151
|
Yu J, Chen T, Xie Z, Liang P, Qu H, Shang M, Mao Q, Ning D, Tang Z, Shi M, Zhou L, Huang Y, Yu X. Oral delivery of Bacillus subtilis spore expressing enolase of Clonorchis sinensis in rat model: induce systemic and local mucosal immune responses and has no side effect on liver function. Parasitol Res 2015; 114:2499-505. [PMID: 25877387 DOI: 10.1007/s00436-015-4449-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 03/20/2015] [Indexed: 12/17/2022]
Abstract
Caused by the consumption of raw or undercooked freshwater fish containing infective metacercariae of Clonorchis sinensis, human clonorchiasis remains a major public health problem in China. In previous study, we had expressed enolase from C. sinensis (CsENO) on the surface of Bacillus subtilis spore and the recombinant spore induced a pronounced protection in terms of reduced worm burden and eggs per gram feces, suggesting B. subtilis spore as an ideal vehicle for antigen delivery by oral treatment and CsENO as a promising vaccine candidate against clonorchiasis. In the current study, we detected CsENO-specific IgG and IgA levels both in serum and in intestinal mucus from rats orally administrated with B. subtilis spore surface expressing CsENO by ELISA. Lysozyme levels in serum and in intestinal mucus were analyzed too. In addition, IgA-secreting cells in intestine epithelium of the rats were detected by immunohistochemistry assay. The intestinal villi lengths of duodenum, jejunum, and ileum were also measured. Rats orally treated with B. subtilis spore or normal saline were used as controls. Our results showed that, compared with the control groups, oral administration of B. subtilis spore expressing CsENO induced both systemic and local mucosal immune response. The recombinant spores also enhanced non-specific immune response in rats. The spores had no side effect on liver function. Moreover, it might facilitate food utilization and digestion of the rats. Our work will pave the way to clarify the involved mechanisms of protective efficacy elicited by B. subtilis spore expressing CsENO and encourage us to carry out more assessment trails of the oral treated spore to develop vaccine against clonorchiasis.
Collapse
Affiliation(s)
- Jinyun Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Musich T, Demberg T, Morgan IL, Estes JD, Franchini G, Robert-Guroff M. Purification and functional characterization of mucosal IgA from vaccinated and SIV-infected rhesus macaques. Clin Immunol 2015; 158:127-39. [PMID: 25840105 DOI: 10.1016/j.clim.2015.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/04/2015] [Accepted: 03/24/2015] [Indexed: 12/30/2022]
Abstract
Vaccine-induced mucosal antibodies are often evaluated using small volumes of secretory fluids. However, fecal matter containing mucosal IgA is abundant. We purified fecal IgA from five SIV-vaccinated and five SIV-infected rhesus macaques by sequential affinity chromatography. The purified IgA was dimeric by native PAGE, contained secretory component, and was analogous to IgA in colostrum and vaginal fluid by western blot. IgA from one infected and four vaccinated animals neutralized H9-derived SIV(mac)251 with IC(50)s as low as 1 μg/mL. Purified IgAs inhibited transcytosis and exhibited phagocytic activity, the latter significantly correlated with SIV(mac)251 Env-specific IgA in the purified samples. Among different affinity resins, peptide M was optimal compared to jacalin, anti-monkey IgA and SSL7 for IgA purification, as confirmed using tandem peptide M/anti-monkey IgA columns. Fecal IgA provided material sufficient for several assays relevant to protective efficacy, and was shown to be multifunctional. Our approach is potentially applicable to human clinical studies.
Collapse
Affiliation(s)
| | | | | | - Jacob D Estes
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | |
Collapse
|
153
|
Moh ES, Thaysen-Andersen M, Packer NH. Relative versus absolute quantitation in disease glycomics. Proteomics Clin Appl 2015; 9:368-82. [DOI: 10.1002/prca.201400184] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 12/21/2014] [Accepted: 02/10/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Edward S.X. Moh
- Department of Chemistry and Biomolecular Sciences; Macquarie University; Sydney Australia
| | | | - Nicolle H. Packer
- Department of Chemistry and Biomolecular Sciences; Macquarie University; Sydney Australia
| |
Collapse
|
154
|
Wang J, Li Q, Xie J, Xu Y. Cigarette smoke inhibits BAFF expression and mucosal immunoglobulin A responses in the lung during influenza virus infection. Respir Res 2015; 16:37. [PMID: 25849069 PMCID: PMC4364338 DOI: 10.1186/s12931-015-0201-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/02/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND It is incompletely understood how cigarette smoke (CS) exposure affects lung mucosal immune responses during viral respiratory infections. B cell activating factor belonging to the tumor necrosis factor family (BAFF) plays an important role in the induction of secretory immunoglobulin A (S-IgA) which is the main effector of the mucosal immune system. We therefore investigated the effects of CS exposure on BAFF expression and S-IgA responses in the lung during influenza virus infection. METHODS Mice were exposed to CS and/or infected with influenza virus. Bronchoalveolar lavage fluid and lung compartments were analyzed for BAFF expression, influenza-specific S-IgA level and histological changes. Lung B cells were isolated and the activation-induced cytidine deaminase (Aicda) expression was determined. BEAS-2B cells were treated with CS extract (CSE), influenza virus, interferon beta or N-acetylcysteine and BAFF expression was measured. RESULTS CS inhibited BAFF expression in the lung, particularly after long-term exposure. BAFF and S-IgA levels were increased during influenza virus infection. Three-month CS exposure prior to influenza virus infection resulted in reduced BAFF and S-IgA levels in the lung as well as augmented pulmonary inflammation on day 7 after infection. Prior CS exposure also caused decreased Aicda expression in lung B cells during infection. Neutralization of BAFF in the lung resulted in reduced S-IgA levels during influenza virus infection. CSE inhibited virus-mediated BAFF induction in a dose-dependent manner in BEAS-2B cells, while this inhibition of BAFF by CSE was prevented by pretreatment with the antioxidant N-acetylcysteine. CONCLUSIONS Our findings indicate that CS may hinder early mucosal IgA responses in the lung during influenza virus infection through oxidative inhibition of BAFF, which might contribute to the increased incidence and severity of viral infections in smokers.
Collapse
|
155
|
Kato LM, Kawamoto S, Maruya M, Fagarasan S. The role of the adaptive immune system in regulation of gut microbiota. Immunol Rev 2015; 260:67-75. [PMID: 24942682 DOI: 10.1111/imr.12185] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gut nourishes rich bacterial communities that affect profoundly the functions of the immune system. The relationship between gut microbiota and the immune system is one of reciprocity. The microbiota contributes to nutrient processing and the development, maturation, and function of the immune system. Conversely, the immune system, particularly the adaptive immune system, plays a key role in shaping the repertoire of gut microbiota. The fitness of host immune system is reflected in the gut microbiota, and deficiencies in either innate or adaptive immunity impact on diversity and structures of bacterial communities in the gut. Here, we discuss the mechanisms that underlie this reciprocity and emphasize how the adaptive immune system via immunoglobulins (i.e. IgA) contributes to diversification and balance of gut microbiota required for immune homeostasis.
Collapse
Affiliation(s)
- Lucia M Kato
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences (IMS-RCAI), RIKEN Yokohama Institute, Yokohama, Japan
| | | | | | | |
Collapse
|
156
|
Gutzeit C, Magri G, Cerutti A. Intestinal IgA production and its role in host-microbe interaction. Immunol Rev 2015; 260:76-85. [PMID: 24942683 DOI: 10.1111/imr.12189] [Citation(s) in RCA: 223] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Complex and diverse communities of bacteria establish mutualistic and symbiotic relationships with the gut after birth. The intestinal immune system responds to bacterial colonization by acquiring a state of hypo-responsiveness against commensals and active readiness against pathogens. The resulting homeostatic balance involves a continuous dialog between the microbiota and lymphocytes with the intermediation of epithelial and dendritic cells. This dialog causes massive production of immunoglobulin A (IgA), a non-inflammatory antibody specialized in mucosal protection. Here, we discuss recent advances on the regulation of intestinal IgA responses and their role in host-microbe interaction.
Collapse
Affiliation(s)
- Cindy Gutzeit
- Immunology Institute, Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | |
Collapse
|
157
|
The higher frequency of IgA deficiency among Swedish twins is not explained by HLA haplotypes. Genes Immun 2015; 16:199-205. [PMID: 25569265 DOI: 10.1038/gene.2014.78] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 12/21/2022]
Abstract
Serum immunoglobulin A (IgA) concentrations were determined in 12 600 adult Swedish twins, applying a high-throughput reverse-phase protein microarray technique. The prevalence of IgA deficiency (IgAD) was found to be 1:241 in monozygotic (MZ) twins and 1:198 in dizygotic (DZ) twins. Hence, the prevalence in twins is markedly elevated as compared with the normal Swedish adult population (1:600). The twins did not show a difference in the frequency of HLA haplotypes in comparison with almost 40 000 healthy Swedish controls. As expected, the risk-conveying HLA alleles A*01, B*08 and DRB1*01 were overrepresented among the IgAD twins and were also associated with significantly lower mean serum IgA concentrations in the twin cohort. In contrast, significantly higher mean IgA concentrations were found among individuals carrying the protective HLA alleles B*07 and DRB1*15. Exome sequencing data from two MZ twin pairs discordant for the deficiency showed no differences between the siblings. Model fitting analyses derived a heritability of 35% and indicate that genetic influences are modestly important for IgAD. The probandwise concordance rates for IgAD were found to be 31% for MZ and 13% for DZ twins.
Collapse
|
158
|
Senger K, Hackney J, Payandeh J, Zarrin AA. Antibody Isotype Switching in Vertebrates. Results Probl Cell Differ 2015; 57:295-324. [PMID: 26537387 DOI: 10.1007/978-3-319-20819-0_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humoral or antibody-mediated immune response in vertebrates has evolved to respond to diverse antigenic challenges in various anatomical locations. Diversification of the immunoglobulin heavy chain (IgH) constant region via isotype switching allows for remarkable plasticity in the immune response, including versatile tissue distribution, Fc receptor binding, and complement fixation. This enables antibody molecules to exert various biological functions while maintaining antigen-binding specificity. Different immunoglobulin (Ig) classes include IgM, IgD, IgG, IgE, and IgA, which exist as surface-bound and secreted forms. High-affinity autoantibodies are associated with various autoimmune diseases such as lupus and arthritis, while defects in components of isotype switching are associated with infections. A major route of infection used by a large number of pathogens is invasion of mucosal surfaces within the respiratory, digestive, or urinary tract. Most infections of this nature are initially limited by effector mechanisms such as secretory IgA antibodies. Mucosal surfaces have been proposed as a major site for the genesis of adaptive immune responses, not just in fighting infections but also in tolerating commensals and constant dietary antigens. We will discuss the evolution of isotype switching in various species and provide an overview of the function of various isotypes with a focus on IgA, which is universally important in gut homeostasis as well as pathogen clearance. Finally, we will discuss the utility of antibodies as therapeutic modalities.
Collapse
Affiliation(s)
- Kate Senger
- Department of Immunology, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Jason Hackney
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Jian Payandeh
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Ali A Zarrin
- Department of Immunology, Genentech Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
159
|
Brüssow H. Microbiota and the human nature: know thyself. Environ Microbiol 2014; 17:10-5. [DOI: 10.1111/1462-2920.12693] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 10/24/2014] [Indexed: 12/27/2022]
Affiliation(s)
- Harald Brüssow
- Nutrition and Health Research; Nestlé Research Center; Lausanne 26 Lausanne CH-1000 Switzerland
| |
Collapse
|
160
|
Abstract
The intestinal mucosa harbors the largest population of antibody (Ab)-secreting plasma cells (PC) in the human body, producing daily several grams of immunoglobulin A (IgA). IgA has many functions, serving as a first-line barrier that protects the mucosal epithelium from pathogens, toxins and food antigens (Ag), shaping the intestinal microbiota, and regulating host-commensal homeostasis. Signals induced by commensal colonization are central for regulating IgA induction, maintenance, positioning and function and the number of IgA(+) PC is dramatically reduced in neonates and germ-free (GF) animals. Recent evidence demonstrates that the innate immune effector molecules tumor necrosis factor α (TNFα) and inducible nitric oxide synthase (iNOS) are required for IgA(+) PC homeostasis during the steady state and infection. Moreover, new functions ascribed to PC independent of Ab secretion continue to emerge, suggesting that PC, including IgA(+) PC, should be re-examined in the context of inflammation and infection. Here, we outline mechanisms of IgA(+) PC generation and survival, reviewing their functions in health and disease.
Collapse
Key Words
- AID, activation-induced deaminase
- APC, antigen-presenting cell
- APRIL, a proliferation-inducing ligand
- Ab, antibody
- Ag, antigen
- Arg, arginase
- Atg, autophagy-related gene
- B cell
- BAFF, B-cell activating factor
- BCMA, B-cell maturation antigen
- BM, bone marrow
- Blimp, B-lymphocyte-induced maturation protein
- CCL, CC chemokine ligand
- CCR, CC chemokine receptor
- CD, cluster of differentiation
- CSR, class-switch recombination
- CXCL, CXC chemokine ligand
- DC, dendritic cell
- ER, endoplasmic reticulum
- FDC, follicular dendritic cells
- FcαR, Fc fragment of IgA receptor
- GALT, gut-associated lymphoid tissues
- GC, germinal center
- GF, germ-free
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- GRP, glucose-regulated proteins
- HIV, human immunodeficiency virus
- IEC, intestinal epithelial cells
- IFN, interferon
- IL, interleukin
- ILC, innate lymphoid cells
- ILF, isolated lymphoid follicles
- IRE, inositol-requiring enzyme
- IRF, interferon regulatory factor
- Id, inhibitor of DNA binding
- IgA, immunoglobulin A
- IgAD, selective IgA deficiency
- L-Arg, L-Arginine
- L-Cit, L-citrulline
- L-Glu, L-Glutamate
- L-Orn, L-Ornithine
- L-Pro, L-Proline
- LIGHT, homologous to lymphotoxin, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes
- LP, lamina propria
- LT, lymphotoxinLTβR, LTβ-receptor
- LTi, lymphoid tissue-inducer
- LTo, lymphoid tissue organizing
- Ly, lymphocyte antigen
- MHC, major histocompatibility complex
- MLN, mesenteric lymph nodes
- NO, nitric oxide
- PC, plasma cells
- PP, Peyer's patch
- Pax, paired box
- ROR, Retionic acid receptor (RAR)- or retinoid-related orphan receptor
- SC, stromal cells
- SHM, somatic hypermutation
- SIGNR, specific intercellular adhesion molecule-3-grabbing non-integrin-related
- SIgAsecretory IgA
- TACI, transmembrane activator and calcium-modulator and cyclophilin ligand interactor
- TD, T-dependent
- TFH, T-follicular helper cells
- TGFβR, transforming growth factor β receptor
- TI, T-independent
- TLR, Toll-like receptor
- TNFR, TNF receptor
- TNFα, tumor necrosis factor α
- Th, T helper cell
- Treg, T-regulatory cell
- UPR, unfolded protein response
- XBP, X-box binding protein
- bcl, B-cell lymphoma
- cGMP, cyclic guanosine monophosphate
- iNOS, inducible nitric oxide synthase
- immunoglobulin A (IgA)
- inducible nitric oxide synthase (iNOS)
- innate immune recognition
- intestinal microbiota
- mucosa
- pIgA, polymeric IgA
- pIgR, polymeric Ig receptor
- plasma cell
Collapse
Affiliation(s)
| | - Olga L Rojas
- Department of Immunology; University of Toronto; Toronto, ON Canada
| | - Jörg H Fritz
- Department of Microbiology and Immunology; Department of Physiology; Complex Traits Group; McGill University; Montreal, QC Canada,Correspondence to: Jörg H Fritz;
| |
Collapse
|
161
|
Stieh DJ, King DF, Klein K, Liu P, Shen X, Hwang KK, Ferrari G, Montefiori DC, Haynes B, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Robb ML, Kim JH, Denny TN, Tomaras GD, Shattock RJ. Aggregate complexes of HIV-1 induced by multimeric antibodies. Retrovirology 2014; 11:78. [PMID: 25274446 PMCID: PMC4193994 DOI: 10.1186/s12977-014-0078-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/30/2014] [Indexed: 11/10/2022] Open
Abstract
Background Antibody mediated viral aggregation may impede viral transfer across mucosal surfaces by hindering viral movement in mucus, preventing transcytosis, or reducing inter-cellular penetration of epithelia thereby limiting access to susceptible mucosal CD4 T cells and dendritic cells. These functions may work together to provide effective immune exclusion of virus from mucosal tissue; however little is known about the antibody characteristics required to induce HIV aggregation. Such knowledge may be critical to the design of successful immunization strategies to facilitate viral immune exclusion at the mucosal portals of entry. Results The potential of neutralizing and non-neutralizing IgG and IgA monoclonals (mAbs) to induce HIV-1 aggregation was assessed by Dynamic light scattering (DLS). Although neutralizing and non-neutralizing IgG mAbs and polyclonal HIV-Ig efficiently aggregated soluble Env trimers, they were not capable of forming viral aggregates. In contrast, dimeric (but not monomeric) IgA mAbs induced stable viral aggregate populations that could be separated from uncomplexed virions. Epitope specificity influenced both the degree of aggregation and formation of higher order complexes by dIgA. IgA purified from serum of uninfected RV144 vaccine trial responders were able to efficiently opsonize viral particles in the absence of significant aggregation, reflective of monomeric IgA. Conclusions These results collectively demonstrate that dIgA is capable of forming stable viral aggregates providing a plausible basis for testing the effectiveness of aggregation as a potential protection mechanism at the mucosal portals of viral entry. Electronic supplementary material The online version of this article (doi:10.1186/s12977-014-0078-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel J Stieh
- Center for Infection, Department of Cellular and Molecular Medicine, St George's, University of London, London, SW17 0RE, UK. .,Current address: Department of Cellular and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Deborah F King
- Mucosal Infection & Immunity Group, Section of Infectious Diseases, Imperial College London, St Mary's Campus, London, W2 1PG, UK.
| | - Katja Klein
- Mucosal Infection & Immunity Group, Section of Infectious Diseases, Imperial College London, St Mary's Campus, London, W2 1PG, UK.
| | - Pinghuang Liu
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. .,Current address: Division of Swine Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China.
| | - Xiaoying Shen
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Kwan Ki Hwang
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Guido Ferrari
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA.
| | - David C Montefiori
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Barton Haynes
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA.
| | | | | | | | | | - Nelson L Michael
- Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America.
| | - Merlin L Robb
- Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America.
| | - Jerome H Kim
- Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America.
| | - Thomas N Denny
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Georgia D Tomaras
- Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Robin J Shattock
- Mucosal Infection & Immunity Group, Section of Infectious Diseases, Imperial College London, St Mary's Campus, London, W2 1PG, UK.
| |
Collapse
|
162
|
Reinhart D, Kunert R. Upstream and downstream processing of recombinant IgA. Biotechnol Lett 2014; 37:241-51. [DOI: 10.1007/s10529-014-1686-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/12/2014] [Indexed: 11/24/2022]
|
163
|
Kawamoto S, Maruya M, Kato L, Suda W, Atarashi K, Doi Y, Tsutsui Y, Qin H, Honda K, Okada T, Hattori M, Fagarasan S. Foxp3+ T Cells Regulate Immunoglobulin A Selection and Facilitate Diversification of Bacterial Species Responsible for Immune Homeostasis. Immunity 2014; 41:152-65. [DOI: 10.1016/j.immuni.2014.05.016] [Citation(s) in RCA: 283] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/09/2014] [Indexed: 02/06/2023]
|
164
|
Stowell SR, Arthur CM, McBride R, Berger O, Razi N, Heimburg-Molinaro J, Rodrigues LC, Gourdine JP, Noll AJ, von Gunten S, Smith DF, Knirel YA, Paulson JC, Cummings RD. Microbial glycan microarrays define key features of host-microbial interactions. Nat Chem Biol 2014; 10:470-6. [PMID: 24814672 DOI: 10.1038/nchembio.1525] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 04/09/2014] [Indexed: 11/09/2022]
Abstract
Genomic approaches continue to provide unprecedented insight into the microbiome, yet host immune interactions with diverse microbiota can be difficult to study. We therefore generated a microbial microarray containing defined antigens isolated from a broad range of microbial flora to examine adaptive and innate immunity. Serological studies with this microarray show that immunoglobulins from multiple mammalian species have unique patterns of reactivity, whereas exposure of animals to distinct microbes induces specific serological recognition. Although adaptive immunity exhibited plasticity toward microbial antigens, immunological tolerance limits reactivity toward self. We discovered that several innate immune galectins show specific recognition of microbes that express self-like antigens, leading to direct killing of a broad range of Gram-negative and Gram-positive microbes. Thus, host protection against microbes seems to represent a balance between adaptive and innate immunity to defend against evolving antigenic determinants while protecting against molecular mimicry.
Collapse
Affiliation(s)
- Sean R Stowell
- 1] Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA. [2]
| | - Connie M Arthur
- 1] Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA. [2]
| | - Ryan McBride
- 1] Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA. [2] Department of and Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA. [3]
| | - Oren Berger
- 1] Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA. [2] Department of and Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - Nahid Razi
- 1] Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA. [2] Department of and Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - Jamie Heimburg-Molinaro
- Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lilian C Rodrigues
- Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jean-Philippe Gourdine
- Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alexander J Noll
- Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - David F Smith
- Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yuriy A Knirel
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - James C Paulson
- 1] Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA. [2] Department of and Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - Richard D Cummings
- Department of Biochemistry and the Glycomics Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
165
|
Guzman-Bautista ER, Garcia-Ruiz CE, Gama-Espinosa AL, Ramirez-Estudillo C, Rojas-Gomez OI, Vega-Lopez MA. Effect of age and maternal antibodies on the systemic and mucosal immune response after neonatal immunization in a porcine model. Immunology 2014; 141:609-16. [PMID: 24754050 DOI: 10.1111/imm.12222] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Newborn mammals are highly susceptible to respiratory infections. Although maternal antibodies (MatAb) offer them some protection, they may also interfere with their systemic immune response to vaccination. However, the impact of MatAb on the neonatal mucosal immune response remains incompletely described. This study was performed to determine the effect of ovalbumin (OVA) -specific MatAb on the anti- OVA antibody response in sera, nasal secretions and saliva from specific pathogen-free Vietnamese miniature piglets immunized at 7 or 14 days of age. Our results demonstrated that MatAb increased antigen-specific IgA and IgG responses in sera, and transiently enhanced an early secretory IgA response in nasal secretions of piglets immunized at 7 days of age. In contrast, we detected a lower mucosal (nasal secretion and saliva) anti- OVA IgG response in piglets with MatAb immunized at 14 days of age, compared with piglets with no MatAb, suggesting a modulatory effect of antigen-specific maternal factors on the isotype transfer to the mucosal immune exclusion system. In our porcine model, we demonstrated that passive maternal immunity positively modulated the systemic and nasal immune responses of animals immunized early in life. Our results, therefore, open the possibility of inducing systemic and respiratory mucosal immunity in the presence of MatAb through early vaccination.
Collapse
|
166
|
Kamada N, Núñez G. Regulation of the immune system by the resident intestinal bacteria. Gastroenterology 2014; 146:1477-88. [PMID: 24503128 PMCID: PMC3995843 DOI: 10.1053/j.gastro.2014.01.060] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/27/2014] [Accepted: 01/30/2014] [Indexed: 02/07/2023]
Abstract
The microbiota is an important factor in the development of the immune response. The interaction between the gastrointestinal tract and resident microbiota is well balanced in healthy individuals, but its breakdown can lead to intestinal and extraintestinal disease. We review current knowledge about the mechanisms that regulate the interaction between the immune system and the microbiota, focusing on the role of resident intestinal bacteria in the development of immune responses. We also discuss mechanisms that prevent immune responses against resident bacteria, and how the indigenous bacteria stimulate the immune system to protect against commensal pathobionts and exogenous pathogens. Unraveling the complex interactions between resident intestinal bacteria and the immune system could improve our understanding of disease pathogenesis and lead to new therapeutic approaches.
Collapse
Affiliation(s)
- Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
167
|
Masahata K, Umemoto E, Kayama H, Kotani M, Nakamura S, Kurakawa T, Kikuta J, Gotoh K, Motooka D, Sato S, Higuchi T, Baba Y, Kurosaki T, Kinoshita M, Shimada Y, Kimura T, Okumura R, Takeda A, Tajima M, Yoshie O, Fukuzawa M, Kiyono H, Fagarasan S, Iida T, Ishii M, Takeda K. Generation of colonic IgA-secreting cells in the caecal patch. Nat Commun 2014; 5:3704. [PMID: 24718324 DOI: 10.1038/ncomms4704] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 03/21/2014] [Indexed: 01/22/2023] Open
Abstract
Gut-associated lymphoid tissues are responsible for the generation of IgA-secreting cells. However, the function of the caecal patch, a lymphoid tissue in the appendix, remains unknown. Here we analyse the role of the caecal patch using germ-free mice colonized with intestinal bacteria after appendectomy. Appendectomized mice show delayed accumulation of IgA(+) cells in the large intestine, but not the small intestine, after colonization. Decreased colonic IgA(+) cells correlate with altered faecal microbiota composition. Experiments using photoconvertible Kaede-expressing mice or adoptive transfer show that the caecal patch IgA(+) cells migrate to the large and small intestines, whereas Peyer's patch cells are preferentially recruited to the small intestine. IgA(+) cells in the caecal patch express higher levels of CCR10. Dendritic cells in the caecal patch, but not Peyer's patches, induce CCR10 on cocultured B cells. Thus, the caecal patch is a major site for generation of IgA-secreting cells that migrate to the large intestine.
Collapse
Affiliation(s)
- Kazunori Masahata
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Department of Pediatric Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Eiji Umemoto
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Hisako Kayama
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Manato Kotani
- Department of Immunology and Cell Biology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takashi Kurakawa
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kazuyoshi Gotoh
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shintaro Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tomonori Higuchi
- Department of Microbiology, Kinki University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yoshihiro Baba
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Makoto Kinoshita
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Yosuke Shimada
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Taishi Kimura
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryu Okumura
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Akira Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Tajima
- The Institute of Experimental Animal Sciences, Faculty of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Osamu Yoshie
- Department of Microbiology, Kinki University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masahiro Fukuzawa
- Department of Pediatric Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroshi Kiyono
- 1] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan [2] Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Tetsuya Iida
- 1] Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan [2] Laboratory of Genomic Research on Pathogenic Bacteria, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Ishii
- 1] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan [2] Department of Immunology and Cell Biology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kiyoshi Takeda
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| |
Collapse
|
168
|
Abstract
Vibrio cholerae is the causative agent of the acute diarrheal disease of cholera. Innate immune responses to V. cholerae are not a major cause of cholera pathology, which is characterized by severe, watery diarrhea induced by the action of cholera toxin. Innate responses may, however, contribute to resolution of infection and must be required to initiate adaptive responses after natural infection and oral vaccination. Here we investigated whether a well-established infant mouse model of cholera can be used to observe an innate immune response. We also used a vaccination model in which immunized dams protect their pups from infection through breast milk antibodies to investigate innate immune responses after V. cholerae infection for pups suckled by an immune dam. At the peak of infection, we observed neutrophil recruitment accompanied by induction of KC, macrophage inflammatory protein 2 (MIP-2), NOS-2, interleukin-6 (IL-6), and IL-17a. Pups suckled by an immunized dam did not mount this response. Accessory toxins RtxA and HlyA played no discernible role in neutrophil recruitment in a wild-type background. The innate response to V. cholerae deleted for cholera toxin-encoding phage (CTX) and part of rtxA was significantly reduced, suggesting a role for CTX-carried genes or for RtxA in the absence of cholera toxin (CTX). Two extracellular V. cholerae DNases were not required for neutrophil recruitment, but DNase-deficient V. cholerae caused more clouds of DNA in the intestinal lumen, which appeared to be neutrophil extracellular traps (NETs), suggesting that V. cholerae DNases combat NETs. Thus, the infant mouse model has hitherto unrecognized utility for interrogating innate responses to V. cholerae infection.
Collapse
|
169
|
Zhu LY, Lin AF, Shao T, Nie L, Dong WR, Xiang LX, Shao JZ. B cells in teleost fish act as pivotal initiating APCs in priming adaptive immunity: an evolutionary perspective on the origin of the B-1 cell subset and B7 molecules. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:2699-714. [PMID: 24532580 DOI: 10.4049/jimmunol.1301312] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The long-held paradigm that B cells cannot uptake nonspecific particulate Ags for the initiation of primary adaptive immunity has been challenged by the recent discovery that teleost B cells have potent phagocytic and microbicidal abilities. This discovery provides preliminary clues that primitive B cells might act as initiating APCs in priming adaptive immunity. In this study, zebrafish B cells clearly showed a potent Ag-presenting ability to both soluble Ags and bacterial particles to prime naive CD4(+) T cell activation. This finding demonstrates the innate-like nature of teleost B cells in the interface of innate and adaptive immunity, indicating that they might consist of a major population of initiating APCs whose performance is similar to that of dendritic cells. Given the functional similarities between teleost B cells and the mammalian B-1 subset, we hypothesize that B-1 lineage and teleost B cells might originate from a common ancestor with potent phagocytic and initiating APC capacities. In addition, CD80/86 and CD83 costimulatory signals were identified as being essential for B cell-initiated adaptive immunity. This result suggests that the costimulatory mechanism originated as early as the origin of adaptive immunity and is conserved throughout vertebrate evolution. In fish, only a single CD80/86 copy exists, which is similar to mammalian CD86 rather than to CD80. Thus, CD86 might be a more primordial B7 family member that originated from fish. This study provides valuable insights into the evolutionary history of professional APCs, B cell lineages, and the costimulatory mechanism underlying adaptive immunity as a whole.
Collapse
Affiliation(s)
- Lv-yun Zhu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
170
|
Antigen targeting to M cells for enhancing the efficacy of mucosal vaccines. Exp Mol Med 2014; 46:e85. [PMID: 24626171 PMCID: PMC3972786 DOI: 10.1038/emm.2013.165] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/06/2013] [Indexed: 01/01/2023] Open
Abstract
Vaccination is one of the most successful applications of immunology and for a long time has depended on parenteral administration protocols. However, recent studies have pointed to the promise of mucosal vaccination because of its ease, economy and efficiency in inducing an immune response not only systemically, but also in the mucosal compartment where many pathogenic infections are initiated. However, successful mucosal vaccination requires the help of an adjuvant for the efficient delivery of vaccine material into the mucosa and the breaking of the tolerogenic environment, especially in oral mucosal immunization. Given that M cells are the main gateway to take up luminal antigens and initiate antigen-specific immune responses, understanding the role and characteristics of M cells is crucial for the development of successful mucosal vaccines. Especially, particular interest has been focused on the regulation of the tolerogenic mucosal microenvironment and the introduction of the luminal antigen into the lymphoid organ by exploiting the molecules of M cells. Here, we review the characteristics of M cells and the immune regulatory factors in mucosa that can be exploited for mucosal vaccine delivery and mucosal immune regulation.
Collapse
|
171
|
Chen Y, Lee T, Hong W, Hsieh H, Chen M. Effects of Lactobacillus kefiranofaciens M1 isolated from kefir grains on enterohemorrhagic Escherichia coli infection using mouse and intestinal cell models. J Dairy Sci 2013; 96:7467-77. [DOI: 10.3168/jds.2013-7015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 08/31/2013] [Indexed: 12/12/2022]
|
172
|
Age-related expression of the polymeric immunoglobulin receptor (pIgR) in the gastric mucosa of young pigs. PLoS One 2013; 8:e81473. [PMID: 24236214 PMCID: PMC3827463 DOI: 10.1371/journal.pone.0081473] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 10/23/2013] [Indexed: 11/19/2022] Open
Abstract
To date few studies have addressed the development and function of the porcine gastric mucosal immune system and this is a major limitation to understanding the immunopathogenesis of infections occurring in young pigs. The polymeric immunoglobulin receptor (pIgR) mediates the transport of secretory immunoglobulins until luminal surface of the gut mucosa and the aim of this study was to investigate the time course of pIgR expression and to determine its localization in three functionally different porcine gastric sites during the suckling period and after weaning. An additional goal was to investigate the time course expression of toll-like receptors (TLRs) in relation to pIgR expression. Gastric samples were collected from the cardiac-to-oxyntic transition (Cd), the oxyntic (Ox), and the pyloric (Py) regions in 84 pigs, slaughtered before weaning (14, 21 and 28 days of age; 23, 23 and 19 pigs, respectively) and 14 days post-weaning (42 days of age, 23 pigs). PIgR was expressed in the mucosa of all the three gastric sites, and its transcript levels were modulated during suckling and after weaning, with regional differences. PIgR expression increased linearly during suckling (P=0.019) and also increased post-weaning (P=0.001) in Cd, it increased post-weaning in Py (P=0.049) and increased linearly during suckling in Ox (P=0.036). TLRs expression was also modulated during development: in Cd, TLR2 increased linearly during suckling (P=0.003); in Ox, TLR2 decreased after weaning (P=0.038) while TLR4 increased linearly during suckling(P=0.008). The expression of TLR2, 3 and 4 in Ox was positively correlated with pIgR expression (P<0.001). Importantly, both pIgR protein and mRNA were localized, by immunohistochemistry and in situ hybridization, respectively, in the gastric glands of the lamina propria. These results indicate that pIgR is actively synthesized in the gastric mucosa and suggest that pIgR could play a crucial role in gastric mucosal immune defense of growing pigs.
Collapse
|
173
|
Kato LM, Kawamoto S, Maruya M, Fagarasan S. Gut TFH and IgA: key players for regulation of bacterial communities and immune homeostasis. Immunol Cell Biol 2013; 92:49-56. [PMID: 24100385 DOI: 10.1038/icb.2013.54] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 02/07/2023]
Abstract
The main function of the immune system is to protect the host against pathogens. However, unlike the systemic immune system, the gut immune system does not eliminate, but instead nourishes complex bacterial communities and establishes advanced symbiotic relationships. Immunoglobulin A (IgA) is the most abundant antibody isotype in mammals, produced mainly in the gut. The primary function of IgA is to maintain homeostasis at mucosal surfaces, and studies in mice have demonstrated that IgA diversification has an essential role in the regulation of gut microbiota. Dynamic diversification and constant adaptation of IgA responses to local microbiota require expression of activation-induced cytidine deaminase by B cells and control from T follicular helper and Foxp3(+) T cells in germinal centers (GCs). We discuss the finely tuned regulatory mechanisms for IgA synthesis in GCs of Peyer's patches and emphasize the roles of CD4(+) T cells for IgA selection and the maintenance of appropriate gut microbial communities required for immune homeostasis.
Collapse
Affiliation(s)
- Lucia M Kato
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences IMS-RCAI, RIKEN Yokohama Institute, Yokohama, Japan
| | - Shimpei Kawamoto
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences IMS-RCAI, RIKEN Yokohama Institute, Yokohama, Japan
| | - Mikako Maruya
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences IMS-RCAI, RIKEN Yokohama Institute, Yokohama, Japan
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences IMS-RCAI, RIKEN Yokohama Institute, Yokohama, Japan
| |
Collapse
|
174
|
da Silva BF, Souza GHMF, Turco EGL, Del Giudice PT, Soler TB, Spaine DM, Borrelli M, Gozzo FC, Pilau EJ, Garcia JS, Ferreira CR, Eberlin MN, Bertolla RP. Differential seminal plasma proteome according to semen retrieval in men with spinal cord injury. Fertil Steril 2013; 100:959-69. [DOI: 10.1016/j.fertnstert.2013.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 11/30/2022]
|
175
|
Kim SH, Lee HY, Jang YS. Targeted Delivery of VP1 Antigen of Foot-and-mouth Disease Virus to M Cells Enhances the Antigen-specific Systemic and Mucosal Immune Response. Immune Netw 2013; 13:157-62. [PMID: 24009543 PMCID: PMC3759713 DOI: 10.4110/in.2013.13.4.157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 08/01/2013] [Accepted: 08/05/2013] [Indexed: 12/01/2022] Open
Abstract
Application of vaccine materials through oral mucosal route confers great economical advantage in animal farming industry due to much less vaccination cost compared with that of injection-based vaccination. In particular, oral administration of recombinant protein antigen against foot-and-mouth disease virus (FMDV) is an ideal strategy because it is safe from FMDV transmission during vaccine production and can induce antigen-specific immune response in mucosal compartments, where FMDV infection has been initiated, which is hardly achievable through parenteral immunization. Given that effective delivery of vaccine materials into immune inductive sites is prerequisite for effective oral mucosal vaccination, M cell-targeting strategy is crucial in successful vaccination since M cells are main gateway for luminal antigen influx into mucosal lymphoid tissue. Here, we applied previously identified M cell-targeting ligand Co1 to VP1 of FMDV in order to test the possible oral mucosal vaccination against FMDV infection. M cell-targeting ligand Co1-conjugated VP1 interacted efficiently with M cells of Peyer's patch. In addition, oral administration of ligand-conjugated VP1 enhanced the induction of VP1-specific IgG and IgA responses in systemic and mucosal compartments, respectively, in comparison with those from oral administration of VP1 alone. In addition, the enhanced VP1-specific immune response was found to be due to antigen-specific Th2-type cytokine production. Collectively, it is suggested that the M cell-targeting strategy could be applied to develop efficient oral mucosal vaccine against FMDV infection.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology, Interdisciplinary Program of Bioactive Materials, and Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Korea
| | | | | |
Collapse
|
176
|
Kim SH, Yang IY, Jang SH, Kim J, Truong TT, Van Pham T, Truong NU, Lee KY, Jang YS. C5a receptor-targeting ligand-mediated delivery of dengue virus antigen to M cells evokes antigen-specific systemic and mucosal immune responses in oral immunization. Microbes Infect 2013; 15:895-902. [PMID: 23892099 DOI: 10.1016/j.micinf.2013.07.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 04/03/2013] [Accepted: 07/16/2013] [Indexed: 12/16/2022]
Abstract
Oral mucosal immunization is a feasible and economic vaccination strategy. In order to achieve a successful oral mucosal vaccination, antigen delivery to gut immune inductive site and avoidance of oral tolerance induction should be secured. One promising approach is exploring the specific molecules expressed on the apical surfaces of M cells that have potential for antigen uptake and immune stimulation. We previously identified complement 5a receptor (C5aR) expression on human M-like cells and mouse M cells and confirmed its non-redundant role as a target receptor for antigen delivery to M cells using a model antigen. Here, we applied the OmpH ligand, which is capable of targeting the ligand-conjugated antigen to M cells to induce specific mucosal and systemic immunities against the EDIII of dengue virus (DENV). Oral immunization with the EDIII-OmpH efficiently targeted the EDIII to M cells and induced EDIII-specific immune responses comparable to those induced by co-administration of EDIII with cholera toxin (CT). Also, the enhanced responses by OmpH were characterized as Th2-skewed responses. Moreover, oral immunization using EDIII-OmpH did not induce systemic tolerance against EDIII. Collectively, we suggest that OmpH-mediated targeting of antigens to M cells could be used for an efficient oral vaccination against DENV infection.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology, Chonbuk National University, Jeonju 561-756, Republic of Korea; Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol 2013; 14:685-90. [PMID: 23778796 PMCID: PMC4083503 DOI: 10.1038/ni.2608] [Citation(s) in RCA: 1066] [Impact Index Per Article: 88.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/09/2013] [Indexed: 02/07/2023]
Abstract
A dense resident microbial community in the gut, referred as the commensal microbiota, coevolved with the host and is essential for many host physiological processes that include enhancement of the intestinal epithelial barrier, development of the immune system and acquisition of nutrients. A major function of the microbiota is protection against colonization by pathogens and overgrowth of indigenous pathobionts that can result from the disruption of the healthy microbial community. The mechanisms that regulate the ability of the microbiota to restrain pathogen growth are complex and include competitive metabolic interactions, localization to intestinal niches and induction of host immune responses. Pathogens, in turn, have evolved strategies to escape from commensal-mediated resistance to colonization. Thus, the interplay between commensals and pathogens or indigenous pathobionts is critical for controlling infection and disease. Understanding pathogen-commensal interactions may lead to new therapeutic approaches to treating infectious diseases.
Collapse
Affiliation(s)
- Nobuhiko Kamada
- Department of Pathology and Comprehensive Cancer Center, University of Michigan, Ann Arbor 48109, USA
| | - Grace Y. Chen
- Department of Internal Medicine and Comprehensive Cancer Center, University of Michigan, Ann Arbor 48109, USA
| | - Naohiro Inohara
- Department of Pathology, University of Michigan, Ann Arbor 48109, USA
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan, Ann Arbor 48109, USA
| |
Collapse
|
178
|
Moldt B, Saye-Francisco K, Schultz N, Burton DR, Hessell AJ. Simplifying the synthesis of SIgA: combination of dIgA and rhSC using affinity chromatography. Methods 2013; 65:127-32. [PMID: 23811333 DOI: 10.1016/j.ymeth.2013.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/17/2013] [Accepted: 06/18/2013] [Indexed: 01/30/2023] Open
Abstract
The mucosal epithelia together with adaptive immune responses, such as local production and secretion of dimeric and polymeric immunoglobulin A (IgA), are a crucial part of the first line of defense against invading pathogens. IgA is primarily secreted as SIgA and plays multiple roles in mucosal defense. The study of SIgA-mediated protection is an important area of research in mucosal immunity but an easy, fast and reproducible method to generate pathogen-specific SIgA in vitro has not been available. We report here a new method to produce SIgA by co-purification of dimeric IgA, containing J chain, and recombinant human SC expressed in CHO cells. We previously reported the generation, production and characterization of the human recombinant monoclonal antibody IgA2 b12. This antibody, derived from the variable regions of the neutralizing anti-HIV-1 mAb IgG1 b12, blocked viral attachment and uptake by epithelial cells in vitro. We used a cloned CHO cell line that expresses monomeric, dimeric and polymeric species of IgA2 b12 for large-scale production of dIgA2 b12. Subsequently, we generated a CHO cell line to express recombinant human secretory component (rhSC). Here, we combined dIgA2 b12 and CHO-expressed rhSC via column chromatography to produce SIgA2 b12 that remains fully intact upon elution with 0.1M citric acid, pH 3.0. We have performed biochemical analysis of the synthesized SIgA to confirm the species is of the expected size and retains the functional properties previously described for IgA2 b12. We show that SIgA2 b12 binds to the HIV-1 gp120 glycoprotein with similar apparent affinity to that of monomeric and dimeric forms of IgA2 b12 and neutralizes HIV-1 isolates with similar potency. An average yield of 6 mg of SIgA2 b12 was achieved from the combination of 20mg of purified dIgA2 b12 and 2L of rhSC-containing CHO cell supernatant. We conclude that synthesized production of stable SIgA can be generated by co-purification. This process introduces a simplified means of generating a variety of pathogen-specific SIgA antibodies for research and clinical applications.
Collapse
Affiliation(s)
- Brian Moldt
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Karen Saye-Francisco
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Niccole Schultz
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R Burton
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Boston, MA, USA
| | - Ann J Hessell
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
179
|
Deal EM, Lahl K, Narváez CF, Butcher EC, Greenberg HB. Plasmacytoid dendritic cells promote rotavirus-induced human and murine B cell responses. J Clin Invest 2013; 123:2464-74. [PMID: 23635775 DOI: 10.1172/jci60945] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 02/26/2013] [Indexed: 12/22/2022] Open
Abstract
B cell-dependent immunity to rotavirus, an important intestinal pathogen, plays a significant role in viral clearance and protects against reinfection. Human in vitro and murine in vivo models of rotavirus infection were used to delineate the role of primary plasmacytoid DCs (pDCs) in initiating B cell responses. Human pDCs were necessary and sufficient for B cell activation induced by rotavirus. Type I IFN recognition by B cells was essential for rotavirus-mediated B cell activation in vitro and murine pDCs and IFN-α/β-mediated B cell activation after in vivo intestinal rotavirus infection. Furthermore, rotavirus-specific serum and mucosal antibody responses were defective in mice lacking functional pDCs at the time of infection. These data demonstrate that optimal B cell activation and virus-specific antibody secretion following mucosal infection were a direct result of pDC-derived type I IFN. Importantly, viral shedding significantly increased in pDC-deficient mice, suggesting that pDC-dependent antibody production influences viral clearance. Thus, mucosal pDCs critically influence the course of rotavirus infection through rotavirus recognition and subsequent IFN production and display powerful adjuvant properties to initiate and enhance humoral immunity.
Collapse
Affiliation(s)
- Emily M Deal
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305-5105, USA
| | | | | | | | | |
Collapse
|
180
|
|
181
|
Maruya M, Kawamoto S, Kato LM, Fagarasan S. Impaired selection of IgA and intestinal dysbiosis associated with PD-1-deficiency. Gut Microbes 2013; 4:165-71. [PMID: 23333864 PMCID: PMC3595078 DOI: 10.4161/gmic.23595] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A major function of immunoglobulin A (IgA) is to maintain balanced bacterial communities in the gut. We have previously shown that diversification of IgA upon somatic hypermutation (SHM) is critical for IgA function yet the principles governing the selection of IgA in the gut have remained elusive. Here we discuss recent progress in understanding this process as revealed by our studies in mice that lack the inhibitory co-receptor programmed cell death-1 (PD-1). We found that PD-1 affects the dynamics of germinal center (GC) B cells by controlling the number and the nature of T helper cells in the Peyer's patches (PPs). Deregulation of the T cell compartment impacts the selection of IgA plasma cells leading to gut dysbiosis. When the PD-1-dependent checkpoint is missing, gut bacteria go beyond the mucosal barrier and induce systemic GCs that can generate antibodies with auto-reactive properties.
Collapse
|
182
|
Schuijt TJ, van der Poll T, de Vos WM, Wiersinga WJ. The intestinal microbiota and host immune interactions in the critically ill. Trends Microbiol 2013; 21:221-9. [PMID: 23454077 DOI: 10.1016/j.tim.2013.02.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/28/2013] [Accepted: 02/01/2013] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract harbors a complex population of microbes that play a fundamental role in the development of the immune system and human health. Besides an important local contribution in the host defense against infections, it has become increasingly clear that intestinal bacteria also modulate immune responses at systemic sites. These new insights can be of profound clinical relevance especially for intensive care medicine where the majority of patients are treated with antibiotics, which have pervasive and long-term effects on the intestinal microbiota. Moreover, considerable progress has been made in defining the role of the intestinal microbiota in both health and disease. In this review, we highlight these aspects and focus on recent key findings addressing the role of intestinal microbiota in antimicrobial defense mechanisms and its impact on intestinal homeostasis in the critically ill.
Collapse
Affiliation(s)
- Tim J Schuijt
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands.
| | | | | | | |
Collapse
|
183
|
Kim JJ, Khan WI. Goblet cells and mucins: role in innate defense in enteric infections. Pathogens 2013; 2:55-70. [PMID: 25436881 PMCID: PMC4235714 DOI: 10.3390/pathogens2010055] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 01/27/2013] [Accepted: 01/28/2013] [Indexed: 12/16/2022] Open
Abstract
Goblet cells reside throughout the gastrointestinal (GI) tract and are responsible for the production and preservation of a protective mucus blanket by synthesizing and secreting high molecular weight glycoproteins known as mucins. The concept of the mucus layer functioning as a dynamic protective barrier is suggested by studies showing changes in mucins in inflammatory conditions of the GI tract, by the altered goblet cell response in germ-free animals, and by the enhanced mucus secretion seen in response to infections. The mucin-containing mucus layer coating the GI epithelium is the front line of innate host defense. Mucins are likely to be the first molecules that invading pathogens interact with at the cell surface and thus, can limit binding to other glycoproteins and neutralize the pathogen. This review will focus on what is known about goblet cell response in various GI infections and the regulatory networks that mediate goblet cell function and mucin production in response to intestinal insults. In addition, we describe the current knowledge on the role of mucins in intestinal innate defense. It is the aim of this review to provide the readers with an update on goblet cell biology and current understanding on the role of mucins in host defense in enteric infections.
Collapse
Affiliation(s)
- Janice J Kim
- Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, Ontario, L8S 4K1, Canada.
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, Ontario, L8S 4K1, Canada.
| |
Collapse
|
184
|
Glycobiome: bacteria and mucus at the epithelial interface. Best Pract Res Clin Gastroenterol 2013; 27:25-38. [PMID: 23768550 DOI: 10.1016/j.bpg.2013.03.001] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/08/2013] [Indexed: 02/06/2023]
Abstract
The human gastrointestinal tract is colonised with a dense and diverse microbial community, that is an important player in human health and physiology. Close to the epithelial cells the mucosal microbiota is separated from the host with a thin lining of host derived glycans, including the cell surface glycocalyx and the extracellular secreted mucus. The mucosa-associated microbial composition differs from the luminal content and could be particularly important for nutrient exchange, communication with the host, development of the immune system, and resistance against invading pathogens. The mucosa-associated microbiota has adapted to the glycan rich environment by the production of mucus-degrading enzymes and mucus-binding extracellular proteins, and include mucus-degrading specialists such as Akkermansia muciniphila and Bacteroides thetaiotaomicron. This review is focussed on the host-microbe interactions within the glycan landscape at the epithelial interface and considers the spatial organisation and composition of the mucosa-associated microbiota in health and disease.
Collapse
|
185
|
Caoili SEC. Antidotes, antibody-mediated immunity and the future of pharmaceutical product development. Hum Vaccin Immunother 2013; 9:294-9. [PMID: 23291934 PMCID: PMC3859750 DOI: 10.4161/hv.22858] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
If new scientific knowledge is to be more efficiently generated and applied toward the advancement of health, human safety must be more effectively addressed in the conduct of research. Given the present difficulties of accurately predicting biological outcomes of novel interventions in vivo, the imperative of human safety suggests the development of novel pharmaceutical products in tandem with their prospective antidotes in anticipation of possible adverse events, to render the risks of initial clinical trials more acceptable from a regulatory standpoint. Antibody-mediated immunity provides a generally applicable mechanistic basis for developing antidotes to both biologicals and small-molecule drugs (such that antibodies may serve as antidotes to pharmaceutical agents as a class including other antibodies) and also for the control and prevention of both infectious and noninfectious diseases via passive or active immunization. Accordingly, the development of prophylactic or therapeutic passive-immunization strategies using antipeptide antibodies is a plausible prelude to the development of corresponding active-immunization strategies using peptide-based vaccines. In line with this scheme, global proliferation of antibody- and vaccine-production technologies, especially those that obviate dependence on the cold chain for storage and transport of finished products, could provide geographically distributed breakout capability against emerging and future health challenges.
Collapse
Affiliation(s)
- Salvador Eugenio C Caoili
- Department of Biochemistry and Molecular Biology; College of Medicine; University of the Philippines Manila; Manila, Philippines
| |
Collapse
|
186
|
|
187
|
Abstract
An important activity of mucosal surfaces is the production of antibodies (Abs) referred to as secretory immunoglobulin A (SIgA) that serve as a first line of defense to repel pathogenic microorganisms and provide a finely tuned balance to guarantee controlled survival of essential commensal bacteria. By excluding bacteria from the epithelial cell, SIgA participates in the cross-talk between the host and its intestinal content, ensuring appropriate homeostasis under normal conditions. Besides the classical view of immune exclusion function, SIgA Abs exhibit the striking feature to adhere to gastrointestinal M cells residing in the follicle-associated epithelium in organized structures called Peyer's patches. Selective binding of SIgA results in transport across the microfold (M) cells, a process that facilitates the association of the Ab with dendritic cells (DCs) located in the underlying subepithelial dome region of Peyer's patches. Limited entry of free SIgA and SIgA-coated bacteria via this pathway is crucial to the modulation of local immune responses in an environment that limits the onset of pro-inflammatory circuits. Such a mechanism would ensure homeostasis by allowing antigen recognition under neutralized conditions and by avoiding tissue dissemination, two features that endow SIgA with non-inflammatory properties in the mucosal environment.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory of the Division of Immunology and Allergy, University State Hospital (CHUV), Rue du Bugnon, 1011 Lausanne, Switzerland.
| |
Collapse
|
188
|
Kim SH, Lee KY, Jang YS. Mucosal Immune System and M Cell-targeting Strategies for Oral Mucosal Vaccination. Immune Netw 2012; 12:165-75. [PMID: 23213309 PMCID: PMC3509160 DOI: 10.4110/in.2012.12.5.165] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 10/05/2012] [Accepted: 10/09/2012] [Indexed: 02/08/2023] Open
Abstract
Vaccination is one of the most effective methods available to prevent infectious diseases. Mucosa, which are exposed to heavy loads of commensal and pathogenic microorganisms, are one of the first areas where infections are established, and therefore have frontline status in immunity, making mucosa ideal sites for vaccine application. Moreover, vaccination through the mucosal immune system could induce effective systemic immune responses together with mucosal immunity in contrast to parenteral vaccination, which is a poor inducer of effective immunity at mucosal surfaces. Among mucosal vaccines, oral mucosal vaccines have the advantages of ease and low cost of vaccine administration. The oral mucosal immune system, however, is generally recognized as poorly immunogenic due to the frequent induction of tolerance against orally-introduced antigens. Consequently, a prerequisite for successful mucosal vaccination is that the orally introduced antigen should be transported across the mucosal surface into the mucosa-associated lymphoid tissue (MALT). In particular, M cells are responsible for antigen uptake into MALT, and the rapid and effective transcytotic activity of M cells makes them an attractive target for mucosal vaccine delivery, although simple transport of the antigen into M cells does not guarantee the induction of specific immune responses. Consequently, development of mucosal vaccine adjuvants based on an understanding of the biology of M cells has attracted much research interest. Here, we review the characteristics of the oral mucosal immune system and delineate strategies to design effective oral mucosal vaccines with an emphasis on mucosal vaccine adjuvants.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Korea
| | | | | |
Collapse
|
189
|
Hasnain SZ, Gallagher AL, Grencis RK, Thornton DJ. A new role for mucins in immunity: insights from gastrointestinal nematode infection. Int J Biochem Cell Biol 2012; 45:364-74. [PMID: 23107603 DOI: 10.1016/j.biocel.2012.10.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/21/2012] [Accepted: 10/23/2012] [Indexed: 12/26/2022]
Abstract
The body's mucosal surfaces are protected from pathogens and physical and chemical attack by the gel-like extracellular matrix, mucus. The framework of this barrier is provided by polymeric, gel-forming mucins. These enormous O-linked glycoproteins are synthesised, stored and secreted by goblet cells that are also the source of other protective factors. Immune regulation of goblet cells during the course of infection impacts on mucin production and properties and ultimately upon barrier function. The barrier function of mucins in protection of the host is well accepted as an important aspect of innate defence. However, it is becoming increasingly clear that mucins have a much more direct role in combating pathogens and parasites and are an important part of the coordinated immune response to infection. Of particular relevance to this review is the finding that mucins are essential anti-parasitic effector molecules. The current understanding of the roles of these multifunctional glycoproteins, and other goblet cell products, in mucosal defence against intestinal dwelling nematodes is discussed.
Collapse
Affiliation(s)
- Sumaira Z Hasnain
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute, Mater Health Services and the University of Queensland, Brisbane, QLD 4029, Australia
| | | | | | | |
Collapse
|
190
|
Inhibitory effect of HIV-specific neutralizing IgA on mucosal transmission of HIV in humanized mice. Blood 2012; 120:4571-82. [PMID: 23065154 DOI: 10.1182/blood-2012-04-422303] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
HIV-1 infections are generally initiated at mucosal sites. Thus, IgA antibody, which plays pivotal roles in mucosal immunity, might efficiently prevent HIV infection. However, mounting a highly effective HIV-specific mucosal IgA response by conventional immunization has been challenging and the potency of HIV-specific IgA against infection needs to be addressed in vivo. Here we show that the polymeric IgA form of anti-HIV antibody inhibits HIV mucosal transmission more effectively than the monomeric IgA or IgG1 form in a comparable range of concentrations in humanized mice. To deliver anti-HIV IgA in a continual manner, we devised a hematopoietic stem/progenitor cell (HSPC)-based genetic approach using an IgA gene. We transplanted human HSPCs transduced with a lentiviral construct encoding a class-switched anti-HIV IgA (b12-IgA) into the humanized bone marrow-liver-thymus (BLT) mice. The transgene was expressed specifically in B cells and plasma cells in lymphoid organs and mucosal sites. After vaginal HIV-1 challenge, mucosal CD4(+) T cells in the b12-IgA-producing mice were protected from virus-mediated depletion. Similar results were also obtained in a second humanized model, "human immune system mice." Our study demonstrates the potential of anti-HIV IgA in immunoprophylaxis in vivo, emphasizing the importance of the mucosal IgA response in defense against HIV/AIDS.
Collapse
|
191
|
Mesin L, Sollid LM, Di Niro R. The intestinal B-cell response in celiac disease. Front Immunol 2012; 3:313. [PMID: 23060888 PMCID: PMC3463893 DOI: 10.3389/fimmu.2012.00313] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 09/18/2012] [Indexed: 12/19/2022] Open
Abstract
The function of intestinal immunity is to provide protection toward pathogens while preserving the composition of the microflora and tolerance to orally fed nutrients. This is achieved via a number of tightly regulated mechanisms including production of IgA antibodies by intestinal plasma cells. Celiac disease is a common gut disorder caused by a dysfunctional immune regulation as signified, among other features, by a massive intestinal IgA autoantibody response. Here we review the current knowledge of this B-cell response and how it is induced, and we discuss key questions to be addressed in future research.
Collapse
Affiliation(s)
- Luka Mesin
- Centre for Immune Regulation, Department of Immunology, Oslo University Hospital-Rikshospitalet, University of Oslo, Oslo, Norway
| | | | | |
Collapse
|
192
|
Zaman M, Abdel-Aal ABM, Fujita Y, Ziora ZM, Batzloff MR, Good MF, Toth I. Structure–Activity Relationship for the Development of a Self-Adjuvanting Mucosally Active Lipopeptide Vaccine against Streptococcus pyogenes. J Med Chem 2012; 55:8515-23. [DOI: 10.1021/jm301074n] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Mehfuz Zaman
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane 4072, Australia
| | - Abu-Baker M. Abdel-Aal
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane 4072, Australia
| | - Yoshio Fujita
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane 4072, Australia
| | - Zyta M. Ziora
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane 4072, Australia
| | | | - Michael F. Good
- Institute for Glycomics, Griffith University, Gold Coast 4215, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane 4072, Australia
- School of Pharmacy, The University
of Queensland, Brisbane, 4072, Australia
| |
Collapse
|
193
|
Development of serum IgA and IgM levels in breast-fed and formula-fed infants during the first week of life. Early Hum Dev 2012; 88:743-5. [PMID: 22784936 DOI: 10.1016/j.earlhumdev.2012.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 03/17/2012] [Accepted: 03/20/2012] [Indexed: 11/23/2022]
Abstract
BACKGROUND IgA and IgM antibodies play important roles to protect infants in early life AIM To study the effects of breast milk feeding versus formula feeding in early infancy on the development of serum IgA and IgM. METHODS A group of 220 healthy infants born after uncomplicated pregnancies and deliveries were enrolled. The infants were divided into three groups according to feeding type: breast-fed (BF), formula-fed (FF), and mixed-fed (MF). Capillary blood was collected for serum IgA and IgM detection at the first week of life. RESULTS The average concentrations of serum IgA and IgM in all infants were 1.171±1.079 and 256.2±165.8 μg/ml, respectively. There were significantly higher concentrations of serum IgA in the FF group than MF group at 3, 4 and 6 days of age and BF group at 5 and 6 days old. Paired serum IgA concentrations revealed that IgA significantly decreased in the BF group, but not in the FF and MF groups. Meanwhile, paired serum IgM concentrations revealed that IgM increased significantly during early infancy in all groups. However, the IgM levels had no difference among the 3 groups within 7 days of age. CONCLUSIONS Our study demonstrated the development of serum IgA and IgM in early life. Formula feeding induced higher serum IgA concentrations than breast-feeding within 7 days of age. However, serum IgM concentration was significantly increased in early life in all groups but had no differences between the different feeding types. Breast-feeding may protect antigen loading in early life.
Collapse
|
194
|
IgA synthesis: a form of functional immune adaptation extending beyond gut. Curr Opin Immunol 2012; 24:261-8. [PMID: 22503962 DOI: 10.1016/j.coi.2012.03.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/22/2012] [Accepted: 03/23/2012] [Indexed: 12/12/2022]
Abstract
Immunoglobulin A (IgA) is the most abundantly produced antibody isotype in mammals. The primary function of IgA is to maintain homeostasis at mucosal surfaces. IgA is generated in specialized gut associated lymphoid tissues (GALT) by T cell-dependent and T cell-independent mechanisms. Studies in mice have demonstrated that IgA diversification has an essential role in the regulation of gut microbiota. Aberrant bacterial growth, by activating innate and adaptive immune cells, has emerged as a risk factor for inflammatory diseases such as metabolic disorders and autoimmune diseases. Dynamic diversification of IgA shields bacterial antigens preventing inflammatory responses, but when IgA regulation is suboptimal aberrant bacterial growth and inflammation can ensue.
Collapse
|
195
|
Association of a protective monoclonal IgA with the O antigen of Salmonella enterica serovar Typhimurium impacts type 3 secretion and outer membrane integrity. Infect Immun 2012; 80:2454-63. [PMID: 22473607 DOI: 10.1128/iai.00018-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Invasion of intestinal epithelial cells by Salmonella enterica serovar Typhimurium is an energetically demanding process, involving the transfer of effector proteins from invading bacteria into host cells via a specialized organelle known as the Salmonella pathogenicity island 1 (SPI-1) type 3 secretion system (T3SS). By a mechanism that remains poorly understood, entry of S. Typhimurium into epithelial cells is inhibited by Sal4, a monoclonal, polymeric IgA antibody that binds an immunodominant epitope within the O-antigen (O-Ag) component of lipopolysaccharide. In this study, we investigated how the binding of Sal4 to the surface of S. Typhimurium influences T3SS activity, bacterial energetics, and outer membrane integrity. We found that Sal4 treatment impaired T3SS-mediated translocon formation and attenuated the delivery of tagged effector proteins into epithelial cells. Sal4 treatment coincided with a partial reduction in membrane energetics and intracellular ATP levels, possibly explaining the impairment in T3SS activity. Sal4's effects on bacterial secretion and energetics occurred concurrently with an increase in O-Ag levels in culture supernatants, alterations in outer membrane permeability, and changes in surface ultrastructure, as revealed by transmission electron microscopy and cryo-electron microscopy. We propose that Sal4, by virtue of its ability to bind and cross-link the O-Ag, induces a form of outer membrane stress that compromises the integrity of the S. Typhimurium cell envelope and temporarily renders the bacterium avirulent.
Collapse
|
196
|
Abstract
UNLABELLED During foetal development, neonatal period and childhood, the immune system is constantly maturing. In the foetus, infection responsiveness is low and associates with spontaneous abortion. During the neonatal period, the infection response shifts towards a more pro-inflammatory response. The immune system of the newborn acquires adaptive features as a result of exposure to microbes. CONCLUSION The development of the human immune system is a continuous process where both accelerated and retarded development is deleterious.
Collapse
Affiliation(s)
- Sofia Ygberg
- The Institution for Woman and Child Health, Unit of Clinical Pediatrics, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
197
|
Kim SH, Jung DI, Yang IY, Kim J, Lee KY, Nochi T, Kiyono H, Jang YS. M cells expressing the complement C5a receptor are efficient targets for mucosal vaccine delivery. Eur J Immunol 2011; 41:3219-29. [PMID: 21887786 DOI: 10.1002/eji.201141592] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 07/21/2011] [Accepted: 08/26/2011] [Indexed: 11/10/2022]
Abstract
In the mucosal immune system, M cells are known as specialized epithelial cells that take up luminal antigens, although the receptors on M cells and the mechanism of antigen uptake into M cells are not well-understood. Here, we report the expression of the complement C5a receptor (C5aR) on the apical surface of M cells. C5ar mRNA expression in co-cultured Caco-2 human M-like cells was six-fold higher than in mono-cultured cells. C5aR expression was detected together with glycoprotein 2, an M-cell-specific protein, on the apical surface of M-like cells and mouse Peyer's patch M cells. Interestingly, after oral administration of Yersinia enterocolitica which expresses outer membrane protein H (OmpH) that is homologous to the Skp α1 domain of Escherichia coli, a ligand of C5aR, dense clustering and phosphorylation of C5aR were detected in M cells. Finally, targeted antigen delivery to M cells using C5aR as a receptor was achieved using the OmpH α1 of Y. enterocolitica such that the induction of ligand-conjugated antigen-specific immune responses was confirmed in mice after oral immunization of the OmpH β1α1-conjugated antigen. Collectively, we identified C5aR expression on M cells and suggest that C5aR could be used as a target receptor for mucosal antigen delivery.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology and Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea
| | | | | | | | | | | | | | | |
Collapse
|
198
|
Meeusen EN. Exploiting mucosal surfaces for the development of mucosal vaccines. Vaccine 2011; 29:8506-11. [PMID: 21945494 DOI: 10.1016/j.vaccine.2011.09.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/19/2011] [Accepted: 09/06/2011] [Indexed: 10/17/2022]
Abstract
Mucosal immunity covers a variety of mucosal surfaces susceptible to different pathogens. This review highlights the diversity of mucosal tissues and the unique microenvironments in which an immune response is generated. It argues that tissue-specific factors present throughout mucosal tissues and lymph nodes determine the differentiation into IgA-producing B cells, which in turn determines their migration patterns. Mucosal immunity can therefore be induced when antigen is delivered at any mucosal tissue without the need for specific 'mucosal adjuvants' or targeting to specialised lymphoid structures. Non-oral vaccination strategies directed at alternative and more accessible mucosal tissue sites, may provide new avenues for both mucosal and systemic immunization, and will be greatly facilitated by the use of large animal models.
Collapse
Affiliation(s)
- Els N Meeusen
- School of Biomedical Sciences, Monash University, Melbourne, Australia.
| |
Collapse
|
199
|
Qiu C, Tian D, Wan Y, Zhang W, Qiu C, Zhu Z, Ye R, Song Z, Zhou M, Yuan S, Shi B, Wu M, Liu Y, Gu S, Wei J, Zhou Z, Zhang X, Zhang Z, Hu Y, Yuan Z, Xu J. Early adaptive humoral immune responses and virus clearance in humans recently infected with pandemic 2009 H1N1 influenza virus. PLoS One 2011; 6:e22603. [PMID: 21886767 PMCID: PMC3160288 DOI: 10.1371/journal.pone.0022603] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 06/25/2011] [Indexed: 11/24/2022] Open
Abstract
Few studies on the humoral immune responses in human during natural influenza infection have been reported. Here, we used serum samples from pandemic 2009 H1N1 influenza infected patients to characterize the humoral immune responses to influenza during natural infection in humans. We observed for the first time that the pandemic 2009 H1N1 influenza induced influenza A-specific IgM within days after symptoms onset, whereas the unit of IgG did not changed. The magnitude of influenza A-specific IgM antibodies might have a value in predicting the rate of virus clearance to some degree. However, the newly developed IgM was not associated with hemagglutination inhibition (HI) activities in the same samples but correlated with HI activities of subsequently collected sera which were mediated by IgG antibodies, indicating that IgM was critical for influenza infection and influences subsequent IgG antibody responses. These findings provide new important insights on the human immunity to natural influenza infection.
Collapse
Affiliation(s)
- Chao Qiu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Di Tian
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yanmin Wan
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wanju Zhang
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chenli Qiu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhaoqin Zhu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ruiqi Ye
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhigang Song
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Mingzhe Zhou
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Songhua Yuan
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Bisheng Shi
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Min Wu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yi Liu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shimin Gu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jun Wei
- Yuncheng CDC, Yuncheng, China
| | - Zhitong Zhou
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory for Infectious Disease Prevention and Control, China CDC, Beijing, China
| | - Zhiyong Zhang
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yunwen Hu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail: (JX); (ZY)
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory for Infectious Disease Prevention and Control, China CDC, Beijing, China
- * E-mail: (JX); (ZY)
| |
Collapse
|
200
|
|