inflammation Mediators/metabolism
- Keratitis/diagnosis
- Keratitis/metabolism
- Keratitis/microbiology
- Lectins, C-Type/metabolism
- Mice
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- beta-Glucans/pharmacology
Collapse
Affiliation(s)
- Nan Jiang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Guiqiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
- * E-mail:
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Liting Hu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Chengye Che
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Cui Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Qian Wang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Qiang Xu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Xudong Peng
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| |
Collapse
|
34
|
Feriotti C, Bazan SB, Loures FV, Araújo EF, Costa TA, Calich VLG. Expression of dectin-1 and enhanced activation of NALP3 inflammasome are associated with resistance to paracoccidioidomycosis. Front Microbiol 2015; 6:913. [PMID: 26388856 PMCID: PMC4558525 DOI: 10.3389/fmicb.2015.00913] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/20/2015] [Indexed: 02/02/2023] Open
Abstract
Dectin-1 is a pattern recognition receptor (PRR) that recognizes β-glucans and plays a major role in the immunity against fungal pathogens. Paracoccidioides brasiliensis, the causative agent of paracoccidioidomycosis, has a sugar-rich cell wall mainly composed of mannans and glucans. To investigate the role of dectin-1 in the innate immunity of resistant (A/J) and susceptible (B10.A) mice to P. brasiliensis infection, we evaluated the role of curdlan (a dectin-1 agonist) and laminarin (a dectin-1 antagonist) in the activation of macrophages from both mouse strains. We verified that curdlan has a negligible role in the activation of B10.A macrophages but enhances the phagocytic and fungicidal abilities of A/J macrophages. Curdlan up-regulated the expression of costimulatory molecules and PRRs in A/J macrophages that express elevated levels of dectin-1, but not in B10.A cells. In addition, curdlan treatment inhibited arginase-1 and enhanced NO-synthase mRNA expression in infected A/J macrophages but had not effect in B10.A cells. In contrast, laminarin reinforced the respective M2/M1 profiles of infected A/J and B10.A macrophages. Following curdlan treatment, A/J macrophages showed significantly higher Syk kinase phosphorylation and expression of intracellular pro-IL-1β than B10.A cells. These findings led us to investigate if the NRLP3 inflammasome was differently activated in A/J and B10.A cells. Indeed, compared with B10.A cells A/J macrophages showed an increased expression of NALP3, ASC, and IL-1β mRNA. They also showed elevated caspase-1 activity and secreted high levels of mature IL-β and IL-18 after curdlan treatment and P. brasiliensis infection. Our data demonstrate that soluble and particulate β-glucans exert opposed modulatory activities on macrophages of diverse genetic patterns. Moreover, the synergistic action of dectin-1 and NALP3 inflammasome were for the first time associated with the innate response of resistant hosts to P. brasiliensis infection.
Collapse
Affiliation(s)
- Claudia Feriotti
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo São Paulo, Brazil
| | - Silvia B Bazan
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo São Paulo, Brazil
| | - Flávio V Loures
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo São Paulo, Brazil
| | - Eliseu F Araújo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo São Paulo, Brazil
| | - Tânia A Costa
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo São Paulo, Brazil
| | - Vera L G Calich
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo São Paulo, Brazil
| |
Collapse
|
35
|
Abstract
Scavenger receptors constitute a large family of evolutionally conserved protein molecules that are structurally and functionally diverse. Although scavenger receptors were originally identified based on their capacity to scavenge modified lipoproteins, these molecules have been shown to recognize and bind to a broad spectrum of ligands, including modified and unmodified host-derived molecules or microbial components. As a major subset of innate pattern recognition receptors, scavenger receptors are mainly expressed on myeloid cells and function in a wide range of biological processes, such as endocytosis, adhesion, lipid transport, antigen presentation, and pathogen clearance. In addition to playing a crucial role in maintenance of host homeostasis, scavenger receptors have been implicated in the pathogenesis of a number of diseases, e.g., atherosclerosis, neurodegeneration, or metabolic disorders. Emerging evidence has begun to reveal these receptor molecules as important regulators of tumor behavior and host immune responses to cancer. This review summarizes our current understanding on the newly identified, distinct functions of scavenger receptors in cancer biology and immunology. The potential of scavenger receptors as diagnostic biomarkers and novel targets for therapeutic interventions to treat malignancies is also highlighted.
Collapse
Affiliation(s)
- Xiaofei Yu
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - John R Subjeck
- Department of Cellular Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA.
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
36
|
Raïch-Regué D, Rosborough BR, Watson AR, McGeachy MJ, Turnquist HR, Thomson AW. mTORC2 Deficiency in Myeloid Dendritic Cells Enhances Their Allogeneic Th1 and Th17 Stimulatory Ability after TLR4 Ligation In Vitro and In Vivo. THE JOURNAL OF IMMUNOLOGY 2015; 194:4767-76. [PMID: 25840913 DOI: 10.4049/jimmunol.1402551] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 03/09/2015] [Indexed: 01/29/2023]
Abstract
The mammalian/mechanistic target of rapamycin (mTOR) is a key integrative kinase that functions in two independent complexes, mTOR complex (mTORC) 1 and mTORC2. In contrast to the well-defined role of mTORC1 in dendritic cells (DC), little is known about the function of mTORC2. In this study, to our knowledge, we demonstrate for the first time an enhanced ability of mTORC2-deficient myeloid DC to stimulate and polarize allogeneic T cells. We show that activated bone marrow-derived DC from conditional Rictor(-/-) mice exhibit lower coinhibitory B7-H1 molecule expression independently of the stimulus and enhanced IL-6, TNF-α, IL-12p70, and IL-23 production following TLR4 ligation. Accordingly, TLR4-activated Rictor(-/-) DC display augmented allogeneic T cell stimulatory ability, expanding IFN-γ(+) and IL-17(+), but not IL-10(+) or CD4(+)Foxp3(+) regulatory T cells in vitro. A similar DC profile was obtained by stimulating Dectin-1 (C-type lectin family member) on Rictor(-/-) DC. Using novel CD11c-specific Rictor(-/-) mice, we confirm the alloreactive Th1 and Th17 cell-polarizing ability of endogenous mTORC2-deficient DC after TLR4 ligation in vivo. Furthermore, we demonstrate that proinflammatory cytokines produced by Rictor(-/-) DC after LPS stimulation are key in promoting Th1/Th17 responses. These data establish that mTORC2 activity restrains conventional DC proinflammatory capacity and their ability to polarize T cells following TLR and non-TLR stimulation. Our findings provide new insight into the role of mTORC2 in regulating DC function and may have implications for emerging therapeutic strategies that target mTOR in cancer, infectious diseases, and transplantation.
Collapse
Affiliation(s)
- Dàlia Raïch-Regué
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Brian R Rosborough
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Alicia R Watson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Mandy J McGeachy
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Hēth R Turnquist
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
37
|
Loures FV, Araújo EF, Feriotti C, Bazan SB, Calich VLG. TLR-4 cooperates with Dectin-1 and mannose receptor to expand Th17 and Tc17 cells induced by Paracoccidioides brasiliensis stimulated dendritic cells. Front Microbiol 2015; 6:261. [PMID: 25873917 PMCID: PMC4379960 DOI: 10.3389/fmicb.2015.00261] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/16/2015] [Indexed: 01/21/2023] Open
Abstract
The concomitant use of diverse pattern recognition receptors (PRRs) by innate immune cells can result in synergistic or inhibitory activities that profoundly influence anti-microbial immunity. Dectin-1 and the mannose receptor (MR) are C-type lectin receptors (CLRs) previously reported to cooperate with toll-like receptors (TLRs) signaling in the initial inflammatory response and in the induction of adaptive Th17 and Tc17 immunity mediated by CD4(+) and CD8(+) T cells, respectively. The protective immunity against paracoccidioidomycosis, the most prevalent fungal infection of Latin America, was previously shown to be influenced by these T cell subsets motivating us to study the contribution of TLRs, Dectin-1, and MR to the development of Th17/Tc17 immunity. First, curdlan a specific Dectin-1 agonist was used to characterize the influence of this receptor in the proliferative response and Th17/Tc17 differentiation of naïve lymphocytes induced by Paracoccidioides brasiliensis activated dendritic cells (DCs) from C57BL/6 mice. Then, wild type (WT), Dectin-1(-/-), TLR-2(-/-), and TLR-4(-/-) DCs treated or untreated with anti-Dectin-1 and anti-MR antibodies were used to investigate the contribution of these receptors in lymphocyte activation and differentiation. We verified that curdlan induces an enhanced lymphocyte proliferation and development of IL-17 producing CD4(+) and CD8(+) T cells. In addition, treatment of WT, TLR-2(-/-), and TLR-4(-/-) DCs by anti-Dectin-1 antibodies or antigen presentation by Dectin-1(-/-) DCs led to decreased lymphoproliferation and impaired Th17 and Tc17 expansion. These responses were also inhibited by anti-MR treatment of DCs, but a synergistic action on Th17/Tc17 differentiation was mediated by TLR-4 and MR. Taken together, our results indicate that diverse TLRs and CLRs are involved in the induction of lymphocyte proliferation and Th17/Tc17 differentiation mediated by P. brasiliensis activated DCs, but a synergist action was restricted to Dectin-1, TLR-4, and MR.
Collapse
Affiliation(s)
- Flávio V Loures
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Eliseu F Araújo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Claudia Feriotti
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Silvia B Bazan
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| | - Vera L G Calich
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, Brazil
| |
Collapse
|
38
|
Aref AM, Hoa NT, Ge L, Agrawal A, Dacosta-Iyer M, Lambrecht N, Ouyang Y, Cornforth AN, Jadus MR. HCA519/TPX2: a potential T-cell tumor-associated antigen for human hepatocellular carcinoma. Onco Targets Ther 2014; 7:1061-70. [PMID: 24966688 PMCID: PMC4063820 DOI: 10.2147/ott.s61442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Immunotherapy for human hepatocellular cancer (HCC) is slowly making progress towards treating these fatal cancers. The identification of new antigens can improve this approach. We describe a possible new antigen, hepatocellular carcinoma‐associated antigen‐519/targeting protein for Xklp‐2 (HCA519/TPX2), for HCC that might be beneficial for T‐cell specific HCC immunotherapy. Methods HCC was studied for the expression for 15 tumor‐associated antigens considered useful for immunotherapy within three HCC cell lines (HepG2, Hep3B, and PLC/PRF/5), lymphocytes, non‐cancerous livers, and clinical HCC. The expression of tumor antigenic precursor proteins (TAPPs) messenger RNA was first screened by reverse transcriptase quantitative real‐time polymerase chain reaction. Results Four antigens (alpha fetoprotein, aspartyl/asparaginyl βhydroxylase, glypican3 and HCA519/TPX2) proved to be the best expressed TAPPs within the HCC specimens by molecular analyses. HCA519/TPX2 was detected by intracellular cell flow cytometry within HCC cell lines by using a specific antibody towards this TAPP. This antibody also detected the protein within primary HCCs. We synthesized two HCA519/TPX2 peptides (HCA519464–472 and HCA519351–359) which can bind to human leukocyte antigen (HLA)‐A*0201. Dendritic cells pulsed with these peptides stimulated cytolytic T lymphocytes (CTLs). These killer T‐cells lysed HLA‐A*0201+ T2 cells exogenously loaded with the correct specific peptide. The CTLs killed HepG2 (HLA‐A2+ and HCA519+), but not the Hep3B and PLC/PRF/5 cell lines, which are HCA519+ but HLA‐A2‐negative. In silico analysis reveals that HCA519/TPX2 has the inherent ability to bind to a very wide variety of HLA antigens. Conclusion HCA519/TPX2 is a viable immunotarget that should be further investigated within HCC patients.
Collapse
Affiliation(s)
- Ahmed M Aref
- Biological Science Department, Modern Sciences and Arts University, Faculty of Dentistry, Cairo, Egypt ; Southern California Institute for Research and Education, Veterans Affairs Medical Center, Long Beach, CA, USA ; Research Health Care Group, Veterans Affairs Medical Center Long Beach, CA, USA
| | - Neil T Hoa
- Research Health Care Group, Veterans Affairs Medical Center Long Beach, CA, USA
| | - Lisheng Ge
- Research Health Care Group, Veterans Affairs Medical Center Long Beach, CA, USA
| | - Anshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California, Irvine, CA, USA
| | - Maria Dacosta-Iyer
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Nils Lambrecht
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Yi Ouyang
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | | | - Martin R Jadus
- Pathology and Laboratory Medicine Department, Veterans Affairs Medical Center Long Beach, CA, USA ; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA ; Neuro-Oncology Program, Chao Comprehensive Cancer Center, University of California, Irvine, CA, USA
| |
Collapse
|
39
|
Loures FV, Araújo EF, Feriotti C, Bazan SB, Costa TA, Brown GD, Calich VLG. Dectin-1 induces M1 macrophages and prominent expansion of CD8+IL-17+ cells in pulmonary Paracoccidioidomycosis. J Infect Dis 2014; 210:762-73. [PMID: 24604821 DOI: 10.1093/infdis/jiu136] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dectin-1, the innate immune receptor that recognizes β-glucan, plays an important role in immunity against fungal pathogens. Paracoccidioides brasiliensis, the etiological agent of paracoccidioidomycosis, has a sugar-rich cell wall mainly composed of mannans and glucans. This fact motivated us to use dectin-1-sufficient and -deficient mice to investigate the role of β-glucan recognition in the immunity against pulmonary paracoccidioidomycosis. Initially, we verified that P. brasiliensis infection reinforced the tendency of dectin-1-deficient macrophages to express an M2 phenotype. This prevalent antiinflammatory activity of dectin-1(-/-) macrophages resulted in impaired fungicidal ability, low nitric oxide production, and elevated synthesis of interleukin 10 (IL-10). Compared with dectin-1-sufficient mice, the fungal infection of dectin-1(-/-) mice was more severe and resulted in enhanced tissue pathology and mortality rates. The absence of dectin-1 has also impaired the production of T-helper type 1 (Th1), Th2, and Th17 cytokines and the activation and migration of T cells to the site of infection. Remarkably, dectin-1 deficiency increased the expansion of regulatory T cells and reduced the differentiation of T cells to the IL-17(+) phenotype, impairing the migration of IL-17(+)CD8(+) T cells and polymorphonuclear cells to infected tissues. In conclusion, dectin-1 exerts an important protective role in pulmonary paracoccidioidomycosis by controlling the innate and adaptive phases of antifungal immunity.
Collapse
Affiliation(s)
- Flávio V Loures
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Eliseu F Araújo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Claudia Feriotti
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Silvia B Bazan
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Tânia A Costa
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Gordon D Brown
- Aberdeen Fungal Group, Section of Immunity and Infection, Institute of Medical Sciences, University of Aberdeen, United Kingdom
| | - Vera L G Calich
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| |
Collapse
|
40
|
Wu TC, Xu K, Banchereau R, Marches F, Yu CI, Martinek J, Anguiano E, Pedroza-Gonzalez A, Snipes GJ, O'Shaughnessy J, Nishimura S, Liu YJ, Pascual V, Banchereau J, Oh S, Palucka K. Reprogramming tumor-infiltrating dendritic cells for CD103+ CD8+ mucosal T-cell differentiation and breast cancer rejection. Cancer Immunol Res 2014; 2:487-500. [PMID: 24795361 DOI: 10.1158/2326-6066.cir-13-0217] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Our studies showed that tumor-infiltrating dendritic cells (DC) in breast cancer drive inflammatory Th2 (iTh2) cells and protumor inflammation. Here, we show that intratumoral delivery of the β-glucan curdlan, a ligand of dectin-1, blocks the generation of iTh2 cells and prevents breast cancer progression in vivo. Curdlan reprograms tumor-infiltrating DCs via the ligation of dectin-1, enabling the DCs to become resistant to cancer-derived thymic stromal lymphopoietin (TSLP), to produce IL-12p70, and to favor the generation of Th1 cells. DCs activated via dectin-1, but not those activated with TLR-7/8 ligand or poly I:C, induce CD8+ T cells to express CD103 (αE integrin), a ligand for cancer cells, E-cadherin. Generation of these mucosal CD8+ T cells is regulated by DC-derived integrin αvβ8 and TGF-β activation in a dectin-1-dependent fashion. These CD103+ CD8+ mucosal T cells accumulate in the tumors, thereby increasing cancer necrosis and inhibiting cancer progression in vivo in a humanized mouse model of breast cancer. Importantly, CD103+ CD8+ mucosal T cells elicited by reprogrammed DCs can reject established cancer. Thus, reprogramming tumor-infiltrating DCs represents a new strategy for cancer rejection.
Collapse
Affiliation(s)
- Te-Chia Wu
- Authors' Affiliations: Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Agrawal S, Gollapudi S, Gupta S, Agrawal A. Dendritic cells from the elderly display an intrinsic defect in the production of IL-10 in response to lithium chloride. Exp Gerontol 2013; 48:1285-92. [PMID: 23988651 DOI: 10.1016/j.exger.2013.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 08/10/2013] [Accepted: 08/13/2013] [Indexed: 10/26/2022]
Abstract
Chronic, low grade inflammation is a characteristic of old age. Innate immune system cells such as dendritic cells (DCs) from the elderly display a pro-inflammatory phenotype associated with increased reactivity to self. Lithium is a well-established anti-inflammatory agent used in the treatment of bipolar disorders. It has also been reported to reduce inflammation in DCs. Here, we investigated whether Lithium is effective in reducing the inflammatory responses in DCs from the elderly. The effect of Lithium Chloride (LiCl) was compared on the response of TLR4 agonist, LPS and TLR2 agonist, PAM3CSK4 stimulated aged and young DCs. LiCl enhanced the production of IL-10 in LPS stimulated young DCs. However, it did not affect TNF-α and IL-6 production. In contrast, in aged DCs, LiCl reduced the secretion of TNF-α and IL-6 in LPS stimulated DCs but did not increase IL-10. LiCl had no significant effect on PAM3CSK4 responses in aged and young DCs. LiCl treated DCs also displayed differences at the level of CD4 T cell priming and polarization. LPS-stimulated young DCs reduced IFN-γ secretion and biased the Th cell response towards Th2/Treg while LiCl treated aged DCs only reduced IFN-γ secretion but did not bias the response towards Th2/Treg. In summary, our data suggests that LiCl reduces inflammation in aged and young DCs via different mechanisms. Furthermore, the effect of LiCl is different on LPS and PAM3CSK4 responses.
Collapse
Affiliation(s)
- Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA 92697, USA
| | | | | | | |
Collapse
|
42
|
Vannucci L, Krizan J, Sima P, Stakheev D, Caja F, Rajsiglova L, Horak V, Saieh M. Immunostimulatory properties and antitumor activities of glucans (Review). Int J Oncol 2013; 43:357-64. [PMID: 23739801 PMCID: PMC3775562 DOI: 10.3892/ijo.2013.1974] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/17/2013] [Indexed: 12/25/2022] Open
Abstract
New foods and natural biological modulators have recently become of scientific interest in the investigation of the value of traditional medical therapeutics. Glucans have an important part in this renewed interest. These fungal wall components are claimed to be useful for various medical purposes and they are obtained from medicinal mushrooms commonly used in traditional Oriental medicine. The immunotherapeutic properties of fungi extracts have been reported, including the enhancement of anticancer immunity responses. These properties are principally related to the stimulation of cells of the innate immune system. The discovery of specific receptors for glucans on dendritic cells (dectin-1), as well as interactions with other receptors, mainly expressed by innate immune cells (e.g., Toll-like receptors, complement receptor-3), have raised new attention toward these products as suitable therapeutic agents. We briefly review the characteristics of the glucans from mycelial walls as modulators of the immunity and their possible use as antitumor treatments.
Collapse
Affiliation(s)
- Luca Vannucci
- Laboratory of Immunotherapy, Department of Immunology and Gnotobiology, Institute of Microbiology, Academy of Sciences of Czech Republic, v.v.i., 142 20 Prague 4, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Chandra LC, Traoré D, French C, Marlow D, D’Offay J, Clarke SL, Smith BJ, Kuvibidila S. White button, portabella, and shiitake mushroom supplementation up-regulates interleukin-23 secretion in acute dextran sodium sulfate colitis C57BL/6 mice and murine macrophage J.744.1 cell line. Nutr Res 2013; 33:388-96. [DOI: 10.1016/j.nutres.2013.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 02/13/2013] [Accepted: 02/20/2013] [Indexed: 01/25/2023]
|
44
|
Agrawal A, Su H, Chen J, Osann K, Agrawal S, Gupta S. Increased IL-21 secretion by aged CD4+T cells is associated with prolonged STAT-4 activation and CMV seropositivity. Aging (Albany NY) 2013; 4:648-59. [PMID: 23064011 PMCID: PMC3492228 DOI: 10.18632/aging.100490] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Advancing age leads to significant decline in immune functions. IL-21 is produced primarily by T follicular helper (Tfh) cells and is required for effective immune cell functions. Here we compared the induction of IL-21 in aged and young subjects. Our investigation demonstrates that CD4+T cells from healthy elderly individuals (age ≥ 65) secreted significantly higher levels of IL-21 on priming with aged and young dendritic cells (DC). Though the aged and young DCs secreted comparable levels of IL-12 on stimulation with anti-CD40 antibody and LPS, culture of DCs with aged CD4+ T cells resulted in increased production of IL-21 as compared to that with young CD4+ T cells. Further examination revealed that the response of aged naïve CD4+ T cells to IL-12 was altered, resulting in increased differentiation of aged Th cells towards Tfh cells. Investigation into the signaling mechanism suggested that phosphorylation of STAT-4 in response to IL-12 was sustained for a longer duration in aged CD4+ T cells as compared to CD4+ T cells from young subjects. Additional analysis demonstrated that increased IL-21 secretion correlated with chronic CMV infection in aged subjects. These findings indicate that chronic CMV infection alters the response of aged CD4+ T cells to IL-12 resulting in an increased secretion of IL-21 and that aging affects Tfh cell responses in humans which may contribute to age-associated inflammation and immune dysfunctions.
Collapse
Affiliation(s)
- Anshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California, Irvine, CA 92617, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Ghosh K, Sharma G, Saha A, Kar S, Das PK, Ukil A. Successful therapy of visceral leishmaniasis with curdlan involves T-helper 17 cytokines. J Infect Dis 2013; 207:1016-25. [PMID: 23255562 DOI: 10.1093/infdis/jis771] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
The aim of this study was to evaluate and characterize the therapeutic potential of curdlan, a naturally occurring β-glucan immunomodulator, against visceral leishmaniasis, a fatal parasitic disease. Curdlan eliminated the liver and spleen parasite burden in a 45-day BALB/c mouse model of visceral leishmaniasis at a dosage of 10 mg/kg/day as determined by Giemsa-stained organ impression smears. Curdlan was associated with production of the disease-resolving T-helper (Th) 1 and Th17-inducing cytokines interleukin (IL)-6, IL-1β, and IL-23, as well as with production of Th17 cytokines IL-17 and IL-22, as determined by enzyme-linked immunosorbent assay (ELISA) and real time polymerase chain reaction (RT-PCR). Reversal of curdlan-mediated protection by anti-IL-17 and anti-IL-23 monoclonal antibodies showed the importance of Th17 cytokines. Significantly decreased production of both IL-17 and IL-22 by mice that received anti-IL-23 antibody suggested the essential role of IL-23 in Th17 differentiation. Although administration of recombinant IL-17 or IL-23 caused significant suppression of the organ parasite burden, with marked generation of interferon γ and nitric oxide (NO), effects were much faster for IL-17. These results documented that although both IL-23 and IL-17 play major roles in the antileishmanial effect of curdlan, the effect of IL-23 may occur indirectly, through the induction of IL-17 production.
Collapse
Affiliation(s)
- Kuntal Ghosh
- Department of Biochemistry, Calcutta University, Kolkata, India
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
We assessed the role of Dectin-1 in the immune response to the pathogenic fungus Coccidioides, both in vitro and in vivo, using mice with a targeted mutation in Clec7a. Elicited peritoneal macrophages responded to formalin-killed spherules (FKS) and alkali-treated FKS by secreting proinflammatory cytokines in a Dectin-1- and β-glucan-dependent manner. The responses of bone marrow-derived dendritic cells (BMDC) to the same stimulants were more complex; interleukin 1β (IL-1β) and tumor necrosis factor alpha (TNF-α) secretion was independent of Dectin-1, while IL-6, IL-10, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were largely but not entirely dependent on Dectin-1. After intranasal infection, Dectin-1(-/-) mice had lower concentrations of IL-12p70, gamma interferon (IFN-γ), IL-1β, and the Th17 cytokines IL-22, IL-23, and 17A in the lung lavage fluid, which may explain why they were significantly more susceptible to pulmonary coccidioidomycosis two weeks after infection. The Dectin-1 mutation was even more deleterious in (B6 × DBA/2)F(2) mice, which are more resistant to coccidioidomycosis than B6 mice by virtue of protective genes from DBA/2, a genetically resistant strain. We also found that two susceptible strains of mice (B6 and BALB/c) expressed much less Dectin-1 in their lungs than did resistant DBA/2 mice. We conclude that Dectin-1 is necessary for resistance to Coccidioides immitis, that Dectin-1 promotes both Th1 and Th17 protective immune responses to this infection, and that there is a correlation between expression of Dectin-1 by the inflammatory infiltrate and resistance to coccidioidomycosis. IMPORTANCE Coccidioidomycosis is a fungal infection endemic in the southwestern United States and neighboring Mexico, causing ~150,000 lung infections in people and resulting in ~17,000 hospitalizations annually in California alone. Very little is known about innate immunity to this fungus. This paper shows that Dectin-1, the primary β-glucan receptor on myeloid cells, is required for resistance to this pathogen. Dectin-1 is part of the innate immune system, and it is needed to direct the acquired immune response toward into a pathway that will lead to macrophage activation. Lungs from infected mice lacking Dectin-1 had lower concentrations of Th1 and Th17 cytokines, two cytokine pathways that are very important for acquired T cell immunity to Coccidioides spp. This is the first demonstration that Dectin-1 is required for host resistance to a dimorphic, primary pathogenic fungus.
Collapse
|
47
|
Dendritic cell activation by polysaccharide isolated from Angelica dahurica. Food Chem Toxicol 2012; 55:241-7. [PMID: 23246459 DOI: 10.1016/j.fct.2012.12.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 12/04/2012] [Accepted: 12/05/2012] [Indexed: 12/15/2022]
Abstract
Angelica dahurica is used in functional foods for the prevention and treatment of various diseases, such as inflammation and cancer. In the present study, we examined the effect of A. dahurica polysaccharide (ADP) on dendritic cell (DC) maturation. ADP increased the expressions of CD86 and MHC-II molecules, the production of IL-12, IL-1β, and TNF-α, and allogeneic T cell activation ability of DCs, and reduced DC endocytosis. As a mechanism of action, the knockdown of TLR4 with small interfering RNA decreased the ADP-induced production of nitric oxide and IL-12 by DCs, suggesting the membrane receptor candidate of ADP. After binding to TLR4, ADP increased the phosphorylation of ERK, JNK, and p38 MAPKs, and the nuclear translocation of NF-κB p50/p65. These results indicate that ADP activates DCs through TLR4 and downstream signalings.
Collapse
|
48
|
Rhee JH, Lee SE, Kim SY. Mucosal vaccine adjuvants update. Clin Exp Vaccine Res 2012; 1:50-63. [PMID: 23596577 PMCID: PMC3623511 DOI: 10.7774/cevr.2012.1.1.50] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 05/25/2012] [Accepted: 06/15/2012] [Indexed: 12/16/2022] Open
Abstract
Mucosal vaccination, capable of inducing protective immune responses both in the mucosal and systemic immune compartments, has many advantages and is regarded as a blue ocean in the vaccine industry. Mucosal vaccines can offer lower costs, better accessability, needle-free delivery, and higher capacity of mass immunizations during pandemics. However, only very limited number of mucosal vaccines was approved for human use in the market yet. Generally, induction of immune responses following mucosal immunization requires the co-administration of appropriate adjuvants that can initiate and support the effective collaboration between innate and adaptive immunity. Classically, adjuvant researches were rather empirical than keenly scientific. However, during last several years, fundamental scientific achievements in innate immunity have been translated into the development of new mucosal adjuvants. This review focuses on recent developments in the concepts of adjuvants and innate immunity, mucosal immunity with special interest of vaccine development, and basic and applied researches in mucosal adjuvant.
Collapse
Affiliation(s)
- Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. ; Department of Microbiology and Research Institute of Vibrio Infections, Chonnam National University Medical School, Gwangju, Korea
| | | | | |
Collapse
|
49
|
Huang H, Ostroff GR, Lee CK, Agarwal S, Ram S, Rice PA, Specht CA, Levitz SM. Relative contributions of dectin-1 and complement to immune responses to particulate β-glucans. THE JOURNAL OF IMMUNOLOGY 2012; 189:312-7. [PMID: 22649195 DOI: 10.4049/jimmunol.1200603] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glucan particles (GPs) are Saccharomyces cerevisiae cell walls chemically extracted so they are composed primarily of particulate β-1,3-D-glucans. GPs are recognized by Dectin-1 and are potent complement activators. Mice immunized with Ag-loaded GPs develop robust Ab and CD4(+) T cell responses. In this study, we examined the relative contributions of Dectin-1 and complement to GP phagocytosis and Ag-specific responses to immunization with OVA encapsulated in GPs. The in vitro phagocytosis of GPs by bone marrow-derived dendritic cells was facilitated by heat-labile serum component(s) independently of Dectin-1. This enhanced uptake was not seen with serum from complement component 3 knockout (C3(-/-)) mice and was also inhibited by blocking Abs directed against complement receptor 3. After i.p. injection, percent phagocytosis of GPs by peritoneal macrophages was comparable in wild-type and Dectin-1(-/-) mice and was not inhibited by the soluble β-glucan antagonist laminarin. In contrast, a much lower percentage of peritoneal macrophages from C3(-/-) mice phagocytosed GPs, and this percentage was further reduced in the presence of laminarin. Subcutaneous immunization of wild-type, Dectin-1(-/-), and C3(-/-) mice with GP-OVA resulted in similar Ag-specific IgG(1) and IgG(2c) type Ab and CD4(+) T cell lymphoproliferative responses. Moreover, while CD4(+) Th1 and Th2 responses measured by ELISPOT assay were similar in the three mouse strains, Th17 responses were reduced in C3(-/-) mice. Thus, although Dectin-1 is necessary for optimal phagocytosis of GPs in the absence of complement, complement dominates when both an intact complement system and Dectin-1 are present. In addition, Th-skewing after GP-based immunization was altered in C3(-/-) mice.
Collapse
Affiliation(s)
- Haibin Huang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Solana R, Tarazona R, Gayoso I, Lesur O, Dupuis G, Fulop T. Innate immunosenescence: effect of aging on cells and receptors of the innate immune system in humans. Semin Immunol 2012; 24:331-41. [PMID: 22560929 DOI: 10.1016/j.smim.2012.04.008] [Citation(s) in RCA: 354] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 03/31/2012] [Accepted: 04/09/2012] [Indexed: 12/13/2022]
Abstract
Components of the innate immune response, including neutrophils and macrophages, are the first line of defense against infections. Their role is to initiate an inflammatory response, phagocyte and kill pathogens, recruit natural killer cells (NK), and facilitate the maturation and migration of dendritic cells that will initiate the adaptive immune response. Extraordinary advances have been made in the last decade on the knowledge of the receptors and mechanisms used by cells of the innate immunity not only to sense and eliminate the pathogen but also to communicate each other and collaborate with cells of adaptive immunity to mount an effective immune response. The analysis of innate immunity in elderly humans has evidenced that aging has a profound impact on the phenotype and functions of these cells. Thus altered expression and/or function of innate immunity receptors and signal transduction leading to defective activation and decreased chemotaxis, phagocytosis and intracellular killing of pathogens have been described. The phenotype and function of NK cells from elderly individuals show significant changes that are compatible with remodeling of the different NK subsets, with a decrease in the CD56bright subpopulation and accumulation of the CD56dim cells, in particular those differentiated NK cells that co-express CD57, as well as a decreased expression of activating natural cytotoxicity receptors. These alterations can be responsible of the decreased production of cytokines and the lower per-cell cytotoxicity observed in the elderly. Considering the relevance of these cells in the initiation of the immune response, the possibility to reactivate the function of innate immune cells should be considered in order to improve the response to pathogens and to vaccination in the elderly.
Collapse
Affiliation(s)
- Rafael Solana
- Immunology Unit, Instituto Maimonides para la Investigacion Biomedica de Cordoba-University of Cordoba-Hospital Reina Sofia, Cordoba, Spain.
| | | | | | | | | | | |
Collapse
|