101
|
MERS-CoV Spike Protein Vaccine and Inactivated Influenza Vaccine Formulated with Single Strand RNA Adjuvant Induce T-Cell Activation through Intranasal Immunization in Mice. Pharmaceutics 2020; 12:pharmaceutics12050441. [PMID: 32397649 PMCID: PMC7284860 DOI: 10.3390/pharmaceutics12050441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/06/2020] [Accepted: 05/09/2020] [Indexed: 12/17/2022] Open
Abstract
The effectiveness of vaccines is enhanced by adding adjuvants. Furthermore, the selection of an inoculation route depends on the type of adjuvant used and is important for achieving optimum vaccine efficacy. We investigated the immunological differences between two types of vaccines—spike protein from the Middle East respiratory syndrome virus and inactivated influenza virus vaccine, in combination with a single-stranded RNA adjuvant—administered through various routes (intramuscular, intradermal, and intranasal) to BALB/c mice. Intramuscular immunization with the RNA adjuvant-formulated spike protein elicited the highest humoral immune response, characterized by IgG1 and neutralizing antibody production. Although intranasal immunization did not elicit a humoral response, it showed extensive T-cell activation through large-scale induction of interferon-γ- and interleukin-2-secreting cells, as well as CD4+ T-cell activation in mouse splenocytes. Moreover, only intranasal immunization induced IgA production. When immunized with the inactivated influenza vaccine, administration of the RNA adjuvant via all routes led to protection after viral challenge, regardless of the presence of a vaccine-specific antibody. Therefore, the inoculation route should depend on the type of immune response needed; i.e., the intramuscular route is suitable for eliciting a humoral immune response, whereas the intranasal route is useful for T-cell activation and IgA induction.
Collapse
|
102
|
Gayet R, Michaud E, Nicoli F, Chanut B, Paul M, Rochereau N, Guillon C, He Z, Papagno L, Bioley G, Corthesy B, Paul S. Impact of IgA isoforms on their ability to activate dendritic cells and to prime T cells. Eur J Immunol 2020; 50:1295-1306. [PMID: 32277709 DOI: 10.1002/eji.201948177] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 02/20/2020] [Accepted: 04/07/2020] [Indexed: 12/23/2022]
Abstract
Human IgA could be from different isotypes (IgA1/IgA2) and/or isoforms (monomeric, dimeric, or secretory). Monomeric IgA mainly IgA1 are considered as an anti-inflammatory isotype whereas dimeric/secretory IgA have clearly dual pro- and anti-inflammatory effects. Here, we show that IgA isotypes and isoforms display different binding abilities to FcαRI, Dectin-1, DC-SIGN, and CD71 on monocyte-derived dendritic cells (moDC). We describe that IgA regulate the expression of their own receptors and trigger modulation of moDC maturation. We also demonstrate that dimeric IgA2 and IgA1 induce different inflammatory responses leading to cytotoxic CD8+ T cells activation. moDC stimulation by dimeric IgA2 was followed by a strong pro-inflammatory effect. Our study highlights differences regarding IgA isotypes and isoforms in the context of DC conditioning. Further investigations are needed on the activation of adaptive immunity by IgA in the context of microbiota/IgA complexes during antibody-mediated immune selection.
Collapse
Affiliation(s)
- Rémi Gayet
- GIMAP/EA3064, Université de Lyon, Saint-Etienne, France
| | - Eva Michaud
- GIMAP/EA3064, Université de Lyon, Saint-Etienne, France
| | - Francesco Nicoli
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Universités, Paris, France
| | | | - Mireille Paul
- SAINBIOSE, INSERM U1059, University of Lyon, Saint-Etienne, France
| | | | - Christophe Guillon
- Retroviruses and Structural Biochemistry, Institut de Biologie et Chimie des Protéines, University of Lyon, CNRS, UMR5086, Lyon, France
| | - Zhiguo He
- BiiGC/EA2521, Université de Lyon, Saint-Etienne, France
| | - Laura Papagno
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Universités, Paris, France
| | - Gilles Bioley
- BiiGC/EA2521, Université de Lyon, Saint-Etienne, France
| | - Blaise Corthesy
- R&D Laboratory of the Division of Immunology and Allergy, CHUV, Centre des Laboratoires d'Epalinges, Epalinges, Switzerland
| | - Stéphane Paul
- GIMAP/EA3064, Université de Lyon, Saint-Etienne, France
| |
Collapse
|
103
|
Verma A, Schmidt BA, Elizaldi SR, Nguyen NK, Walter KA, Beck Z, Trinh HV, Dinasarapu AR, Lakshmanappa YS, Rane NN, Matyas GR, Rao M, Shen X, Tomaras GD, LaBranche CC, Reimann KA, Foehl DH, Gach JS, Forthal DN, Kozlowski PA, Amara RR, Iyer SS. Impact of T h1 CD4 Follicular Helper T Cell Skewing on Antibody Responses to an HIV-1 Vaccine in Rhesus Macaques. J Virol 2020; 94:e01737-19. [PMID: 31827000 PMCID: PMC7158739 DOI: 10.1128/jvi.01737-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Generating durable humoral immunity through vaccination depends upon effective interactions of follicular helper T (Tfh) cells with germinal center (GC) B cells. Th1 polarization of Tfh cells is an important process shaping the success of Tfh-GC B cell interactions by influencing costimulatory and cytokine-dependent Tfh help to B cells. However, the question remains as to whether adjuvant-dependent modulation of Tfh cells enhances HIV-1 vaccine-induced antienvelope (anti-Env) antibody responses. We investigated whether an HIV-1 vaccine platform designed to increase the number of Th1-polarized Tfh cells enhances the magnitude and quality of anti-Env antibodies. Utilizing a novel interferon-induced protein 10 (IP-10)-adjuvanted HIV-1 DNA prime followed by a monophosphoryl lipid A and QS-21 (MPLA+QS-21)-adjuvanted Env protein boost (DIP-10 PALFQ) in macaques, we observed higher anti-Env serum IgG titers with greater cross-clade reactivity, specificity for V1V2, and effector functions than in macaques primed with DNA lacking IP-10 and boosted with MPLA-plus-alum-adjuvanted Env protein (DPALFA) The DIP-10 PALFQ vaccine regimen elicited higher anti-Env IgG1 and lower IgG4 antibody levels in serum, showing for the first time that adjuvants can dramatically impact the IgG subclass profile in macaques. The DIP-10 PALFQ regimen also increased vaginal and rectal IgA antibodies to a greater extent. Within lymph nodes, we observed augmented GC B cell responses and the promotion of Th1 gene expression profiles in GC Tfh cells. The frequency of GC Tfh cells correlated with both the magnitude and avidity of anti-Env serum IgG. Together, these data suggest that adjuvant-induced stimulation of Th1-Tfh cells is an effective strategy for enhancing the magnitude and quality of anti-Env antibody responses.IMPORTANCE The results of the RV144 trial demonstrated that vaccination could prevent HIV transmission in humans and that longevity of anti-Env antibodies may be key to this protection. Efforts to improve upon the prime-boost vaccine regimen used in RV144 have indicated that booster immunizations can increase serum anti-Env antibody titers but only transiently. Poor antibody durability hampers efforts to develop an effective HIV-1 vaccine. This study was designed to identify the specific elements involved in the immunological mechanism necessary to produce robust HIV-1-specific antibodies in rhesus macaques. By clearly defining immune-mediated pathways that improve the magnitude and functionality of the anti-HIV-1 antibody response, we will have the foundation necessary for the rational development of an HIV-1 vaccine.
Collapse
Affiliation(s)
- Anil Verma
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Brian A Schmidt
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Sonny R Elizaldi
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- Graduate Group in Immunology, UC Davis, Davis, California, USA
| | - Nancy K Nguyen
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Korey A Walter
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zoltan Beck
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Hung V Trinh
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ashok R Dinasarapu
- Emory Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | | | - Niharika N Rane
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Gary R Matyas
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Celia C LaBranche
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Keith A Reimann
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - David H Foehl
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Johannes S Gach
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Donald N Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Smita S Iyer
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- California National Primate Research Center, School of Veterinary Medicine, UC Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, California, USA
| |
Collapse
|
104
|
Host Factors Affecting Generation of Immunity Against Porcine Epidemic Diarrhea Virus in Pregnant and Lactating Swine and Passive Protection of Neonates. Pathogens 2020; 9:pathogens9020130. [PMID: 32085410 PMCID: PMC7168134 DOI: 10.3390/pathogens9020130] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly virulent re-emerging enteric coronavirus that causes acute diarrhea, dehydration, and up to 100% mortality in neonatal suckling piglets. Despite this, a safe and effective PEDV vaccine against highly virulent strains is unavailable, making PEDV prevention and control challenging. Lactogenic immunity induced via the gut-mammary gland-secretory IgA (sIgA) axis, remains the most promising and effective way to protect suckling piglets from PEDV. Therefore, a successful PEDV vaccine must induce protective maternal IgA antibodies that passively transfer into colostrum and milk. Identifying variables that influence lymphocyte migration and IgA secretion during gestation and lactation is imperative for designing maternal immunization strategies that generate the highest amount of lactogenic immune protection against PEDV in suckling piglets. Because pregnancy-associated immune alterations influence viral pathogenesis and adaptive immune responses in many different species, a better understanding of host immune responses to PEDV in pregnant swine may translate into improved maternal immunization strategies against enteric pathogens for multiple species. In this review, we discuss the role of host factors during pregnancy on antiviral immunity and their implications for generating protective lactogenic immunity in suckling neonates.
Collapse
|
105
|
Bai Y, Wang G, Qi H, Wang Y, Xu C, Yue L, Hou X, Yu L. Immunogenicity of 987P fimbriae of enterotoxigenic Escherichia coli surface-displayed on Lactobacillus casei. Res Vet Sci 2020; 128:308-314. [DOI: 10.1016/j.rvsc.2019.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 12/10/2019] [Accepted: 12/22/2019] [Indexed: 12/27/2022]
|
106
|
Liu J, Clayton K, Gao W, Li Y, Zealey C, Budylowski P, Schwartz J, Yue FY, Bie Y, Rini J, Ostrowski M. Trimeric HIV-1 gp140 fused with APRIL, BAFF, and CD40L on the mucosal gp140-specific antibody responses in mice. Vaccine 2020; 38:2149-2159. [PMID: 32014267 DOI: 10.1016/j.vaccine.2020.01.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/04/2019] [Accepted: 01/19/2020] [Indexed: 12/12/2022]
Abstract
HIV-1 envelope (Env)-specific antibody present at mucosal surfaces can block entry of HIV-1 into these portals and thus should be elicited by an HIV-1 preventive vaccine. Since three molecules of tumor necrosis factor superfamily (TNFSF), APRIL, BAFF, and CD40L, could promote mucosal antibody responses, we made fusion constructs of them with an HIV-1 gp140 trimer and tested the mucosal gp140-specific antibody elicited by the fusion constructs in mice using a DNA prime-protein boost vaccination regimen. The fusion constructs formed trimers and displayed both broadly neutralizing antibody epitopes and non-broadly neutralizing antibody epitopes. Compared with the control construct, trimeric gp140, trimeric gp140-APRIL and gp140-BAFF fusion proteins mildly promoted B cell proliferation in vitro, enhanced HIV-1 gp140-binding IgG responses in vaginal lavage or fecal pellets, respectively, and decreased HIV-1 gp140-binding IgA in sera. Gp140-APRIL also augmented HIV-1 gp140-binding IgG in sera. Surprisingly, gp140-CD40L did not promote B cell proliferation in vitro and inhibited mucosal and systemic HIV-1 gp140-binding IgG or IgA. These results suggest that APRIL and BAFF should be further explored as molecular adjuvants for HIV-1 vaccines to enhance mucosal antibody responses, but covalent fusion of TNFSFs to gp140 may hinder their adjuvancy due to steric interactions.
Collapse
Affiliation(s)
- Jun Liu
- Clinical Sciences Division, University of Toronto, Toronto, Ontario, Canada.
| | - Kiera Clayton
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Wenbo Gao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yu Li
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Chris Zealey
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Budylowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Jordan Schwartz
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Feng Yun Yue
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yuan Bie
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - James Rini
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Clinical Sciences Division, University of Toronto, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
107
|
The Effects of Secretory IgA in the Mucosal Immune System. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2032057. [PMID: 31998782 PMCID: PMC6970489 DOI: 10.1155/2020/2032057] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/14/2019] [Indexed: 12/18/2022]
Abstract
Immunoglobulin A (IgA) is the most abundant antibody isotype in the mucosal immune system. Structurally, IgA in the mucosal surface is a polymeric structure, while serum IgA is monomeric. Secretory IgA (sIgA) is one of the polymeric IgAs composed of dimeric IgA, J chain, and secretory component (SC). Most of sIgAs were generated by gut and have effects in situ. Besides the function of “immune exclusion,” a nonspecific immune role, recent studies found it also played an important role in the specific immunity and immunoregulation. Thanks to the critical role of sIgA during the mucosal immune system homeostasis between commensal microorganisms and pathogens; it has been an important field exploring the relationship between sIgA and commensal microorganisms.
Collapse
|
108
|
Pabst O, Slack E. IgA and the intestinal microbiota: the importance of being specific. Mucosal Immunol 2020; 13:12-21. [PMID: 31740744 PMCID: PMC6914667 DOI: 10.1038/s41385-019-0227-4] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 02/04/2023]
Abstract
Secretory IgA has long been a divisive molecule. Some immunologists point to the mild phenotype of IgA deficiency to justify ignoring it, while some consider its abundance and evolutionary history as grounds for its importance. Further, there is extensive and growing disagreement over the relative importance of affinity-matured, T cell-dependent IgA vs. "natural" and T cell-independent IgA in both microbiota and infection control. As with all good arguments, there is good data supporting different opinions. Here we revisit longstanding questions in IgA biology. We start the discussion from the question of intestinal IgA antigen specificity and critical definitions regarding IgA induction, specificity, and function. These definitions must then be tessellated with the cellular and molecular pathways shaping IgA responses, and the mechanisms by which IgA functions. On this basis we propose how IgA may contribute to the establishment and maintenance of beneficial interactions with the microbiota.
Collapse
Affiliation(s)
- Oliver Pabst
- 0000 0001 0728 696Xgrid.1957.aInstitute of Molecular Medicine, RWTH Aachen University, Aachen, Germany
| | - Emma Slack
- 0000 0001 2156 2780grid.5801.cInstitute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
109
|
Probiotic Propionibacterium freudenreichii requires SlpB protein to mitigate mucositis induced by chemotherapy. Oncotarget 2019; 10:7198-7219. [PMID: 31921383 PMCID: PMC6944450 DOI: 10.18632/oncotarget.27319] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Propionibacterium freudenreichii CIRM-BIA 129 (P. freudenreichii wild type, WT) is a probiotic bacterium, which exerts immunomodulatory effects. This strain possesses extractable surface proteins, including SlpB, which are involved in anti-inflammatory effect and in adhesion to epithelial cells. We decided to investigate the impact of slpB gene mutation on immunomodulation in vitro and in vivo. In an in vitro assay, P. freudenreichii WT reduced expression of IL-8 (p<0.0001) and TNF-α (p<0.0001) cytokines in LPS-stimulated HT-29 cells. P. freudenreichii ΔslpB, lacking the SlpB protein, failed to do so. Subsequently, both strains were investigated in vivo in a 5-FU-induced mucositis mice model. Mucositis is a common side effect of cytotoxic chemotherapy with 5-FU, characterized by mucosal injury, inflammation, diarrhea, and weight loss. The WT strain prevented weight loss, reduced inflammation and consequently histopathological scores. Furthermore, it regulated key markers, including Claudin-1 (cld1, p<0.0005) and IL-17a (Il17a, p<0.0001) genes, as well as IL-12 (p<0.0001) and IL-1β (p<0.0429) cytokines levels. Mutant strain displayed opposite regulatory effect on cld1 expression and on IL-12 levels. This work emphasizes the importance of SlpB in P. freudenreichii ability to reduce mucositis inflammation. It opens perspectives for the development of probiotic products to decrease side effects of chemotherapy using GRAS bacteria with immunomodulatory surface protein properties.
Collapse
|
110
|
Ren Z, Bütz DE, Whelan R, Naranjo V, Arendt MK, Ramuta MD, Yang X, Crenshaw TD, Cook ME. Effects of dietary methionine plus cysteine levels on growth performance and intestinal antibody production in broilers during Eimeria challenge. Poult Sci 2019; 99:374-384. [PMID: 32416822 PMCID: PMC7587792 DOI: 10.3382/ps/pez503] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Research has shown that methionine+ cysteine (M+C) requirements may be higher when chickens are infected with Eimeria app. In a 4 × 2 factorial design, broilers (11 to 21 D) were fed one of 4 corn–soybean meal-based diets containing either 0.6, 0.8, 0.9, or 1.0% standardized ileal digestible (SID) M+C; on day 14, broilers from each diet were gavaged with either phosphate-buffered saline (PBS) or a commercial coccidiosis vaccine (at 100 × vaccine dose) which provide a mixture of live Eimeria acervulina, Eimeria maxima, and Eimeria tenella oocysts. Growth performance was recorded from day 11 to 21. Plasma and intestinal luminal samples were collected on days 14 and 21. Intestine lesion scores and fecal oocyst counts were conducted on day 21. Regardless of dietary SID M+C levels, compared to PBS gavaged broilers, the Eimeria-challenged broilers had (1) decreased (P < 0.05) body weight gain (BWG), feed intake (FI), and gain-to-feed ratio (G:F); (2) increased (P < 0.05) intestinal lesion scores and fecal oocyst counts; (3) increased (P < 0.05) plasma anti-Eimeria IgG, and intestinal luminal total IgA and anti-Eimeria IgA concentrations; and (4) increased (P < 0.05) levels of duodenum luminal gamma interferon (IFN-γ) and interleukin-10 (IL-10), as well as jejunum and cecum luminal IFN-γ concentrations. Regardless of Eimeria challenge, when compared to 0.6% SID M+C, broilers fed ≥0.8% SID M+C had (1) increased (P < 0.05) BWG, FI, and G:F and (2) increased (P < 0.05) levels of jejunum luminal total IgA. After Eimeria challenge, broilers fed 0.8% SID M+C had increased (P < 0.05) levels of jejunum luminal anti-Eimeria IgA compared to broilers fed diets containing 0.6 and 1.0% SID M+C. Collectively, in 11- to 21-D broilers, the growth suppression caused by Eimeria infection could not be mitigated by further increasing dietary M+C alone ≥0.8%. Further research should investigate interactions between dietary M+C and other nutrients for support of immune function and growth in pathogen-challenged broilers.
Collapse
Affiliation(s)
- Zhouzheng Ren
- College of Animal Science and Technology, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi 712100, China.
| | - Daniel E Bütz
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA
| | - Rose Whelan
- Evonik Nutrition & Care GmbH, 4 Rodenbacher Chaussee, Hanau-Wolfgang 63457, Germany
| | - Victor Naranjo
- Evonik Nutrition & Care GmbH, 4 Rodenbacher Chaussee, Hanau-Wolfgang 63457, Germany
| | - Maria K Arendt
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA
| | - Mitchell D Ramuta
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Thomas D Crenshaw
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA
| | - Mark E Cook
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA
| |
Collapse
|
111
|
Fujimoto K, Uematsu S. Development of prime-boost-type next-generation mucosal vaccines. Int Immunol 2019; 32:597-603. [PMID: 31882997 DOI: 10.1093/intimm/dxz085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 12/27/2019] [Indexed: 12/11/2022] Open
Abstract
Our bodies are constantly exposed to a wide variety of pathogenic micro-organisms through mucosal sites. Therefore, effective vaccines that can protect at the mucosa are vital; however, only a few clinically established mucosal vaccines are available. Although conventional injectable vaccines can induce antigen-specific serum immunoglobulin G (IgG) and prevent severe infection, it is difficult to efficiently inhibit the invasion of pathogens at mucosal surfaces because of the inadequate ability to induce antigen-specific IgA. Recently, we have developed a parenteral vaccine with emulsified curdlan and CpG oligodeoxynucleotides and reported its application. Unlike other conventional injectable vaccines, this immunization contributes to the induction of antigen-specific mucosal and systemic immune responses. Even if antigen-specific IgA at the mucosa disappears, this immunization can induce high-titer IgA after boosting with a small amount of antigen on the target mucosal surface. Indeed, vaccination with Streptococcus pneumoniae antigen effectively prevented lung infection induced by this bacterium. In addition, vaccination with Clostridium ramosum, which is a representative pathobiont associated with obesity and diabetes in humans, reduced obesity in mice colonized with this microorganism. This immunization approach might be an effective treatment for intestinal bacteria-mediated diseases that have been difficult to regulate so far, as well as common infectious diseases.
Collapse
Affiliation(s)
- Kosuke Fujimoto
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Asahi-machi, Abeno-ku, Osaka, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Asahi-machi, Abeno-ku, Osaka, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan.,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Yayoi, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
112
|
Pathak M, Padghan P, Halder N, Shilpi, Kulkarni N, Sonar SA, Lal G. CCR9 signaling in dendritic cells drives the differentiation of Foxp3 + Tregs and suppresses the allergic IgE response in the gut. Eur J Immunol 2019; 50:404-417. [PMID: 31755547 DOI: 10.1002/eji.201948327] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/11/2019] [Accepted: 11/20/2019] [Indexed: 12/29/2022]
Abstract
The chemokine receptor CCR9 and its only known ligand CCL25 play an important role in gut inflammation and autoimmune colitis. The function of CCR9-CCL25 in the migration of immune cells is well characterized. However, its role in the immune cell differentiation is mostly not known. Using dextran sodium sulfate (DSS)-induced gut inflammation model, we showed that CCR9+ dendritic cells (DCs) specifically CD11b- CD103+ DCs were significantly increased in the gut-associated lymphoid tissues (GALT) compared to control mice. These CCR9+ DCs express lower MHC II and CD86 molecules and had regulatory surface markers (FasL and latency-associated peptide, LAP) in the GALT. In the presence of CCL25, CCR9+ DCs promoted in vitro differentiation of Foxp3+ regulatory CD4+ T cells (Tregs). CCL25-induced differentiation of Tregs was due to intrinsic signaling in the DCs but not through CD4+ T cells, which was driven by the production of thymic stromal lymphopoietin (TSLP) and not IL-10. Furthermore, adoptive transfer of CCR9+ DCs in C57BL/6 mice promoted Tregs but reduced the Th17 cells in the GALT, and also suppressed the OVA-specific gut-allergic response. Our results suggest CCR9+ DCs have a regulatory function and may provide a new cellular therapeutic strategy to control gut inflammation and allergic immune reaction.
Collapse
Affiliation(s)
- Manisha Pathak
- National Centre for Cell Science, SP Pune University campus, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Priyanka Padghan
- National Centre for Cell Science, SP Pune University campus, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Namrita Halder
- National Centre for Cell Science, SP Pune University campus, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Shilpi
- National Centre for Cell Science, SP Pune University campus, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Neeraja Kulkarni
- National Centre for Cell Science, SP Pune University campus, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Sandip A Sonar
- National Centre for Cell Science, SP Pune University campus, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Girdhari Lal
- National Centre for Cell Science, SP Pune University campus, Ganeshkhind, Pune, Maharashtra, 411007, India
| |
Collapse
|
113
|
Arciniega-Martínez IM, Drago-Serrano ME, Salas-Pimentel M, Ventura-Juárez J, Reséndiz-Albor AA, Campos-Rodríguez R. Anterior subdiaphragmatic vagotomy decreases the IgA antibody response in the small intestines of BALB/c mice. J Neuroimmunol 2019; 337:577072. [DOI: 10.1016/j.jneuroim.2019.577072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/04/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023]
|
114
|
Livshits G, Kalinkovich A. Inflammaging as a common ground for the development and maintenance of sarcopenia, obesity, cardiomyopathy and dysbiosis. Ageing Res Rev 2019; 56:100980. [PMID: 31726228 DOI: 10.1016/j.arr.2019.100980] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022]
Abstract
Sarcopenia, obesity and their coexistence, obese sarcopenia (OBSP) as well as atherosclerosis-related cardio-vascular diseases (ACVDs), including chronic heart failure (CHF), are among the greatest public health concerns in the ageing population. A clear age-dependent increased prevalence of sarcopenia and OBSP has been registered in CHF patients, suggesting mechanistic relationships. Development of OBSP could be mediated by a crosstalk between the visceral and subcutaneous adipose tissue (AT) and the skeletal muscle under conditions of low-grade local and systemic inflammation, inflammaging. The present review summarizes the emerging data supporting the idea that inflammaging may serve as a mutual mechanism governing the development of sarcopenia, OBSP and ACVDs. In support of this hypothesis, various immune cells release pro-inflammatory mediators in the skeletal muscle and myocardium. Subsequently, the endothelial structure is disrupted, and cellular processes, such as mitochondrial activity, mitophagy, and autophagy are impaired. Inflamed myocytes lose their contractile properties, which is characteristic of sarcopenia and CHF. Inflammation may increase the risk of ACVD events in a hyperlipidemia-independent manner. Significant reduction of ACVD event rates, without the lowering of plasma lipids, following a specific targeting of key pro-inflammatory cytokines confirms a key role of inflammation in ACVD pathogenesis. Gut dysbiosis, an imbalanced gut microbial community, is known to be deeply involved in the pathogenesis of age-associated sarcopenia and ACVDs by inducing and supporting inflammaging. Dysbiosis induces the production of trimethylamine-N-oxide (TMAO), which is implicated in atherosclerosis, thrombosis, metabolic syndrome, hypertension and poor CHF prognosis. In OBSP, AT dysfunction and inflammation induce, in concert with dysbiosis, lipotoxicity and other pathophysiological processes, thus exacerbating sarcopenia and CHF. Administration of specialized, inflammation pro-resolving mediators has been shown to ameliorate the inflammatory manifestations. Considering all these findings, we hypothesize that sarcopenia, OBSP, CHF and dysbiosis are inflammaging-oriented disorders, whereby inflammaging is common and most probably the causative mechanism driving their pathogenesis.
Collapse
Affiliation(s)
- Gregory Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel.; Adelson School of Medicine, Ariel University, Ariel, Israel..
| | - Alexander Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
115
|
Song B, Zheng C, Zha C, Hu S, Yang X, Wang L, Xiao H. Dietary leucine supplementation improves intestinal health of mice through intestinal SIgA secretion. J Appl Microbiol 2019; 128:574-583. [PMID: 31562837 DOI: 10.1111/jam.14464] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022]
Abstract
AIMS Leucine supplementation promotes intestinal health, but the mechanism is largely unknown. This study aimed to elucidate the mechanisms underlying the beneficial effects of leucine on intestinal homeostasis. METHODS AND RESULTS Female ICR mice (6-week-old) were randomly assigned into three groups: (i) mice received a basal diet; (ii) mice received a dietary 0·5% crystalline l-leucine supplementation; and (iii) mice received a dietary 1·0% crystalline l-leucine supplementation. Our results showed that leucine supplementation stimulated the secretion of SIgA in mice ileum and expression of cytokines related to SIgA production. Moreover, leucine supplementation improved the expression of mTOR and p70S6K1 expression. Further study showed that leucine supplementation markedly decreased microbiota richness and induced a shift in the Firmicutes : Bacteroidetes ratio in favour of Firmicutes. CONCLUSIONS Therefore, our data suggested that leucine supplementation could enhance intestinal health through the regulation of mTOR pathway and promoting SIgA secretion in the mouse intestine, which might be associated with intestinal microbiota. SIGNIFICANCE AND IMPACT OF THE STUDY The present study found that dietary leucine supplementation of mice could improve intestinal health by enhancing intestinal SIgA secretion via a nonexclusive mechanism, which might include T cell-dependent pathway, T cell-independent pathway and gut microbiota.
Collapse
Affiliation(s)
- B Song
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - C Zheng
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - C Zha
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - S Hu
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - X Yang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - L Wang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - H Xiao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
116
|
Onodera T, Hashi K, Shukla RK, Miki M, Takai-Todaka R, Fujimoto A, Kuraoka M, Miyoshi T, Kobayashi K, Hasegawa H, Ato M, Kelsoe G, Katayama K, Takahashi Y. Immune-Focusing Properties of Virus-like Particles Improve Protective IgA Responses. THE JOURNAL OF IMMUNOLOGY 2019; 203:3282-3292. [PMID: 31704880 DOI: 10.4049/jimmunol.1900481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023]
Abstract
Virus-like particles (VLPs) provide a well-established vaccine platform; however, the immunogenic properties acquired by VLP structure remain poorly understood. In this study, we showed that systemic vaccination with norovirus VLP recalls human IgA responses at higher magnitudes than IgG responses under a humanized mouse model that was established by introducing human PBMCs in severely immunodeficient mice. The recall responses elicited by VLP vaccines depended on VLP structure and the disruption of VLP attenuated recall responses, with a more profound reduction being observed in IgA responses. The IgA-focusing property was also conserved in a murine norovirus-primed model under which murine IgA responses were recalled in a manner dependent on VLP structure. Importantly, the VLP-driven IgA response preferentially targeted virus-neutralizing epitopes located in the receptor-binding domain. Consequently, VLP-driven IgA responses were qualitatively superior to IgG responses in terms of the virus-neutralizing activity in vitro. Furthermore, the IgA in mucosa obtained remarkable protective function toward orally administrated virus in vivo. Thus, our results indicate the immune-focusing properties of the VLP vaccine that improve the quality/quantity of mucosal IgA responses, a finding with important implications for developing mucosal vaccines.
Collapse
Affiliation(s)
- Taishi Onodera
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kana Hashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Rajni Kant Shukla
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Motohiro Miki
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan.,Vaccine & Biomedicine Department, Life Innovation Research Institute, Denka Innovation Center, Denka Co., Ltd., Tokyo 194-8560, Japan
| | - Reiko Takai-Todaka
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan
| | - Akira Fujimoto
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan
| | - Masayuki Kuraoka
- Department of Immunology and Human Vaccine Institute, Duke University, Durham, NC 27710
| | - Tatsuya Miyoshi
- Sakai City Institute of Public Health, Osaka 590-0953, Japan
| | - Kazuo Kobayashi
- Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Manabu Ato
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Garnett Kelsoe
- Department of Immunology and Human Vaccine Institute, Duke University, Durham, NC 27710
| | - Kazuhiko Katayama
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;
| |
Collapse
|
117
|
Class-switch recombination to IgA in the Peyer's patches requires natural thymus-derived Tregs and appears to be antigen independent. Mucosal Immunol 2019; 12:1268-1279. [PMID: 31501516 DOI: 10.1038/s41385-019-0202-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/05/2019] [Accepted: 08/15/2019] [Indexed: 02/04/2023]
Abstract
Our understanding of how class-switch recombination (CSR) to IgA occurs in the gut is still incomplete. Earlier studies have indicated that Tregs are important for IgA CSR and these cells were thought to transform into follicular helper T cells (Tfh), responsible for germinal center formation in the Peyer's patches (PP). Following adoptive transfer of T-cell receptor-transgenic (TCR-Tg) CD4 T cells into nude mice, we unexpectedly found that oral immunization did not require an adjuvant to induce strong gut IgA and systemic IgG responses, suggesting an altered regulatory environment in the PP. After sorting of splenic TCR-Tg CD4 T cells into CD25+ or CD25- cells we observed that none of these fractions supported a gut IgA response, while IgG responses were unperturbed in mice receiving the CD25- cell fraction. Hence, while Tfh functions resided in the CD25- fraction the IgA CSR function in the PP was dependent on CD25+ Foxp3+ Tregs, which were found to be Helios+ neuropilin-1+ thymus-derived Tregs. This is the first study to demonstrate that Tfh and IgA CSR functions are indeed, unique, and separate functions in the PP with the former being TCR-dependent while the latter appeared to be antigen independent.
Collapse
|
118
|
Albright AR, Kabat J, Li M, Raso F, Reboldi A, Muppidi JR. TGFβ signaling in germinal center B cells promotes the transition from light zone to dark zone. J Exp Med 2019; 216:2531-2545. [PMID: 31506281 PMCID: PMC6829600 DOI: 10.1084/jem.20181868] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/18/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022] Open
Abstract
GCs are polarized into LZ and DZ to allow for the spatial separation of selection and proliferation. In this study, we describe the previously unknown function of TGFβ in promoting the transition of GCBs from LZ to DZ. B cells in germinal centers (GCs) cycle between light zone (LZ) and dark zone (DZ). The cues in the GC microenvironment that regulate the transition from LZ to DZ have not been well characterized. In Peyer’s patches (PPs), transforming growth factor-β (TGFβ) promotes IgA induction in activated B cells that can then differentiate into GC B cells. We show here that TGFβ signaling occurs in B cells in GCs and is distinct from signaling that occurs in activated B cells in PPs. Whereas in activated B cells TGFβ signaling is required for IgA induction, in the GC it was instead required for the transition from LZ to DZ. In the absence of TGFβ signaling, there was an accumulation of LZ GC B cells and reduced antibody affinity maturation likely due to reduced activation of Foxo1. This work identifies TGFβ as a microenvironmental cue that is critical for GC homeostasis and function.
Collapse
Affiliation(s)
- Anne R Albright
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Moyi Li
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Fiona Raso
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA
| | - Jagan R Muppidi
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
119
|
BLT1 mediates commensal bacteria-dependent innate immune signals to enhance antigen-specific intestinal IgA responses. Mucosal Immunol 2019; 12:1082-1091. [PMID: 31142830 DOI: 10.1038/s41385-019-0175-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 04/26/2019] [Accepted: 05/08/2019] [Indexed: 02/06/2023]
Abstract
Leukotriene B4 receptor 1 (BLT1) triggers the migration of granulocytes and activated T cells; however, its role in B-cell function remains unclear. Here we report that BLT1 is required to induce the production of antigen-specific IgA against oral vaccine through mediating innate immune signals from commensal bacteria. B cells acquire BLT1 expression during their differentiation to IgA+ B cells and plasma cells in Peyer's patches and the small intestinal lamina propria, respectively. BLT1 KO mice exhibited impaired production of antigen-specific fecal IgA to oral vaccine despite normal IgG responses to systemically immunized antigen. Expression of MyD88 was decreased in BLT1 KO gut B cells and consequently led to diminished proliferation of commensal bacteria-dependent plasma cells. These results indicate that BLT1 enhances the proliferation of commensal bacteria-dependent IgA+ plasma cells through the induction of MyD88 and thereby plays a key role in the production of antigen-specific intestinal IgA.
Collapse
|
120
|
Chen XY, Butt AM, Mohd Amin MCI. Molecular Evaluation of Oral Immunogenicity of Hepatitis B Antigen Delivered by Hydrogel Microparticles. Mol Pharm 2019; 16:3853-3872. [PMID: 31398038 DOI: 10.1021/acs.molpharmaceut.9b00483] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of oral vaccine formulation is crucial to facilitate an effective mass immunization program for various vaccine-preventable diseases. In this work, the efficacy of hepatitis B antigen delivered by bacterial nanocellulose/poly(acrylic acid) composite hydrogel microparticles (MPs) as oral vaccine carriers was assessed to induce both local and systemic immunity. Optimal pH-responsive swelling, mucoadhesiveness, protein drug loading, and drug permeability were characterized by MPs formulated with minimal irradiation doses and acrylic acid concentration. The composite hydrogel materials of bacterial nanocellulose and poly(acrylic acid) showed significantly greater antigen release in simulated intestinal fluid while ensuring the integrity of antigen. In in vivo study, mice orally vaccinated with antigen-loaded hydrogel MPs showed enhanced vaccine immunogenicity with significantly higher secretion of mucosal immunoglobulin A, compared to intramuscular vaccinated control. The splenocytes from the same group demonstrated lymphoproliferation and significant increased secretion of interleukin-2 cytokines upon stimulation with hepatitis B antigen. Expression of CD69 in CD4+ T lymphocytes and CD19+ B lymphocytes in splenocytes from mice orally vaccinated with antigen-loaded hydrogel MPs was comparable to that of the intramuscular vaccinated control, indicating early activation of lymphocytes elicited by our oral vaccine formulation in just two doses. These results demonstrated the potential of antigen-loaded hydrogel MPs as an oral vaccination method for hepatitis B.
Collapse
Affiliation(s)
- Xiang Yi Chen
- Centre for Drug Delivery Research, Faculty of Pharmacy , Universiti Kebangsaan Malaysia , Jalan Raja Muda Abdul Aziz , 50300 Kuala Lumpur , Malaysia
| | - Adeel Masood Butt
- Department of Pharmacy , The University of Lahore , Gujrat Campus, Adjacent Chenab Bridge, Main GT Road , 50700 Gujrat , Pakistan
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Research, Faculty of Pharmacy , Universiti Kebangsaan Malaysia , Jalan Raja Muda Abdul Aziz , 50300 Kuala Lumpur , Malaysia
| |
Collapse
|
121
|
Fasting-Refeeding Impacts Immune Cell Dynamics and Mucosal Immune Responses. Cell 2019; 178:1072-1087.e14. [DOI: 10.1016/j.cell.2019.07.047] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/30/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
|
122
|
Komban RJ, Strömberg A, Biram A, Cervin J, Lebrero-Fernández C, Mabbott N, Yrlid U, Shulman Z, Bemark M, Lycke N. Activated Peyer's patch B cells sample antigen directly from M cells in the subepithelial dome. Nat Commun 2019; 10:2423. [PMID: 31160559 PMCID: PMC6547658 DOI: 10.1038/s41467-019-10144-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/23/2019] [Indexed: 01/13/2023] Open
Abstract
The germinal center (GC) reaction in Peyer's patches (PP) requires continuous access to antigens, but how this is achieved is not known. Here we show that activated antigen-specific CCR6+CCR1+GL7- B cells make close contact with M cells in the subepithelial dome (SED). Using in situ photoactivation analysis of antigen-specific SED B cells, we find migration of cells towards the GC. Following antigen injection into ligated intestinal loops containing PPs, 40% of antigen-specific SED B cells bind antigen within 2 h, whereas unspecifc cells do not, indicating B cell-receptor involvment. Antigen-loading is not observed in M cell-deficient mice, but is unperturbed in mice depleted of classical dendritic cells (DC). Thus, we report a M cell-B cell antigen-specific transporting pathway in PP that is independent of DC. We propose that this antigen transporting pathway has a critical role in gut IgA responses, and should be taken into account when developing mucosal vaccines.
Collapse
Affiliation(s)
- Rathan Joy Komban
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Anneli Strömberg
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Adi Biram
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Jakob Cervin
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Cristina Lebrero-Fernández
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Neil Mabbott
- The Roslin Institute, Edinburgh University, Edinburgh, EH25 9RG, Scotland
| | - Ulf Yrlid
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Ziv Shulman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Mats Bemark
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden.
| | - Nils Lycke
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden.
| |
Collapse
|
123
|
Cyster JG, Allen CDC. B Cell Responses: Cell Interaction Dynamics and Decisions. Cell 2019; 177:524-540. [PMID: 31002794 PMCID: PMC6538279 DOI: 10.1016/j.cell.2019.03.016] [Citation(s) in RCA: 623] [Impact Index Per Article: 103.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/21/2022]
Abstract
B cells and the antibodies they produce have a deeply penetrating influence on human physiology. Here, we review current understanding of how B cell responses are initiated; the different paths to generate short- and long-lived plasma cells, germinal center cells, and memory cells; and how each path impacts antibody diversity, selectivity, and affinity. We discuss how basic research is informing efforts to generate vaccines that induce broadly neutralizing antibodies against viral pathogens, revealing the special features associated with allergen-reactive IgE responses and uncovering the antibody-independent mechanisms by which B cells contribute to health and disease.
Collapse
Affiliation(s)
- Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Christopher D C Allen
- Cardiovascular Research Institute, Department of Anatomy, and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
124
|
BCR affinity differentially regulates colonization of the subepithelial dome and infiltration into germinal centers within Peyer's patches. Nat Immunol 2019; 20:482-492. [PMID: 30833793 DOI: 10.1038/s41590-019-0325-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 01/18/2019] [Indexed: 01/17/2023]
Abstract
Gut-derived antigens trigger immunoglobulin A (IgA) immune responses that are initiated by cognate B cells in Peyer's patches (PPs). These cells colonize the subepithelial domes (SEDs) of the PPs and subsequently infiltrate pre-existing germinal centers (GCs). Here we defined the pre-GC events and the micro-anatomical site at which affinity-based B cell selection occurred in PPs. Using whole-organ imaging, we showed that the affinity of the B cell antigen receptor (BCR) regulated the infiltration of antigen-specific B cells into GCs but not clonal competition in the SED. Follicular helper-like T cells resided in the SED and promoted its B cell colonization, independently of the magnitude of BCR affinity. Imaging and immunoglobulin sequencing indicated that selective clonal expansion ensued during infiltration into GCs. Thus, in contrast to the events in draining lymph nodes and spleen, in PPs, T cells promoted mainly the population expansion of B cells without clonal selection during pre-GC events. These findings have major implications for the design of oral vaccines.
Collapse
|
125
|
Lin S, Haque A, Raeman R, Guo L, He P, Denning TL, El-Rayes B, Moolenaar WH, Yun CC. Autotaxin determines colitis severity in mice and is secreted by B cells in the colon. FASEB J 2019; 33:3623-3635. [PMID: 30481488 PMCID: PMC6404565 DOI: 10.1096/fj.201801415rr] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022]
Abstract
Autotaxin (ATX or ENPP2) is a secreted lysophospholipase D that produces lysophosphatidic acid (LPA), a pleiotropic lipid mediator acting on specific GPCRs. ATX and LPA have been implicated in key (patho)physiologic processes, including embryonic development, lymphocyte homing, inflammation, and cancer progression. Using LPA receptor knockout mice, we previously uncovered a role for LPA signaling in promoting colitis and colorectal cancer. Here, we examined the role of ATX in experimental colitis through inducible deletion of Enpp2 in adult mice. ATX expression was increased upon induction of colitis, whereas ATX deletion reduced the severity of inflammation in both acute and chronic colitis, accompanied by transient weight loss. ATX expression in lymphocytes was strongly reduced in Rag1-/- and μMT mice, suggesting B cells as a major ATX-producing source, which was validated by immunofluorescence and biochemical analyses. ATX secretion by B cells from control, but not Enpp2 knockout, mice led to ERK activation in colorectal cancer cells and promoted T cell migration. We conclude that ATX deletion suppresses experimental colitis and that B cells are a major source of ATX in the colon. Our study suggests that pharmacological inhibition of ATX could be a therapeutic strategy in colitis.-Lin, S., Haque, A., Raeman, R., Guo, L., He, P., Denning, T. L., El-Rayes, B., Moolenaar, W. H., Yun, C. C. Autotaxin determines colitis severity in mice and is secreted by B cells in the colon.
Collapse
Affiliation(s)
- Songbai Lin
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA
- Division of Digestive Diseases, Emory University, Atlanta, Georgia, USA
| | - Abedul Haque
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA
- Division of Digestive Diseases, Emory University, Atlanta, Georgia, USA
| | - Reben Raeman
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Leilei Guo
- Division of Digestive Diseases, Emory University, Atlanta, Georgia, USA
| | - Peijian He
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA
- Division of Digestive Diseases, Emory University, Atlanta, Georgia, USA
| | - Timothy L. Denning
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA; and
| | - Wouter H. Moolenaar
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C. Chris Yun
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA
- Division of Digestive Diseases, Emory University, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA; and
| |
Collapse
|
126
|
Gut intraepithelial T cells calibrate metabolism and accelerate cardiovascular disease. Nature 2019; 566:115-119. [PMID: 30700910 PMCID: PMC6367023 DOI: 10.1038/s41586-018-0849-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 12/05/2018] [Indexed: 12/25/2022]
|
127
|
IgA targeting on the α-molecular recognition element (α-MoRE) of viral phosphoprotein inhibits measles virus replication by interrupting formation and function of P-N complex intracellularly. Antiviral Res 2019; 161:144-153. [DOI: 10.1016/j.antiviral.2018.11.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 12/21/2022]
|
128
|
Different flavors of IL-21 in regulation of intestinal IgA to commensals. Mucosal Immunol 2019; 12:36-38. [PMID: 30361536 PMCID: PMC6533621 DOI: 10.1038/s41385-018-0099-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/19/2018] [Accepted: 09/28/2018] [Indexed: 02/04/2023]
|
129
|
Defective IgA response to atypical intestinal commensals in IL-21 receptor deficiency reshapes immune cell homeostasis and mucosal immunity. Mucosal Immunol 2019; 12:85-96. [PMID: 30087442 PMCID: PMC6301133 DOI: 10.1038/s41385-018-0056-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/18/2018] [Accepted: 05/31/2018] [Indexed: 02/04/2023]
Abstract
Despite studies indicating the effects of IL-21 signaling in intestinal inflammation, its roles in intestinal homeostasis and infection are not yet clear. Here, we report potent effects of commensal microbiota on the phenotypic manifestations of IL-21 receptor deficiency. IL-21 is produced highly in the small intestine and appears to be critical for mounting an IgA response against atypical commensals such as segmented filamentous bacteria and Helicobacter, but not to the majority of commensals. In the presence of these atypical commensals, IL-21R-deficient mice exhibit reduced numbers of germinal center and IgA+ B cells and expression of activation-induced cytidine deaminase in Peyer's patches as well as a significant decrease in small intestine IgA+ plasmablasts and plasma cells, leading to higher bacterial burdens and subsequent expansion of Th17 and Treg cells. These microbiota-mediated secondary changes in turn enhance T cell responses to an oral antigen and strikingly dampen Citrobacter rodentium-induced immunopathology, demonstrating a complex interplay between IL-21-mediated mucosal immunity, microbiota, and pathogens.
Collapse
|
130
|
Pérez-López JA, Rojas-Hernández S, Campos-Rodríguez R, Arciniega-Martínez IM, Cruz-Hernández TR, Reséndiz-Albor AA, Drago-Serrano ME. Posterior Subdiaphragmatic Vagotomy Downmodulates the IgA Levels in the Small Intestine of BALB/c Mice. Neuroimmunomodulation 2019; 26:292-300. [PMID: 31918430 DOI: 10.1159/000505097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/28/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The posterior vagus nerve trunk innervates the entire small intestine, and elucidating its modulatory role in the IgA response was the aim of this study. METHODS Two groups of six male BALB/c mice underwent sham or posterior subdiaphragmatic vagotomy and were euthanized on the 14th postoperative day; then, the small intestines were dissected. The intestinal fluid was harvested for antibody analysis by ELISA, and cell suspensions from Peyer's patches and lamina propria were prepared for cytofluorometric analysis of plasma cells and T lymphocytes. The CD4+ T cells were labeled for the intracellular IgA-producing interleukins (ILs)-4, -5, -6, and -10; transforming growth factor (TGF)-β; and the inflammatory cytokines tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and IL-12. In the intestinal tissue samples, myeloperoxidase (MPO) visualization and the enzymatic activity were assessed by immunohistochemistry and ELISA, respectively. The data were analyzed by Student's t test, and the differences were considered significant at p < 0.05. RESULTS In the vagotomy group, the IgA levels and the CD4+ T cells labeled with mediators that promote IgA secretion, including IL-4 (only at lamina propria), TNF-α, and IFN-γ, were decreased, whereas the lamina propria IgA+ plasma cells and MPO presence/activity were increased; changes in the IgM levels, IgM+ plasma cells, and CD4+ T cells labeled with TGF-β, which have a role in class switch recombination, were not observed. CONCLUSION The downmodulating impact of vagotomy on IgA levels may result from defective IgA secretion without affecting class switch recombination, whereas vagotomy evoked a proinflammatory response regarding MPO. These findings may reflect the role of the vagus nerve on the control of the IgA response in the small intestine.
Collapse
Affiliation(s)
- José Alfredo Pérez-López
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Saúl Rojas-Hernández
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rafael Campos-Rodríguez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ivonne Maciel Arciniega-Martínez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Teresita Rocío Cruz-Hernández
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Aldo Arturo Reséndiz-Albor
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City, Mexico,
| |
Collapse
|
131
|
Suzuki H, Hosomi K, Nasu A, Kondoh M, Kunisawa J. Development of Adjuvant-Free Bivalent Food Poisoning Vaccine by Augmenting the Antigenicity of Clostridium perfringens Enterotoxin. Front Immunol 2018; 9:2320. [PMID: 30356722 PMCID: PMC6189403 DOI: 10.3389/fimmu.2018.02320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/18/2018] [Indexed: 12/28/2022] Open
Abstract
Clostridium perfringens enterotoxin (CPE) is a common cause of food poisoning and hyperkalemia-associated death. Previously, we reported that fusion of pneumococcal surface protein A (PspA) to C-terminal fragment of CPE (C-CPE) efficiently bound mucosal epithelium so that PspA-specific immune responses could be provoked. In this study, we found that fusion of C-CPE with PspA augmented the antigenicity of C-CPE itself. These findings allowed us to hypothesize that fusion of C-CPE and another food poisoning vaccine act as a bivalent food poisoning vaccine. Therefore, we constructed an adjuvant-free bivalent vaccine against CPE and cholera toxin (CT), which is a major food poisoning in developing country, by genetically fusing CT B subunit to C-CPE. Because of the low antigenicity of C-CPE, immunization of mice with C-CPE alone did not induce C-CPE-specific immune responses. However, immunization with our vaccine induced both C-CPE- and CT-specific neutralizing antibody. The underlying mechanism of the augmented antigenicity of C-CPE included the activation of T cells by CTB. Moreover, neutralizing antibodies lasted for at least 48 weeks and the quality of the antibody was dependent on the binding activity of CTB–C-CPE to its receptors. These findings suggest that our fusion protein is a potential platform for the development of an adjuvant-free bivalent vaccine against CPE and CT.
Collapse
Affiliation(s)
- Hidehiko Suzuki
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Ayaka Nasu
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Sciences, The University of Tokyo, Tokyo, Japan.,Department of Microbiology and Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan.,Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Suita, Japan
| |
Collapse
|
132
|
Cordeiro BF, Oliveira ER, da Silva SH, Savassi BM, Acurcio LB, Lemos L, Alves JDL, Carvalho Assis H, Vieira AT, Faria AMC, Ferreira E, Le Loir Y, Jan G, Goulart LR, Azevedo V, Carvalho RDDO, do Carmo FLR. Whey Protein Isolate-Supplemented Beverage, Fermented by Lactobacillus casei BL23 and Propionibacterium freudenreichii 138, in the Prevention of Mucositis in Mice. Front Microbiol 2018; 9:2035. [PMID: 30258413 PMCID: PMC6143704 DOI: 10.3389/fmicb.2018.02035] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022] Open
Abstract
Mucositis is a clinically important gastrointestinal inflammatory infirmity, generated by antineoplastic drugs cytotoxic effects. The inflammatory process caused by this disease frequently leads to derangements in the alimentary tract and great malaise for the patient. Novel strategies are necessary for its prevention or treatment, as currently available treatments of mucositis have several limitations in relieving its symptoms. In this context, several research groups have investigated the use of probiotic bacteria, and in particular dairy bacterial strains. Compelling evidences reveal that milk fermented by certain probiotic bacteria has the capacity to ameliorate intestinal inflammatory disorders. In addition, innovative probiotic delivery strategies, based on probiotics incorporation into protective matrices, such as whey proteins, were able to increase the therapeutic effect of probiotic strains by providing extra protection for bacteria against environmental stresses. Therefore, in this study, we evaluated the role of the whey protein isolate (WPI), when added to skim milk fermented by Lactobacillus casei BL23 (L. casei BL23) or by Propionibacterium freudenreichii CIRM-BIA138 (P. freudenreichii 138), as a protective matrix against in vitro stress challenges. In addition, we investigated the therapeutic effect of these fermented beverages in a murine model of mucositis induced by 5-Fluorouracil (5-FU). Our results demonstrated that milk supplementation with 30% (w/v) of WPI increases the survival rate of both strains when challenged with acid, bile salts, high temperature and cold storage stresses, compared to fermented skim milk without the addition of WPI. Moreover, treatment with the probiotic beverages prevented weight loss and intestinal damages in mice receiving 5-FU. We conclude that the presence of WPI maximizes the anti-inflammatory effects of L. casei BL23, but not for P. freudenreichii 138, suggesting that whey protein enhancement of probiotic activity might be strain-dependent.
Collapse
Affiliation(s)
- Bárbara F. Cordeiro
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Emiliano R. Oliveira
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Sara H. da Silva
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Bruna M. Savassi
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Leonardo B. Acurcio
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Luisa Lemos
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Juliana de L. Alves
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Helder Carvalho Assis
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Angélica T. Vieira
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Ana M. C. Faria
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Enio Ferreira
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | | | - Gwénaël Jan
- STLO, INRA, Agrocampus Ouest, Rennes, France
| | - Luiz R. Goulart
- Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | - Vasco Azevedo
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Rodrigo D. de O. Carvalho
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Fillipe L. R. do Carmo
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
- STLO, INRA, Agrocampus Ouest, Rennes, France
| |
Collapse
|
133
|
The molecular mechanism for activating IgA production by Pediococcus acidilactici K15 and the clinical impact in a randomized trial. Sci Rep 2018; 8:5065. [PMID: 29567956 PMCID: PMC5864838 DOI: 10.1038/s41598-018-23404-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/12/2018] [Indexed: 01/19/2023] Open
Abstract
IgA secretion at mucosal sites is important for host defence against pathogens as well as maintaining the symbiosis with microorganisms present in the small intestine that affect IgA production. In the present study, we tested the ability of 5 strains of lactic acid bacteria stimulating IgA production, being Pediococcus acidilactici K15 selected as the most effective on inducing this protective immunoglobulin. We found that this response was mainly induced via IL-10, as efficiently as IL-6, secreted by K15-stimulated dendritic cells. Furthermore, bacterial RNA was largely responsible for the induction of these cytokines; double-stranded RNA was a major causative molecule for IL-6 production whereas single-stranded RNA was critical factor for IL-10 production. In a randomized, double-blind, placebo-controlled clinical trial, ingestion of K15 significantly increased the secretory IgA (sIgA) concentration in saliva compared with the basal level observed before this intervention. These results indicate that functional lactic acid bacteria induce IL-6 and IL-10 production by dendritic cells, which contribute to upregulating the sIgA concentration at mucosal sites in humans.
Collapse
|