1
|
Khairnar V, Ngo VN. Editorial: Role of carcinoembryonic antigen-related cell adhesion molecules in pathogen responses, tumorigenicity, and immune modulation. Front Immunol 2024; 15:1388453. [PMID: 38504992 PMCID: PMC10949890 DOI: 10.3389/fimmu.2024.1388453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/21/2024] Open
Affiliation(s)
| | - Vu N. Ngo
- Department of Systems Biology, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| |
Collapse
|
2
|
Lee SH, Lee H, You HS, Sung HJ, Hyun SH. Metabolic pathway prediction of core microbiome based on enterotype and orotype. Front Cell Infect Microbiol 2023; 13:1173085. [PMID: 37424791 PMCID: PMC10325833 DOI: 10.3389/fcimb.2023.1173085] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Identification of key microbiome components has been suggested to help address the maintenance of oral and intestinal health in humans. The core microbiome is similar in all individuals, whereas the diverse microbiome varies across individuals, based on their unique lifestyles and phenotypic and genotypic determinants. In this study, we aimed to predict the metabolism of core microorganisms in the gut and oral environment based on enterotyping and orotyping. Materials and methods Gut and oral samples were collected from 83 Korean women aged 50 years or older. The extracted DNA was subjected to next-generation sequencing analysis of 16S rRNA hypervariable regions V3-V4. Results Gut bacteria were clustered into three enterotypes, while oral bacteria were clustered into three orotypes. Sixty-three of the core microbiome between the gut and oral population were correlated, and different metabolic pathways were predicted for each type. Eubacterium_g11, Actinomyces, Atopobium, and Enterococcus were significantly positively correlated between the gut and oral abundance. The four bacteria were classified as type 3 in orotype and type 2 in enterotype. Conclusion Overall, the study suggested that collapsing the human body's multidimensional microbiome into a few categories may help characterize the microbiomes better and address health issues more deeply.
Collapse
Affiliation(s)
- Song Hee Lee
- Department of Biomedical Laboratory Science, Graduate School, Eulji University, Uijeongbu, Republic of Korea
| | - Han Lee
- Department of Biomedical Laboratory Science, Graduate School, Eulji University, Uijeongbu, Republic of Korea
| | - Hee Sang You
- Laboratory of Gastrointestinal Mucosal Immunology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Ho-joong Sung
- Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam, Republic of Korea
| | - Sung Hee Hyun
- Department of Biomedical Laboratory Science, Graduate School, Eulji University, Uijeongbu, Republic of Korea
| |
Collapse
|
3
|
CEACAMS 1, 5, and 6 in disease and cancer: interactions with pathogens. Genes Cancer 2023; 14:12-29. [PMID: 36741860 PMCID: PMC9891707 DOI: 10.18632/genesandcancer.230] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
The CEA family comprises 18 genes and 11 pseudogenes located at chromosome 19q13.2 and is divided into two main groups: cell surface anchored CEA-related cell adhesion molecules (CEACAMs) and the secreted pregnancy-specific glycoproteins (PSGs). CEACAMs are highly glycosylated cell surface anchored, intracellular, and intercellular signaling molecules with diverse functions, from cell differentiation and transformation to modulating immune responses associated with infection, inflammation, and cancer. In this review, we explore current knowledge surrounding CEACAM1, CEACAM5, and CEACAM6, highlight their pathological significance in the areas of cancer biology, immunology, and inflammatory disease, and describe the utility of murine models in exploring questions related to these proteins.
Collapse
|
4
|
Zhang Z, La Placa D, Gugiu G, Thunen A, Le K, Shively JE. Reversal of obesity development in Ceacam1 -/- male mice by bone marrow transplantation or introduction of the human CEACAM1 gene. Obesity (Silver Spring) 2022; 30:1351-1356. [PMID: 35785480 PMCID: PMC9541698 DOI: 10.1002/oby.23457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/20/2022] [Accepted: 04/08/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Although Ceacam1-/- male mice become obese on normal chow, the effect of bone marrow transplantation or introduction of the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) gene has not been studied, to the knowledge of the authors. METHODS This study analyzed Ceacam1-/- mice on normal diet or high-fat diet (HFD), including effects of bone marrow transplantation or introduction of the CEACAM1 gene. RESULTS Male Ceacam1-/- mice on normal diet versus HFD for 24 weeks gained significantly more weight than controls, and Ceacam1-/- mice aged up to 2 years had a high frequency of liver cancer. Transplantation of wild-type bone marrow into Ceacam1-/- mice or introduction of the human CEACAM1 gene fully or partially reversed the obesity phenotype. Liver lipidomics on Ceacam1-/- versus wild-type controls on an HFD revealed a significant increase in diacyl glycerides. An increase in fatty acid transporter CD36 levels further suggests that loss of Ceacam1 leads to a major dysregulation of free fatty acid uptake. CONCLUSIONS CEACAM1 expression in both the liver and immune cells regulates obesity and lipid storage pathways in the liver. Bone marrow reconstitution of the immune system or introduction of the human CEACAM1 gene can fully or partially reverse the phenotype.
Collapse
Affiliation(s)
- Zhifang Zhang
- Department of Immunology & TheranosticsArthur Riggs Diabetes and Metabolism Research Institute, City of Hope Cancer CenterDuarteCaliforniaUSA
| | - Deirdre La Placa
- Department of Immunology & TheranosticsArthur Riggs Diabetes and Metabolism Research Institute, City of Hope Cancer CenterDuarteCaliforniaUSA
| | - Gabriel Gugiu
- Department of Immunology & TheranosticsArthur Riggs Diabetes and Metabolism Research Institute, City of Hope Cancer CenterDuarteCaliforniaUSA
| | - Alyssa Thunen
- Department of Immunology & TheranosticsArthur Riggs Diabetes and Metabolism Research Institute, City of Hope Cancer CenterDuarteCaliforniaUSA
- Irell and Manella Graduate School of Biological Sciences, City of Hope Cancer CenterDuarteCaliforniaUSA
| | - Keith Le
- Department of Immunology & TheranosticsArthur Riggs Diabetes and Metabolism Research Institute, City of Hope Cancer CenterDuarteCaliforniaUSA
| | - John E. Shively
- Department of Immunology & TheranosticsArthur Riggs Diabetes and Metabolism Research Institute, City of Hope Cancer CenterDuarteCaliforniaUSA
| |
Collapse
|
5
|
Heydarian M, Rühl E, Rawal R, Kozjak-Pavlovic V. Tissue Models for Neisseria gonorrhoeae Research—From 2D to 3D. Front Cell Infect Microbiol 2022; 12:840122. [PMID: 35223556 PMCID: PMC8873371 DOI: 10.3389/fcimb.2022.840122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 12/02/2022] Open
Abstract
Neisseria gonorrhoeae is a human-specific pathogen that causes gonorrhea, the second most common sexually transmitted infection worldwide. Disease progression, drug discovery, and basic host-pathogen interactions are studied using different approaches, which rely on models ranging from 2D cell culture to complex 3D tissues and animals. In this review, we discuss the models used in N. gonorrhoeae research. We address both in vivo (animal) and in vitro cell culture models, discussing the pros and cons of each and outlining the recent advancements in the field of three-dimensional tissue models. From simple 2D monoculture to complex advanced 3D tissue models, we provide an overview of the relevant methodology and its application. Finally, we discuss future directions in the exciting field of 3D tissue models and how they can be applied for studying the interaction of N. gonorrhoeae with host cells under conditions closely resembling those found at the native sites of infection.
Collapse
|
6
|
Exploring the Ability of Meningococcal Vaccines to Elicit Mucosal Immunity: Insights from Humans and Mice. Pathogens 2021; 10:pathogens10070906. [PMID: 34358056 PMCID: PMC8308890 DOI: 10.3390/pathogens10070906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 12/16/2022] Open
Abstract
Neisseria meningitidis causes a devastating invasive disease but is also a normal colonizer of the human nasopharynx. Due to the rapid progression of disease, the best tool to protect individuals against meningococcal infections is immunization. Clinical experience with polysaccharide conjugate vaccines has revealed that an ideal meningococcal vaccine must prevent both invasive disease and nasal colonization, which confers herd immunity. However, not all meningococcal vaccines are equal in their ability to prevent nasal colonization, for unknown reasons. Herein, we describe recent efforts to utilize humanized mouse models to understand the impact of different meningococcal vaccines on nasal colonization. These mice are susceptible to nasal colonization, and they become immune following live nasal infection or immunization with matched capsule-conjugate or protein-based vaccines, replicating findings from human work. We bring together insights regarding meningococcal colonization and immunity from clinical work with findings using humanized mouse models, providing new perspective into the different determinants of mucosal versus systemic immunity. Then, we use this as a framework to help focus future studies toward understanding key mechanistic aspects left unresolved, including the bacterial factors required for colonization and immune evasion, determinants of nasal mucosal protection, and characteristics of an ideal meningococcal vaccine.
Collapse
|
7
|
Heydarian M, Schweinlin M, Schwarz T, Rawal R, Walles H, Metzger M, Rudel T, Kozjak-Pavlovic V. Triple co-culture and perfusion bioreactor for studying the interaction between Neisseria gonorrhoeae and neutrophils: A novel 3D tissue model for bacterial infection and immunity. J Tissue Eng 2021; 12:2041731420988802. [PMID: 33796248 PMCID: PMC7970704 DOI: 10.1177/2041731420988802] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/31/2020] [Indexed: 01/13/2023] Open
Abstract
Gonorrhea, a sexually transmitted disease caused by the bacteria Neisseria gonorrhoeae, is characterized by a large number of neutrophils recruited to the site of infection. Therefore, proper modeling of the N. gonorrhoeae interaction with neutrophils is very important for investigating and understanding the mechanisms that gonococci use to evade the immune response. We have used a combination of a unique human 3D tissue model together with a dynamic culture system to study neutrophil transmigration to the site of N. gonorrhoeae infection. The triple co-culture model consisted of epithelial cells (T84 human colorectal carcinoma cells), human primary dermal fibroblasts, and human umbilical vein endothelial cells on a biological scaffold (SIS). After the infection of the tissue model with N. gonorrhoeae, we introduced primary human neutrophils to the endothelial side of the model using a perfusion-based bioreactor system. By this approach, we were able to demonstrate the activation and transmigration of neutrophils across the 3D tissue model and their recruitment to the site of infection. In summary, the triple co-culture model supplemented by neutrophils represents a promising tool for investigating N. gonorrhoeae and other bacterial infections and interactions with the innate immunity cells under conditions closely resembling the native tissue environment.
Collapse
Affiliation(s)
| | - Matthias Schweinlin
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Thomas Schwarz
- Translational Centre Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), Würzburg, Bayern, Germany
| | - Ravisha Rawal
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Heike Walles
- Research Center "Dynamic Systems: Systems Engineering" (CDS), Otto von-Guericke-University, Magdeburg, Sachsen-Anhalt, Germany
| | - Marco Metzger
- Translational Centre Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), Würzburg, Bayern, Germany
| | - Thomas Rudel
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
8
|
Rayes RF, Vourtzoumis P, Bou Rjeily M, Seth R, Bourdeau F, Giannias B, Berube J, Huang YH, Rousseau S, Camilleri-Broet S, Blumberg RS, Beauchemin N, Najmeh S, Cools-Lartigue J, Spicer JD, Ferri LE. Neutrophil Extracellular Trap-Associated CEACAM1 as a Putative Therapeutic Target to Prevent Metastatic Progression of Colon Carcinoma. THE JOURNAL OF IMMUNOLOGY 2020; 204:2285-2294. [PMID: 32169849 DOI: 10.4049/jimmunol.1900240] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 02/06/2020] [Indexed: 12/12/2022]
Abstract
Neutrophils promote tumor growth and metastasis at multiple stages of cancer progression. One mechanism through which this occurs is via release of neutrophil extracellular traps (NETs). We have previously shown that NETs trap tumor cells in both the liver and the lung, increasing their adhesion and metastasis following postoperative complications. Multiple studies have since shown that NETs play a role in tumor progression and metastasis. NETs are composed of nuclear DNA-derived web-like structures decorated with neutrophil-derived proteins. However, it is unknown which, if any, of these NET-affiliated proteins is responsible for inducing the metastatic phenotype. In this study, we identify the NET-associated carcinoembryonic Ag cell adhesion molecule 1 (CEACAM1) as an essential element for this interaction. Indeed, blocking CEACAM1 on NETs, or knocking it out in a murine model, leads to a significant decrease in colon carcinoma cell adhesion, migration and metastasis. Thus, this work identifies NET-associated CEACAM1 as a putative therapeutic target to prevent the metastatic progression of colon carcinoma.
Collapse
Affiliation(s)
- Roni F Rayes
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Phil Vourtzoumis
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Marianne Bou Rjeily
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Rashmi Seth
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - France Bourdeau
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Betty Giannias
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Julie Berube
- Meakins-Christie Laboratories, Department of Medicine, McGill University and the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Yu-Hwa Huang
- Department of Medicine, Harvard University, Boston, MA 02115
| | - Simon Rousseau
- Meakins-Christie Laboratories, Department of Medicine, McGill University and the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Sophie Camilleri-Broet
- Department of Pathology, McGill University Health Center, Montreal, Quebec H4A 3J1, Canada; and
| | | | - Nicole Beauchemin
- Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Sara Najmeh
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Jonathan Cools-Lartigue
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Jonathan D Spicer
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Lorenzo E Ferri
- Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Center, Montreal, Quebec H4A 3J1, Canada;
| |
Collapse
|
9
|
The HopQ-CEACAM Interaction Controls CagA Translocation, Phosphorylation, and Phagocytosis of Helicobacter pylori in Neutrophils. mBio 2020; 11:mBio.03256-19. [PMID: 32019805 PMCID: PMC7002351 DOI: 10.1128/mbio.03256-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori is highly adapted to humans and evades host immunity to allow its lifelong colonization. However, the H. pylori mouse model is artificial for H. pylori, and few adapted strains allow gastric colonization. Here, we show that human or CEACAM-humanized, but not mouse neutrophils are manipulated by the H. pylori HopQ-CEACAM interaction. Human CEACAMs are responsible for CagA phosphorylation, activation, and processing in neutrophils, whereas CagA translocation and tyrosine phosphorylation in DCs and macrophages is independent of the HopQ-CEACAM interaction. H. pylori affects the secretion of distinct chemokines in CEACAM-humanized neutrophils and macrophages. Most importantly, human CEACAMs on neutrophils enhance binding, oxidative burst, and phagocytosis of H. pylori and enhance bacterial survival in the phagosome. The H. pylori-CEACAM interaction modulates PMNs to reduce the H. pylori CagA translocation efficiency in vivo and to fine-tune the expression of CEACAM receptors on neutrophils to limit translocation of CagA and gastric pathology. The cag type IV secretion system (cag-T4SS) of Helicobacter pylori exploits specific cellular carcinoembryonic antigen-related cell adhesion molecules (CEACAMs), such as CEACAM1, -3, -5, and -6, as cellular receptors for CagA translocation into human gastric epithelial cells. We studied the interaction of H. pylori with human CEACAM1, CEACAM3, and CEACAM6 receptors (hCEACAMs) expressed on myeloid cells from CEACAM-humanized mice. Human and CEACAM-humanized mouse polymorphonuclear neutrophils (PMNs) allowed a specific HopQ-dependent interaction strongly enhancing CagA translocation. Translocated CagA was tyrosine phosphorylated, which was not seen in wild-type (wt) murine neutrophils. In contrast, human or murine bone marrow-derived macrophages and dendritic cells (DCs) revealed a low hCEACAM expression and bacterial binding. CagA translocation and tyrosine-phosphorylation was low and independent of the HopQ-CEACAM interaction. Neutrophils, but not macrophages or DCs, from CEACAM-humanized mice, significantly upregulated the proinflammatory chemokine MIP-1α. However, macrophages showed a significantly reduced amount of CXCL1 (KC) and CCL2 (MCP-1) secretion in CEACAM-humanized versus wt cells. Thus, H. pylori, via the HopQ-CEACAM interaction, controls the production and secretion of chemokines differently in PMNs, macrophages, and DCs. We further show that upon H. pylori contact the oxidative burst of neutrophils and phagocytosis of H. pylori was strongly enhanced, but hCEACAM3/6 expression on neutrophils allowed the extended survival of H. pylori within neutrophils in a HopQ-dependent manner. Finally, we demonstrate that during a chronic mouse infection, H. pylori is able to systemically downregulate hCEACAM1 and hCEACAM6 receptor expression on neutrophils, probably to limit CagA translocation efficiency and most likely gastric pathology.
Collapse
|
10
|
Klaile E, Müller MM, Zubiría-Barrera C, Brehme S, Klassert TE, Stock M, Durotin A, Nguyen TD, Feer S, Singer BB, Zipfel PF, Rudolphi S, Jacobsen ID, Slevogt H. Unaltered Fungal Burden and Lethality in Human CEACAM1-Transgenic Mice During Candida albicans Dissemination and Systemic Infection. Front Microbiol 2019; 10:2703. [PMID: 31849868 PMCID: PMC6889641 DOI: 10.3389/fmicb.2019.02703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/07/2019] [Indexed: 12/29/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1, CD66a) is a receptor for Candida albicans. It is crucial for the immune response of intestinal epithelial cells to this opportunistic pathogen. Moreover, CEACAM1 is of importance for the mucosal colonization by different bacterial pathogens. We therefore studied the influence of the human CEACAM1 receptor in human CEACAM1-transgenic mice on the C. albicans colonization and infection utilizing a colonization/dissemination and a systemic infection mouse model. Our results showed no alterations in the host response between the transgenic mice and the wild-type littermates to the C. albicans infections. Both mouse strains showed comparable C. albicans colonization and mycobiota, similar fungal burdens in various organs, and a similar survival in the systemic infection model. Interestingly, some of the mice treated with anti-bacterial antibiotics (to prepare them for C. albicans colonization via oral infection) also showed a strong reduction in endogenous fungi instead of the normally observed increase in fungal numbers. This was independent of the expression of human CEACAM1. In the systemic infection model, the human CEACAM1 expression was differentially regulated in the kidneys and livers of Candida-infected transgenic mice. Notably, in the kidneys, a total loss of the largest human CEACAM1 isoform was observed. However, the overwhelming immune response induced in the systemic infection model likely covered any CEACAM1-specific effects in the transgenic animals. In vitro studies using bone marrow-derived neutrophils from both mouse strains also revealed no differences in their reaction to C. albicans. In conclusion, in contrast to bacterial pathogens interacting with CEACAM1 on different mucosal surfaces, the human CEACAM1-transgenic mice did not reveal a role of human CEACAM1 in the in vivo candidiasis models used here. Further studies and different approaches will be needed to reveal a putative role of CEACAM1 in the host response to C. albicans.
Collapse
Affiliation(s)
- Esther Klaile
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Mario M Müller
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Cristina Zubiría-Barrera
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Saskia Brehme
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Tilman E Klassert
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Magdalena Stock
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Adrian Durotin
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Tien D Nguyen
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Sabina Feer
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Bernhard B Singer
- Medical Faculty, Institute of Anatomy, University Duisburg-Essen, Essen, Germany
| | - Peter F Zipfel
- Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Sven Rudolphi
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.,Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Center for Sepsis Control and Care (CSCC), University Hospital Jena, Jena, Germany
| | - Ilse D Jacobsen
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.,Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Center for Sepsis Control and Care (CSCC), University Hospital Jena, Jena, Germany
| | - Hortense Slevogt
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| |
Collapse
|
11
|
Muenstermann M, Strobel L, Klos A, Wetsel RA, Woodruff TM, Köhl J, Johswich KO. Distinct roles of the anaphylatoxin receptors C3aR, C5aR1 and C5aR2 in experimental meningococcal infections. Virulence 2019; 10:677-694. [PMID: 31274379 PMCID: PMC6650196 DOI: 10.1080/21505594.2019.1640035] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 04/08/2019] [Accepted: 06/28/2019] [Indexed: 02/08/2023] Open
Abstract
The complement system is pivotal in the defense against invasive disease caused by Neisseria meningitidis (Nme, meningococcus), particularly via the membrane attack complex. Complement activation liberates the anaphylatoxins C3a and C5a, which activate three distinct G-protein coupled receptors, C3aR, C5aR1 and C5aR2 (anaphylatoxin receptors, ATRs). We recently discovered that C5aR1 exacerbates the course of the disease, revealing a downside of complement in Nme sepsis. Here, we compared the roles of all three ATRs during mouse nasal colonization, intraperitoneal infection and human whole blood infection with Nme. Deficiency of complement or ATRs did not alter nasal colonization, but significantly affected invasive disease: Compared to WT mice, the disease was aggravated in C3ar-/- mice, whereas C5ar1-/- and C5ar2-/- mice showed increased resistance to meningococcal sepsis. Surprisingly, deletion of either of the ATRs resulted in lower cytokine/chemokine responses, irrespective of the different susceptibilities of the mice. This was similar in ex vivo human whole blood infection using ATR inhibitors. Neutrophil responses to Nme were reduced in C5ar1-/- mouse blood. Upon stimulation with C5a plus Nme, mouse macrophages displayed reduced phosphorylation of ERK1/2, when C5aR1 or C5aR2 were ablated or inhibited, suggesting that both C5a-receptors prime an initial macrophage response to Nme. Finally, in vivo blockade of C5aR1 alone (PMX205) or along with C5aR2 (A8Δ71-73) resulted in ameliorated disease, whereas neither antagonizing C3aR (SB290157) nor its activation with a "super-agonist" peptide (WWGKKYRASKLGLAR) demonstrated a benefit. Thus, C5aR1 and C5aR2 augment disease pathology and are interesting targets for treatment, whereas C3aR is protective in experimental meningococcal sepsis.
Collapse
Affiliation(s)
- Marcel Muenstermann
- Institut für Hygiene und Mikrobiologie, Universität Würzburg, Würzburg, Germany
| | - Lea Strobel
- Institut für Hygiene und Mikrobiologie, Universität Würzburg, Würzburg, Germany
| | - Andreas Klos
- Institut für Medizinische Mikrobiologie und Krankenhaushygiene, Medizinische Hochschule Hannover, Hannover, Germany
| | - Rick A. Wetsel
- Institute of Molecular Medicine Center for Immunology and Autoimmune Diseases, The University of Texas Health Science Center, Houston, TX, USA
| | - Trent M. Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kay O. Johswich
- Institut für Hygiene und Mikrobiologie, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
Zhang Z, La Placa D, Nguyen T, Kujawski M, Le K, Li L, Shively JE. CEACAM1 regulates the IL-6 mediated fever response to LPS through the RP105 receptor in murine monocytes. BMC Immunol 2019; 20:7. [PMID: 30674283 PMCID: PMC6345024 DOI: 10.1186/s12865-019-0287-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background Systemic inflammation and the fever response to pathogens are coordinately regulated by IL-6 and IL-1β. We previously showed that CEACAM1 regulates the LPS driven expression of IL-1β in murine neutrophils through its ITIM receptor. Results We now show that the prompt secretion of IL-6 in response to LPS is regulated by CEACAM1 expression on bone marrow monocytes. Ceacam1−/− mice over-produce IL-6 in response to an i.p. LPS challenge, resulting in prolonged surface temperature depression and overt diarrhea compared to their wild type counterparts. Intraperitoneal injection of a 64Cu-labeled LPS, PET imaging agent shows confined localization to the peritoneal cavity, and fluorescent labeled LPS is taken up by myeloid splenocytes and muscle endothelial cells. While bone marrow monocytes and their progenitors (CD11b+Ly6G−) express IL-6 in the early response (< 2 h) to LPS in vitro, these cells are not detected in the bone marrow after in vivo LPS treatment perhaps due to their rapid and complete mobilization to the periphery. Notably, tissue macrophages are not involved in the early IL-6 response to LPS. In contrast to human monocytes, TLR4 is not expressed on murine bone marrow monocytes. Instead, the alternative LPS receptor RP105 is expressed and recruits MD1, CD14, Src, VAV1 and β-actin in response to LPS. CEACAM1 negatively regulates RP105 signaling in monocytes by recruitment of SHP-1, resulting in the sequestration of pVAV1 and β-actin from RP105. Conclusion This novel pathway and regulation of IL-6 signaling by CEACAM1 defines a novel role for monocytes in the fever response of mice to LPS. Electronic supplementary material The online version of this article (10.1186/s12865-019-0287-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhifang Zhang
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA.
| | - Deirdre La Placa
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Tung Nguyen
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Maciej Kujawski
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Keith Le
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - Lin Li
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA
| | - John E Shively
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, 1500E Duarte Road, Duarte, CA91010, USA.
| |
Collapse
|
13
|
Specific Binding to Differentially Expressed Human Carcinoembryonic Antigen-Related Cell Adhesion Molecules Determines the Outcome of Neisseria gonorrhoeae Infections along the Female Reproductive Tract. Infect Immun 2018; 86:IAI.00092-18. [PMID: 29760215 DOI: 10.1128/iai.00092-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/05/2018] [Indexed: 02/07/2023] Open
Abstract
The gonococcal Opa proteins are an antigenically variable family of surface adhesins that bind human carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), CEACAM3, CEACAM5, and/or CEACAM6, cell surface glycoproteins that are differentially expressed on a broad spectrum of human cells and tissues. While they are presumed to be important for infection, the significance of various Opa-CEACAM-mediated cellular interactions in the context of the genital tract has remained unclear. Here, we observed that CEACAM1 and CEACAM5 are differentially expressed on epithelia lining the upper and lower portions of the human female genital tract, respectively. Using transgenic mouse lines expressing human CEACAMs in a manner that reflects this differential pattern, we considered the impact of Opa-CEACAM interactions during uncomplicated lower genital tract infections versus during pelvic inflammatory disease. Our results demonstrate that Opa-CEACAM5 binding on vaginal epithelia facilitates the long-term colonization of the lower genital tract, while Opa protein binding to CEACAM1 on uterine epithelia enhances gonococcal association and penetration into these tissues. While these Opa-dependent interactions with CEACAM-expressing epithelial surfaces promote infection, Opa binding by neutrophil-expressed CEACAMs counterbalances this by facilitating more effective gonococcal clearance. Furthermore, during uterine infections, CEACAM-dependent tissue invasion aggravates disease pathology by increasing the acute inflammatory response. Together, these findings demonstrate that the outcome of infection is determined by both the cell type-specific expression of human CEACAMs and the CEACAM specificity of the Opa variants expressed, which combine to determine the level of gonococcal association with the genital mucosa versus the extent of CEACAM-dependent inflammation and gonococcal clearance by neutrophils.
Collapse
|
14
|
CEACAM1 promotes CD8 + T cell responses and improves control of a chronic viral infection. Nat Commun 2018; 9:2561. [PMID: 29967450 PMCID: PMC6028648 DOI: 10.1038/s41467-018-04832-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 05/23/2018] [Indexed: 12/26/2022] Open
Abstract
Dysfunction of CD8+ T cells can lead to the development of chronic viral infection. Identifying mechanisms responsible for such T cell dysfunction is therefore of great importance to understand how to prevent persistent viral infection. Here we show using lymphocytic choriomeningitis virus (LCMV) infection that carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is fundamental for recruiting lymphocyte-specific protein kinase (Lck) into the T cell receptor complex to form an efficient immunological synapse. CEACAM1 is essential for activation of CD8+ T cells, and the absence of CEACAM1 on virus-specific CD8+ T cells limits the antiviral CD8+ T cell response. Treatment with anti-CEACAM1 antibody stabilizes Lck in the immunological synapse, prevents CD8+ T cell exhaustion, and improves control of virus infection in vivo. Treatment of human virus-specific CD8+ T cells with anti-CEACAM1 antibody similarly enhances their proliferation. We conclude that CEACAM1 is an important regulator of virus-specific CD8+ T cell functions in mice and humans and represents a promising therapeutic target for modulating CD8+ T cells. Chronic viral infections are frequently associated with the dysfunction of CD8+ T cells which includes loss of function and results in CD8+ T cell exhaustion. Here the authors show a role of CEACAM1 in promoting responsive CD8+ T cells in the context of a chronic lymphocytic choriomeningitis virus (LCMV) infection model.
Collapse
|
15
|
Abstract
The host-adapted human pathogen Neisseria gonorrhoeae is the causative agent of gonorrhoea. Consistent with its proposed evolution from an ancestral commensal bacterium, N. gonorrhoeae has retained features that are common in commensals, but it has also developed unique features that are crucial to its pathogenesis. The continued worldwide incidence of gonorrhoeal infection, coupled with the rising resistance to antimicrobials and the difficulties in controlling the disease in developing countries, highlights the need to better understand the molecular basis of N. gonorrhoeae infection. This knowledge will facilitate disease prevention, surveillance and control, improve diagnostics and may help to facilitate the development of effective vaccines or new therapeutics. In this Review, we discuss sex-related symptomatic gonorrhoeal disease and provide an overview of the bacterial factors that are important for the different stages of pathogenesis, including transmission, colonization and immune evasion, and we discuss the problem of antibiotic resistance.
Collapse
Affiliation(s)
- Sarah Jane Quillin
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - H Steven Seifert
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
16
|
Käser T, Renois F, Wilson HL, Cnudde T, Gerdts V, Dillon JAR, Jungersen G, Agerholm JS, Meurens F. Contribution of the swine model in the study of human sexually transmitted infections. INFECTION GENETICS AND EVOLUTION 2017; 66:346-360. [PMID: 29175001 DOI: 10.1016/j.meegid.2017.11.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/18/2017] [Accepted: 11/22/2017] [Indexed: 12/12/2022]
Abstract
The pig has garnered more and more interest as a model animal to study various conditions in humans. The growing success of the pig as an experimental animal model is explained by its similarities with humans in terms of anatomy, genetics, immunology, and physiology, by their manageable behavior and size, and by the general public acceptance of using pigs for experimental purposes. In addition, the immunological toolbox of pigs has grown substantially in the last decade. This development led to a boost in the use of pigs as a preclinical model for various human infections including sexually transmitted diseases (STIs) like Chlamydia trachomatis. In the current review, we discuss the use of animal models for biomedical research on the major human STIs. We summarize results obtained in the most common animal models and focus on the contributions of the pig model towards the understanding of pathogenesis and the host immune response. In addition, we present the main features of the porcine model that are particularly relevant for the study of pathogens affecting human female and male genital tracts. We also inform on the technological advancements in the porcine toolbox to facilitate new discoveries in this biologically important animal model. There is a continued need for improvements in animal modeling for biomedical research inclusive STI research. With all its advantages and the highly improved toolbox, the porcine model can play a crucial role in STI research and open the door to new exciting discoveries.
Collapse
Affiliation(s)
- Tobias Käser
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, 27607 Raleigh, NC, USA
| | - Fanny Renois
- LUNAM Université, Oniris, Laboratoire d'Étude des Résidus et Contaminants dans les Aliments (LABERCA), UMR INRA 1329, 44307 Nantes, France
| | - Heather L Wilson
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, S7N 5E3 Saskatoon, Saskatchewan, Canada
| | - Thomas Cnudde
- BIOMAP, Laboratoire Biomédicaments Anti-Parasitaires, ISP, UMR INRA 1282, Université Tours, 37380 Nouzilly, France
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, S7N 5E3 Saskatoon, Saskatchewan, Canada
| | - Jo-Anne R Dillon
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, S7N 5E3 Saskatoon, Saskatchewan, Canada; Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Canada
| | - Gregers Jungersen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Jørgen S Agerholm
- Section for Veterinary Reproduction and Obstetrics, Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
17
|
Buckwalter CM, Currie EG, Tsang RSW, Gray-Owen SD. Discordant Effects of Licensed Meningococcal Serogroup B Vaccination on Invasive Disease and Nasal Colonization in a Humanized Mouse Model. J Infect Dis 2017; 215:1590-1598. [PMID: 28368526 DOI: 10.1093/infdis/jix162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 03/23/2017] [Indexed: 11/12/2022] Open
Abstract
Background The multicomponent meningococcal serogroup B vaccine (4CMenB) is an outer membrane vesicle and recombinant protein-based vaccine licensed to protect against serogroup B meningococcal disease. It remains unknown whether this vaccine will prevent carriage or transmission, key aspects in long-term vaccine success and disease eradication. Methods Using a "humanized" transgenic mouse model of nasal colonization, we took a systematic approach to estimate the potential for carriage prevention against antigenically diverse Neisseria meningitidis strains and to compare this protection to an invasive meningococcal disease challenge model. Results The 4CMenB vaccine prevented morbidity and mortality after lethal invasive doses of all meningococcal strains tested. Immunization effectively prevented carriage with only 1 of 4 single antigen-matched strains but reduced or prevented nasal colonization by all 4 isolates with multiple cross-reacting antigens. Each immunized mouse had substantial immunoglobulin G targeting the challenge strains, indicating that antibody correlates with protection against sepsis but not nasal carriage. Conclusions Immunization with the 4CMenB vaccine elicits a robust humoral response that correlates with protection against invasive challenge but not with prevention of asymptomatic colonization. This suggests that widespread use of this vaccine will reduce morbidity and mortality rates in immunized individuals, with the potential to contribute to herd protection against a subset of strains.
Collapse
Affiliation(s)
| | - Elissa G Currie
- Department of Molecular Genetics, University of Toronto, Ontario and
| | - Raymond S W Tsang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba
| | - Scott D Gray-Owen
- Department of Molecular Genetics, University of Toronto, Ontario and
| |
Collapse
|
18
|
Moreau MR, Massari P, Genco CA. The ironclad truth: how in vivo transcriptomics and in vitro mechanistic studies shape our understanding of Neisseria gonorrhoeae gene regulation during mucosal infection. Pathog Dis 2017; 75:3829888. [PMID: 28520925 PMCID: PMC5808646 DOI: 10.1093/femspd/ftx057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/16/2017] [Indexed: 11/12/2022] Open
Abstract
Neisseria gonorrhoeae is one of the most prevalent sexually transmitted infections worldwide. This obligate human pathogen has been extensively studied in vitro, where bacterial factors that are known to contribute to gonococcal disease and their regulation are relatively well defined. However, these in vitro experimental conditions only loosely replicate the host specific environment encountered by the bacteria in vivo. We recently reported on the complete gonococcal transcriptome expressed during natural human mucosal infection using RNA-seq analysis. Gene transcripts expressed in vivo (in vivo expressed factors) included genes encoding antibiotic resistance determinants, and a large number of hypothetical genes. A comparison of the gonococcal transcriptome expressed in vivo with the corresponding strain grown in vitro identified sets of genes regulated by infection, including those regulated by iron and the transcriptional regulatory protein Fur. We highlight here the role of Fur and gonococcal-specific regulatory processes important for infection and pathogenicity. We have determined that the genes controlled by Fur follow the same expression pattern in vivo as described previously in vitro, confirming Fur's regulatory role during infection. Collectively, these studies provide new insights into how bacterial fitness and pathogenicity are modulated during human mucosal infection.
Collapse
Affiliation(s)
- Matthew R. Moreau
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Paola Massari
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Caroline A. Genco
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
19
|
A native outer membrane vesicle vaccine confers protection against meningococcal colonization in human CEACAM1 transgenic mice. Vaccine 2015; 33:1317-1323. [PMID: 25662856 DOI: 10.1016/j.vaccine.2015.01.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/30/2014] [Accepted: 01/22/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND The effect of protein-based meningococcal vaccines on prevention of nasopharyngeal colonization has been difficult to investigate experimentally because a reliable animal colonization model did not exist. METHODS Human CEACAM1 transgenic mice, which can be colonized by meningococci, were immunized IP with one of two meningococcal native outer membrane vesicle (NOMV) vaccines prepared from mutants with attenuated endotoxin (lpxL1 knockout) and over-expressed sub-family B Factor H-binding proteins (FHbp). Animals were challenged intranasally two weeks after the third dose with wild-type strain H44/76, or were treated IP with anti-NOMV serum before and during the bacterial challenge. RESULTS The NOMV-1 vaccine, prepared from the serogroup B H44/76 mutant, elicited ∼40-fold higher serum bactericidal antibody titers against the wild-type H44/76 challenge strain than the NOMV-2 vaccine prepared from a heterologous serogroup W mutant strain with different PorA and FHbp amino acid sequence variants. Compared to aluminum hydroxide-immunized control mice, the efficacy for prevention of any H44/76 colonization was 93% (95% confidence interval, 52-99, P<0.0001) for the NOMV-1 vaccine, and 19% (-3-36, P=0.23) for NOMV-2. NOMV-2-vaccinated mice had a 5.6-fold decrease in geometric mean CFU of bacteria per animal in tracheal washes compared to control mice (P=0.007). The efficacy of passive administration of serum from NOMV-1-vaccinated mice to immunologically naïve mice against colonization was 44% (17-61; P=0.002). CONCLUSIONS Both NOMV vaccines protected against meningococcal colonization but there was greater protection by the NOMV-1 vaccine with antigens matched with the challenge strain. Meningococcal vaccines that target protein antigens have potential to decrease colonization.
Collapse
|
20
|
Takeyama A, Yoshikawa Y, Ikeo T, Morita S, Hieda Y. Expression patterns of CD66a and CD117 in the mouse submandibular gland. Acta Histochem 2015; 117:76-82. [PMID: 25498293 DOI: 10.1016/j.acthis.2014.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/09/2014] [Accepted: 11/10/2014] [Indexed: 02/05/2023]
Abstract
The epithelial tissue of the salivary gland consists of the acinar and ductal parts, the latter of which is further divided into the intercalated, striated and excretory duct segments and is the residential site for salivary stem/progenitor cells. In the present study, the expression patterns of two cell surface molecules, CD66a and CD117, were investigated in the adult mouse submandibular glands (SMG) by immunofluorescence microscopy. Combinations of the two molecules differentially marked several types of SMG epithelial cells, including acinar cells (CD66a-intense, CD117-negative), intercalated duct cells (CD66a-intense, CD117-positive), a subset of the striated and excretory duct cells (CD66a-weak, CD117-positive). Most of the CD117-positive ductal cells were negative for cytokeratin 5 and overlapped with the NKCC1-expressing cells. The CD117- and keratin 5-positive cells resided only in the excretory duct were suggested to correspond to the recently identified salivary stem cells. CD66a and CD117 may be useful markers to isolate several cell types consisting of SMG epithelium and to analyze their molecular and cellular nature. Our data also suggest that CD117-expressing epithelial cells of the gland include at least two distinct populations of the stem/progenitor cells.
Collapse
Affiliation(s)
- Akira Takeyama
- Department of Oral and Maxillofacial Surgery 1, Osaka Dental University, Osaka, Japan.
| | | | - Takashi Ikeo
- Department of Biochemistry, Osaka Dental University, Osaka, Japan
| | - Shosuke Morita
- Department of Oral and Maxillofacial Surgery 1, Osaka Dental University, Osaka, Japan
| | - Yohki Hieda
- Department of Biology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
21
|
Sterilizing immunity elicited by Neisseria meningitidis carriage shows broader protection than predicted by serum antibody cross-reactivity in CEACAM1-humanized mice. Infect Immun 2014; 83:354-63. [PMID: 25368118 DOI: 10.1128/iai.02495-14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neisseria meningitidis asymptomatically colonizes the human upper respiratory tract but is also the cause of meningitis and severe septicemia. Carriage or disease evokes an immune response against the infecting strain. Hitherto, we have known little about the breadth of immunity induced by natural carriage of a single strain or its implications for subsequent infectious challenge. In this study, we establish that transgenic mice expressing human CEACAM1 support nasal colonization by a variety of strains of different capsular types. Next, we nasally challenged these mice with either of the N. meningitidis strains H44/76 (serogroup B, ST-32) and 90/18311 (serogroup C, ST-11), while following the induction of strain-specific immunoglobulin. When these antisera were tested for reactivity with a diverse panel of N. meningitidis strains, very low levels of antibody were detected against all meningococcal strains, yet a mutually exclusive "fingerprint" of high-level cross-reactivity toward certain strains became apparent. To test the efficacy of these responses for protection against subsequent challenge, CEACAM1-humanized mice exposed to strain 90/18311 were then rechallenged with different N. meningitidis strains. As expected, the mice were immune to challenge with the same strain and with a closely related ST-11 strain, 38VI, while H44/76 (ST-32) could still colonize these animals. Notably, however, despite the paucity of detectable humoral response against strain 196/87 (ST-32), this strain was unable to colonize the 90/18311-exposed mice. Combined, our data suggest that current approaches may underestimate the actual breadth of mucosal protection gained through natural exposure to N. meningitidis strains.
Collapse
|
22
|
Global analysis of neutrophil responses to Neisseria gonorrhoeae reveals a self-propagating inflammatory program. PLoS Pathog 2014; 10:e1004341. [PMID: 25188454 PMCID: PMC4154863 DOI: 10.1371/journal.ppat.1004341] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/15/2014] [Indexed: 11/19/2022] Open
Abstract
An overwhelming neutrophil-driven response causes both acute symptoms and the lasting sequelae that result from infection with Neisseria gonorrhoeae. Neutrophils undergo an aggressive opsonin-independent response to N. gonorrhoeae, driven by the innate decoy receptor CEACAM3. CEACAM3 is exclusively expressed by human neutrophils, and drives a potent binding, phagocytic engulfment and oxidative killing of Opa-expressing bacteria. In this study, we sought to explore the contribution of neutrophils to the pathogenic inflammatory process that typifies gonorrhea. Genome-wide microarray and biochemical profiling of gonococcal-infected neutrophils revealed that CEACAM3 engagement triggers a Syk-, PKCδ- and Tak1-dependent signaling cascade that results in the activation of an NF-κB-dependent transcriptional response, with consequent production of pro-inflammatory cytokines. Using an in vivo model of N. gonorrhoeae infection, we show that human CEACAM-expressing neutrophils have heightened migration toward the site of the infection where they may be further activated upon Opa-dependent binding. Together, this study establishes that the role of CEACAM3 is not restricted to the direct opsonin-independent killing by neutrophils, since it also drives the vigorous inflammatory response that typifies gonorrhea. By carrying the potential to mobilize increasing numbers of neutrophils, CEACAM3 thereby represents the tipping point between protective and pathogenic outcomes of N. gonorrhoeae infection. Gonorrhea is a sexually transmitted infection caused by the bacteria Neisseria gonorrhoeae. These bacteria have re-emerged as a public health priority due to its acquisition of resistance to multiple antibiotics, leading to fears of untreatable infection. The symptoms of gonorrhea include an intense inflammatory response that may lead to pus discharged from the infected genital tract and scarring of the reproductive tract caused by neutrophils recruited to the site of infection. Past studies have detailed molecular interactions that lead to neutrophil binding and engulfment of N. gonorrhoeae, yet it remains unclear why N. gonorrhoeae elicits such a pathogenic inflammatory response. In this study, we reveal that N. gonorrhoeae binding to the human innate decoy receptor, CEACAM3, elicits a potent intracellular signaling cascade that leads to neutrophil expression of cytokines that actively recruit other neutrophils to the infected tissues. As they encounter the gonococci, the next wave of neutrophils becomes similarly activated, leading to the progressive expansion in phagocytic cell numbers until they overwhelm the infected tissues. While this process promotes a rapid response to a troubling pathogen early during infection, the unrestrained recruitment of neutrophils and their toxic antimicrobial arsenal also lead to the pathogenic consequences associated with gonorrhea.
Collapse
|
23
|
Tchoupa AK, Schuhmacher T, Hauck CR. Signaling by epithelial members of the CEACAM family - mucosal docking sites for pathogenic bacteria. Cell Commun Signal 2014; 12:27. [PMID: 24735478 PMCID: PMC4057559 DOI: 10.1186/1478-811x-12-27] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 03/24/2014] [Indexed: 11/22/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) comprise a group of immunoglobulin-related vertebrate glycoproteins. Several family members, including CEACAM1, CEA, and CEACAM6, are found on epithelial tissues throughout the human body. As they modulate diverse cellular functions, their signaling capacity is in the focus of current research. In this review we will summarize the knowledge about common signaling processes initiated by epithelial CEACAMs and suggest a model of signal transduction by CEACAM family members lacking significant cytoplasmic domains. As pathogenic and non-pathogenic bacteria exploit these receptors during mucosal colonization, we try to highlight the connection between CEACAMs, microbes, and cellular responses. Special emphasis in this context is placed on the functional interplay between CEACAMs and integrins that influences matrix adhesion of epithelial cells. The cooperation between these two receptor families provides an intriguing example of the fine tuning of cellular responses and their manipulation by specialized microorganisms.
Collapse
Affiliation(s)
| | | | - Christof R Hauck
- Lehrstuhl für Zellbiologie, Universität Konstanz, 78457 Konstanz, Germany.
| |
Collapse
|
24
|
Inhibition of the alternative pathway of nonhuman infant complement by porin B2 contributes to virulence of Neisseria meningitidis in the infant rat model. Infect Immun 2014; 82:2574-84. [PMID: 24686052 DOI: 10.1128/iai.01517-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neisseria meningitidis utilizes capsular polysaccharide, lipooligosaccharide (LOS) sialic acid, factor H binding protein (fHbp), and neisserial surface protein A (NspA) to regulate the alternative pathway (AP) of complement. Using meningococcal mutants that lacked all four of the above-mentioned molecules (quadruple mutants), we recently identified a role for PorB2 in attenuating the human AP; inhibition was mediated by human fH, a key downregulatory protein of the AP. Previous studies showed that fH downregulation of the AP via fHbp or NspA is specific for human fH. Here, we report that PorB2-expressing quadruple mutants also regulate the AP of baby rabbit and infant rat complement. Blocking a human fH binding region on PorB2 of the quadruple mutant of strain 4243 with a chimeric protein that comprised human fH domains 6 and 7 fused to murine IgG Fc enhanced AP-mediated baby rabbit C3 deposition, which provided evidence for an fH-dependent mechanism of nonhuman AP regulation by PorB2. Using isogenic mutants of strain H44/76 that differed only in their PorB molecules, we confirmed a role for PorB2 in resistance to killing by infant rat serum. The PorB2-expressing strain also caused higher levels of bacteremia in infant rats than its isogenic PorB3-expressing counterpart, thus providing a molecular basis for increased survival of PorB2 isolates in this model. These studies link PorB2 expression with infection of infant rats, which could inform the choice of meningococcal strains for use in animal models, and reveals, for the first time, that PorB2-expressing strains of N. meningitidis regulate the AP of baby rabbits and rats.
Collapse
|
25
|
Beauchemin N, Arabzadeh A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev 2013; 32:643-71. [DOI: 10.1007/s10555-013-9444-6] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Johswich KO, McCaw SE, Islam E, Sintsova A, Gu A, Shively JE, Gray-Owen SD. In vivo adaptation and persistence of Neisseria meningitidis within the nasopharyngeal mucosa. PLoS Pathog 2013; 9:e1003509. [PMID: 23935487 PMCID: PMC3723569 DOI: 10.1371/journal.ppat.1003509] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 06/04/2013] [Indexed: 12/13/2022] Open
Abstract
Neisseria meningitidis (Nme) asymptomatically colonizes the human nasopharynx, yet can initiate rapidly-progressing sepsis and meningitis in rare instances. Understanding the meningococcal lifestyle within the nasopharyngeal mucosa, a phase of infection that is prerequisite for disease, has been hampered by the lack of animal models. Herein, we compare mice expressing the four different human carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) that can bind the neisserial Opa protein adhesins, and find that expression of human CEACAM1 is necessary and sufficient to establish intranasal colonization. During infection, in vivo selection for phase variants expressing CEACAM1-specific Opa proteins occurs, allowing mucosal attachment and entry into the subepithelial space. Consistent with an essential role for Opa proteins in this process, Opa-deficient meningococci were unable to colonize the CEACAM1-humanized mice. While simple Opa-mediated attachment triggered an innate response regardless of meningococcal viability within the inoculum, persistence of viable Opa-expressing bacteria within the CEACAM1-humanized mice was required for a protective memory response to be achieved. Parenteral immunization with a capsule-based conjugate vaccine led to the accumulation of protective levels of Nme-specific IgG within the nasal mucus, yet the sterilizing immunity afforded by natural colonization was instead conferred by Nme-specific IgA without detectable IgG. Considered together, this study establishes that the availability of CEACAM1 helps define the exquisite host specificity of this human-restricted pathogen, displays a striking example of in vivo selection for the expression of desirable Opa variants, and provides a novel model in which to consider meningococcal infection and immunity within the nasopharyngeal mucosa. Neisseria meningitidis (Nme), a common cause of bacterial meningitis, are carried asymptomatically in the nasopharynx by a substantial proportion of healthy individuals. Their strict adaptation to the human as host has so far impeded the development of animal models to study the meningococcal lifestyle in vivo. While several human CEACAMs are recognized by the neisserial Opa protein adhesins, we show here that the expression of human CEACAM1 in transgenic mice is necessary and sufficient to allow nasal colonization by Nme. The dependence on human CEACAM1 is attributable to the Opa proteins, since intranasal infection with Opa-negative colonies of Nme selects for bacteria expressing Opa proteins, and genetically Opa-deficient meningococci are unable to colonize these animals. We use this new mouse model to examine how innate immune factors such as neutrophils and complement limit colonization. Furthermore, we compare how adaptive responses elicited by colonization and those generated by parenteral vaccination differentially confer sterilizing immunity. Together, this work provides the first evidence of the critical nature of Opa-CEACAM1 binding in vivo, demonstrates that this is a major determinant of the host restriction by Nme, and reveals a clear disparity between immune correlates of sterilizing immunity conferred by natural colonization versus parenteral immunization.
Collapse
Affiliation(s)
- Kay O Johswich
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
27
|
Miyao M, Ozeki M, Abiru H, Manabe S, Kotani H, Tsuruyama T, Tamaki K. Bile canalicular abnormalities in the early phase of a mouse model of sclerosing cholangitis. Dig Liver Dis 2013; 45:216-25. [PMID: 23107486 DOI: 10.1016/j.dld.2012.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 09/15/2012] [Accepted: 09/19/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND The bile canaliculus is the smallest and first biliary channel and is formed by two or three adjacent hepatocytes. Previous studies of chronic cholangiopathies such as primary sclerosing cholangitis have focused on the bile ductules. However, little is known about the pathological alterations in bile canaliculi in the early phase of cholangiopathies. AIM To characterize the bile canalicular morphology in the early phase of sclerosing cholangitis we used 3,5-diethoxycarbonyl-1,4-dihydrocollidine-induced mouse model of sclerosing cholangitis. METHODS Mice were fed a diet with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (0.1%). Serum biochemical, histological, immunohistochemical, and electron microscopic analyses were performed 1, 2, 4, and 7 days after feeding. RESULTS All experimental groups showed significantly increased serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase levels. From day 1, bile canalicular abnormalities such as dilatation and meandering and loss of microvilli were observed. After bile canalicular abnormalities had appeared, substantial infiltration of inflammatory cells was observed amongst the necrotic cells and periductal region. After these inflammatory changes, cholangiocytes proliferated in the portal area and formed ductular reactions. Finally, periductal fibrosis appeared. CONCLUSION This study provides novel evidence of the occurrence of bile canalicular abnormalities during the early phase of sclerosing cholangitis.
Collapse
Affiliation(s)
- Masashi Miyao
- Department of Forensic Medicine and Molecular Pathology, Kyoto University Graduate School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Shyam K Sharan KB, Sharan SK. Manipulating the Mouse Genome Using Recombineering. ADVANCES IN GENETICS 2013; 2. [PMID: 31404315 DOI: 10.4172/2169-0111.1000108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genetically engineered mouse models are indispensable for understanding the biological function of genes, understanding the genetic basis of human diseases and for preclinical testing of novel therapies. Generation of such mouse models has been possible because of our ability to manipulate the mouse genome. Recombineering is a highly efficient recombination-based method of genetic engineering that has revolutionized our ability to generate mouse models. Since recombineering technology is not dependent on the availability of restriction enzyme recognition sites, it allows us to modify the genome with great precision. It requires homology arms as short as 40 bases for recombination, which makes it relatively easy to generate targeting constructs to insert, change or delete either a single nucleotide or a DNA fragment several kb in size; insert selectable markers, reporter genes or add epitope tags to any gene of interest. In this review, we focus on the development of recombineering technology and its application in the generation of transgenic and knockout or knock-in mouse models. High throughput generation of gene targeting vectors, used to construct knockout alleles in mouse embryonic stem cells, is now feasible because of this technology. The challenge now is to use the "designer" mice to develop novel therapies to prevent, cure or effectively manage some the most debilitating human diseases.
Collapse
Affiliation(s)
| | - Shyam K Sharan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland 21702
| |
Collapse
|
29
|
Johswich KO, Zhou J, Law DKS, St. Michael F, McCaw SE, Jamieson FB, Cox AD, Tsang RSW, Gray-Owen SD. Invasive potential of nonencapsulated disease isolates of Neisseria meningitidis. Infect Immun 2012; 80:2346-53. [PMID: 22508859 PMCID: PMC3416473 DOI: 10.1128/iai.00293-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 04/03/2012] [Indexed: 01/07/2023] Open
Abstract
The capsule of Neisseria meningitidis is the major virulence factor that enables this bacterium to overcome host immunity elicited by complement and phagocytes, rendering it capable of surviving in blood. As such, nonencapsulated N. meningitidis isolates are generally considered nonpathogenic. Here, we consider the inherent virulence of two nonencapsulated N. meningitidis isolates obtained from our national surveillance of infected blood cultures in Canada. Capsule deficiency of both strains was confirmed by serology and PCR for the ctrA to ctrD genes and siaA to siaC genes, as well as siaD genes specific to serogroups B, C, Y, and W135. In both strains, the capsule synthesis genes were replaced by the capsule null locus, cnl-2. In accordance with a lack of capsule, both strains were fully susceptible to killing by both human and baby rabbit complement. However, in the presence of cytidine-5' monophospho-N-acetylneuraminic acid (CMP-NANA), allowing for lipooligosaccharide (LOS) sialylation, a significant increase of resistance to complement killing was observed. Mass spectrometry of purified LOS did not reveal any uncommon modifications that would explain their invasive phenotype. Finally, in a mouse intraperitoneal challenge model, these nonencapsulated isolates displayed enhanced virulence relative to an isogenic mutant of serogroup B strain MC58 lacking capsule (MC58ΔsiaD). Virulence of all nonencapsulated isolates tested was below that of encapsulated serogroup B strains MC58 and B16B6. However, whereas no mortality was observed with MC58ΔsiaD, 5/10 mice succumbed to infection with strain 2275 and 2/11 mice succumbed to strain 2274. Our results suggest the acquisition of a new virulence phenotype by these nonencapsulated strains.
Collapse
Affiliation(s)
- Kay O. Johswich
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jianwei Zhou
- Vaccine Preventable Bacterial Diseases, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Dennis K. S. Law
- Vaccine Preventable Bacterial Diseases, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Frank St. Michael
- Institute for Biological Sciences, National Research Council, Ottawa, Ontario, Canada
| | - Shannon E. McCaw
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Andrew D. Cox
- Institute for Biological Sciences, National Research Council, Ottawa, Ontario, Canada
| | - Raymond S. W. Tsang
- Vaccine Preventable Bacterial Diseases, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Scott D. Gray-Owen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Li G, Jiao H, Yan H, Wang J, Wang X, Ji M. Establishment of a human CEACAM1 transgenic mouse model for the study of gonococcal infections. J Microbiol Methods 2011; 87:350-4. [PMID: 21986029 DOI: 10.1016/j.mimet.2011.09.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 09/25/2011] [Accepted: 09/26/2011] [Indexed: 12/30/2022]
Abstract
Neisseria gonorrhoeae is the causative microorganism for the sexually transmitted disease (STD) gonorrhea and humans are its only natural host. An animal model would be a useful tool for gonorrhea research, therefore we developed the hCEACAM1 transgenic mice, using an eukaryotic expression vector, pCDPCAM1-GI. This construct was microinjected into the zygotes of C57BL/6 mice and 22 F0 generation transgenic mice were obtained. Four (lines 50, 53, 54, and 59) of the F0 generation were found to carry the transgene by PCR and sequence analysis, respectively. Western blotting and Fluorescence-Activated Cell Sorting Analysis demonstrated that hCEACAM1 was expressed on the cell membrane of various tissues in the line 53 transgenic mouse. To initiate the disease in the animal model, the F2 or F3 transgenic mice were inoculated with N. gonorrhoeae intravaginally. Compared with normal mice, N. gonorrhoeae can successfully infect and cause inflammation in the transgenic mice. These data suggested the feasibility of using hCEACAM1 transgenic mice as an animal model for gonococcal infections.
Collapse
Affiliation(s)
- Guocai Li
- Department of Pathogen Biology and Immunology, Yangzhou University School of Medicine, Yangzhou, China.
| | | | | | | | | | | |
Collapse
|
31
|
Trivedi K, Tang CM, Exley RM. Mechanisms of meningococcal colonisation. Trends Microbiol 2011; 19:456-63. [PMID: 21816616 DOI: 10.1016/j.tim.2011.06.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/16/2011] [Accepted: 06/28/2011] [Indexed: 01/05/2023]
Abstract
Despite advances against infectious diseases over the past century, Neisseria meningitidis remains a major causative agent of meningitis and septicaemia worldwide. Its adaptation for survival in the human nasopharynx makes the meningococcus a highly successful commensal bacterium. Recent progress has been made in understanding the mechanisms that enable neisserial colonisation, in terms of the role of type IV pili, the impact of other adhesins, biofilm formation, nutrient acquisition and resistance to host immune defences. Refinements in cell-based and in vivo models will lead to improved understanding of the colonisation process, and hopefully to more effective vaccines and therapeutic strategies.
Collapse
Affiliation(s)
- Kaushali Trivedi
- Centre for Molecular Microbiology and Infection, Faculty of Medicine, Flowers Building, Imperial College London, London SW7 2AZ, UK
| | | | | |
Collapse
|
32
|
Zhu W, Chen CJ, Thomas CE, Anderson JE, Jerse AE, Sparling PF. Vaccines for gonorrhea: can we rise to the challenge? Front Microbiol 2011; 2:124. [PMID: 21687431 PMCID: PMC3109613 DOI: 10.3389/fmicb.2011.00124] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 05/19/2011] [Indexed: 12/14/2022] Open
Abstract
Immune responses to the gonococcus after natural infection ordinarily result in little immunity to reinfection, due to antigenic variation of the gonococcus, and redirection or suppression of immune responses. Brinton and colleagues demonstrated that parenteral immunization of male human volunteers with a purified pilus vaccine gave partial protection against infection by the homologous strain. However, the vaccine failed in a clinical trial. Recent vaccine development efforts have focused on the female mouse model of genital gonococcal infection. Here we discuss the state of the field, including our unpublished data regarding efficacy in the mouse model of either viral replicon particle (VRP) vaccines, or outer membrane vesicle (OMV) vaccines. The OMV vaccines failed, despite excellent serum and mucosal antibody responses. Protection after a regimen consisting of a PorB-VRP prime plus recombinant PorB boost was correlated with apparent Th1, but not with antibody, responses. Protection probably was due to powerful adjuvant effects of the VRP vector. New tools including novel transgenic mice expressing human genes required for gonococcal infection should enable future research. Surrogates for immunity are needed. Increasing antimicrobial resistance trends among gonococci makes development of a vaccine more urgent.
Collapse
Affiliation(s)
- Weiyan Zhu
- Department of Medicine, University of North Carolina Chapel Hill, NC, USA
| | | | | | | | | | | |
Collapse
|
33
|
Johnson MB, Criss AK. Resistance of Neisseria gonorrhoeae to neutrophils. Front Microbiol 2011; 2:77. [PMID: 21747795 PMCID: PMC3128980 DOI: 10.3389/fmicb.2011.00077] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 03/31/2011] [Indexed: 01/04/2023] Open
Abstract
Infection with the human-specific bacterial pathogen Neisseria gonorrhoeae triggers a potent, local inflammatory response driven by polymorphonuclear leukocytes (neutrophils or PMNs). PMNs are terminally differentiated phagocytic cells that are a vital component of the host innate immune response and are the first responders to bacterial and fungal infections. PMNs possess a diverse arsenal of components to combat microorganisms, including the production of reactive oxygen species and release of degradative enzymes and antimicrobial peptides. Despite numerous PMNs at the site of gonococcal infection, N. gonorrhoeae can be cultured from the PMN-rich exudates of individuals with acute gonorrhea, indicating that some bacteria resist killing by neutrophils. The contribution of PMNs to gonorrheal pathogenesis has been modeled in vivo by human male urethral challenge and murine female genital inoculation and in vitro using isolated primary PMNs or PMN-derived cell lines. These systems reveal that some gonococci survive and replicate within PMNs and suggest that gonococci defend themselves against PMNs in two ways: they express virulence factors that defend against PMNs' oxidative and non-oxidative antimicrobial components, and they modulate the ability of PMNs to phagocytose gonococci and to release antimicrobial components. In this review, we will highlight the varied and complementary approaches used by N. gonorrhoeae to resist clearance by human PMNs, with an emphasis on gonococcal gene products that modulate bacterial-PMN interactions. Understanding how some gonococci survive exposure to PMNs will help guide future initiatives for combating gonorrheal disease.
Collapse
Affiliation(s)
| | - Alison K. Criss
- Department of Microbiology, University of VirginiaCharlottesville, VA, USA
| |
Collapse
|
34
|
Sadarangani M, Pollard AJ, Gray-Owen SD. Opa proteins and CEACAMs: pathways of immune engagement for pathogenic Neisseria. FEMS Microbiol Rev 2011; 35:498-514. [PMID: 21204865 DOI: 10.1111/j.1574-6976.2010.00260.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neisseria meningitidis and Neisseria gonorrhoeae are globally important pathogens, which in part owe their success to their ability to successfully evade human immune responses over long periods. The phase-variable opacity-associated (Opa) adhesin proteins are a major surface component of these organisms, and are responsible for bacterial adherence and entry into host cells and interactions with the immune system. Most immune interactions are mediated via binding to members of the carcinoembryonic antigen cell adhesion molecule (CEACAM) family. These Opa variants are able to bind to different receptors of the CEACAM family on epithelial cells, neutrophils, and T and B lymphocytes, influencing the innate and adaptive immune responses. Increased epithelial cell adhesion creates the potential for prolonged infection, invasion and dissemination. Furthermore, Opa proteins may inhibit T-lymphocyte activation and proliferation, B-cell antibody production, and innate inflammatory responses by infected epithelia, in addition to conferring increased resistance to antibody-dependent, complement-mediated killing. While vaccines containing Opa proteins could induce adhesion-blocking and bactericidal antibodies, the consequence of CEACAM binding by a candidate Opa-containing vaccine requires further investigation. This review summarizes current knowledge of the immunological consequences of the interaction between meningococcal and gonococcal Opa proteins and human CEACAMs, considering the implications for pathogenesis and vaccine development.
Collapse
Affiliation(s)
- Manish Sadarangani
- Oxford Vaccine Group, Department of Paediatrics, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford, UK.
| | | | | |
Collapse
|