51
|
Li W, Xu X, Wen H, Wang Z, Ding C, Liu X, Gao Y, Su H, Zhang J, Han Y, Xia Y, Wang X, Gu H, Yao X. Inverse Association Between the Skin and Oral Microbiota in Atopic Dermatitis. J Invest Dermatol 2019; 139:1779-1787.e12. [PMID: 30802424 DOI: 10.1016/j.jid.2019.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/21/2022]
Abstract
Previous studies have shown independently that the skin and gut microbiota are closely associated with atopic dermatitis (AD); however, the microbiota across different habitats of AD patients as an integrated community has not been characterized. In the present study, we comparatively analyzed the structure and function of the microbial communities in the skin, oral cavity, and gut of 172 AD patients and 120 healthy controls through 16S ribosomal RNA gene amplicon sequencing. The skin and oral cavity, but not the gut, of AD patients demonstrated differential reduction in the microbial diversity, and these were distinctly correlated with disease severity. Different degrees of shifts in the community structure were found among different habitats, and the lineage distance between the skin and oral microbiota of AD patients was closer than that observed in the controls. The different habitats of AD patients exhibited site-specific alterations at the genus level, and many oral-specific microbes of AD showed opposing directions of enrichment in the skin and oral cavity. Most interestingly, an inverse association in the functional pathways was found between the skin and oral microbiota of AD patients. Additionally, the alterations of the microbiota in different body sites of AD patients were differentially affected by age.
Collapse
Affiliation(s)
- Wei Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiaoqiang Xu
- Aimigene Institute, Shenzhen, People's Republic of China
| | - He Wen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Zhifeng Wang
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Chao Ding
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China; Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Xiaochun Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yingxia Gao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Huichun Su
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Jingxi Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yue Han
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yan Xia
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Xiaokai Wang
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Heng Gu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China.
| | - Xu Yao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China.
| |
Collapse
|
52
|
Cornejo Ulloa P, van der Veen MH, Krom BP. Review: modulation of the oral microbiome by the host to promote ecological balance. Odontology 2019; 107:437-448. [PMID: 30719639 PMCID: PMC6732124 DOI: 10.1007/s10266-019-00413-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023]
Abstract
The indivisible relationship between the human host and its oral microbiome has been shaped throughout the millennia, by facing various changes that have forced the adaptation of oral microorganisms to new environmental conditions. In this constant crosstalk between the human host and its microbiome, a bidirectional relationship has been established. The microorganisms provide the host with functions it cannot perform on its own and at the same time the host provides its microbes with a suitable environment for their growth and development. These host factors can positively affect the microbiome, promoting diversity and balance between different species, resulting in a state of symbiosis and absence of pathology. In contrast, other host factors can negatively influence the composition of the oral microbiome and drive the interaction towards a dysbiotic state, where the balance tilts towards a harmful relationship between the host and its microbiome. The aim of this review is to describe the role host factors play in cultivating and maintaining a healthy oral ecology and discuss mechanisms that can prevent its drift towards dysbiosis.
Collapse
Affiliation(s)
- Pilar Cornejo Ulloa
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, G. Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Monique H van der Veen
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, G. Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands.
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, G. Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands.
| |
Collapse
|
53
|
Bauer HK, Flesch D, Walenta S, Unger RE, Schwab R, Nezi-Cahn S, Hasenburg A, Heller M, Brenner W. Primary Mucosal Epithelial Cell Cultivation: A Reliable and Accelerated Isolation. Tissue Eng Part C Methods 2019; 25:82-92. [DOI: 10.1089/ten.tec.2018.0327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Heide-Katharina Bauer
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Daniela Flesch
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Urology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Walenta
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ronald E. Unger
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Roxana Schwab
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sandra Nezi-Cahn
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Annette Hasenburg
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Martin Heller
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Walburgis Brenner
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
54
|
Creighton RL, Woodrow KA. Microneedle-Mediated Vaccine Delivery to the Oral Mucosa. Adv Healthc Mater 2019; 8:e1801180. [PMID: 30537400 PMCID: PMC6476557 DOI: 10.1002/adhm.201801180] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/12/2018] [Indexed: 12/28/2022]
Abstract
The oral mucosa is a minimally invasive and immunologically rich site that is underutilized for vaccination due to physiological and immunological barriers. To develop effective oral mucosal vaccines, key questions regarding vaccine residence time, uptake, adjuvant formulation, dose, and delivery location must be answered. However, currently available dosage forms are insufficient to address all these questions. An ideal oral mucosal vaccine delivery system would improve both residence time and epithelial permeation while enabling efficient delivery of physicochemically diverse vaccine formulations. Microneedles have demonstrated these capabilities for dermal vaccine delivery. Additionally, microneedles enable precise control over delivery properties like depth, uniformity, and dosing, making them an ideal tool to study oral mucosal vaccination. Select studies have demonstrated the feasibility of microneedle-mediated oral mucosal vaccination, but they have only begun to explore the broad functionality of microneedles. This review describes the physiological and immunological challenges related to oral mucosal vaccine delivery and provides specific examples of how microneedles can be used to address these challenges. It summarizes and compares the few existing oral mucosal microneedle vaccine studies and offers a perspective for the future of the field.
Collapse
Affiliation(s)
- Rachel L Creighton
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
55
|
Haruna T, Kariya S, Fujiwara T, Yuta A, Higaki T, Zhao P, Ogawa Y, Kanai K, Hirata Y, Oka A, Nishizaki K, Okano M. Role of whole saliva in the efficacy of sublingual immunotherapy in seasonal allergic rhinitis. Allergol Int 2019; 68:82-89. [PMID: 30166059 DOI: 10.1016/j.alit.2018.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/08/2018] [Accepted: 07/24/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The development of methods to predict the clinical effectiveness of sublingual immunotherapy (SLIT) for allergic diseases is a crucial matter. We sought to determine whether whole saliva, which is the first body component that contacts allergen extracts during SLIT, is associated with the clinical effectiveness of SLIT in Japanese cedar pollinosis. METHODS Blood monocytes or monocytic THP-1 cells were cultured in the presence or absence of either whole saliva or pure saliva with or without treatments including filtration and blockade of TLR2 and/or TLR4 signaling. IL-10 levels in the supernatants were then measured. Whole saliva-induced IL-10 production by THP-1 cells was compared between asymptomatic and disease-onset patients during peak pollen dispersal after SLIT. RESULTS Both monocytes and THP-1 cells produced substantial amounts of IL-10 in response to whole saliva. IL-10 production was significantly reduced in response to pure saliva and 0.2 μm-filtered saliva. Simultaneous treatment with polymyxin B and TL2.1, a neutralizing antibody against TLR2, also reduced IL-10 production. IL-10 levels produced by THP-1 cells in response to whole saliva collected prior to SLIT were significantly higher in asymptomatic patients determined by symptom-medication scores than disease-onset patients following SLIT. Such differences were not seen in saliva collected 3 months after the initiation of SLIT or saliva collected during peak pollen dispersal. CONCLUSIONS Our results provide a basis for why the sublingual route is effective and preferable in allergen immunotherapy. Saliva-induced IL-10 levels produced by THP-1 cells may be a predictive marker for clinical remission after SLIT.
Collapse
Affiliation(s)
- Takenori Haruna
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shin Kariya
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tazuko Fujiwara
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Takaya Higaki
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Pengfei Zhao
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Kengo Kanai
- Department Otorhinolaryngology-Head & Neck Surgery, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Yuji Hirata
- Department Otorhinolaryngology-Head & Neck Surgery, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Aiko Oka
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazunori Nishizaki
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mitsuhiro Okano
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Department of Otorhinolaryngology, International University of Health and Welfare School of Medicine, Narita, Japan.
| |
Collapse
|
56
|
Naftulin JS, Penzi LR, Manatis-Lornell A, Yasuda MR, Porter ML, Saavedra A, Senna MM. Longstanding alopecia and nail dystrophy are associated with more severe overall chronic graft-versus-host disease in adults. Bone Marrow Transplant 2018; 54:469-472. [DOI: 10.1038/s41409-018-0309-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/30/2018] [Accepted: 08/05/2018] [Indexed: 11/09/2022]
|
57
|
Barreto de Albuquerque J, Silva Dos Santos D, Stein JV, de Meis J. Oral Versus Intragastric Inoculation: Similar Pathways of Trypanosoma cruzi Experimental Infection? From Target Tissues, Parasite Evasion, and Immune Response. Front Immunol 2018; 9:1734. [PMID: 30100907 PMCID: PMC6072848 DOI: 10.3389/fimmu.2018.01734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
Currently, oral infection is the most frequent transmission mechanism of Chagas disease in Brazil and others Latin American countries. This transmission pathway presents increased mortality rate in the first 2 weeks, which is higher than the calculated mortality after the biting of infected insect vectors. Thus, the oral route of Trypanosoma cruzi infection, and the consequences in the host must be taken into account when thinking on the mechanisms underlying the natural history of the disease. Distinct routes of parasite entry may differentially affect immune circuits, stimulating regional immune responses that impact on the overall profile of the host protective immunity. Experimental studies related to oral infection usually comprise inoculation in the mouth (oral infection, OI) or gavage (gastrointestinal infection, GI), being often considered as similar routes of infection. Hence, establishing a relationship between the inoculation site (OI or GI) with disease progression and the mounting of T. cruzi-specific regional immune responses is an important issue to be considered. Here, we provide a discussion on studies performed in OI and GI in experimental models of acute infections, including T. cruzi infection.
Collapse
Affiliation(s)
| | - Danielle Silva Dos Santos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| |
Collapse
|
58
|
Immune quiescence in the oral mucosa is maintained by a uniquely large population of highly activated Foxp3 + regulatory T cells. Mucosal Immunol 2018; 11:1092-1102. [PMID: 29743613 PMCID: PMC6035783 DOI: 10.1038/s41385-018-0027-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/05/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
Abstract
The oral mucosa is a critical barrier tissue that protects the oral cavity against invading pathogens and foreign antigens. Interestingly, inflammation in the oral cavity is rarely observed, indicating that overt immune activation in this site is actively suppressed. Whether Foxp3+ Treg cells are involved in controlling immunity of the oral mucosa, however, is not fully understood. Here, we show that the oral mucosa is highly enriched in Foxp3+ Treg cells, and that oral mucosa Treg cells are phenotypically distinct from those of LN or spleen, as they expressed copious amounts of the tissue-retention molecule CD103 and unusually high-levels of CTLA4. Acute depletion of Foxp3+ Treg cells had catastrophic effects, resulting in marked infiltration of activated effector T cells that were associated with autoimmunity and tissue destruction of the oral mucosa. Moreover, adoptive transfer of naive CD4 T cells revealed that the oral mucosa is highly ineffective in inducing Foxp3+ Treg cells in situ, so that it depends on recruitment and migration of exogenous Treg cells to populate this mucosal site. Collectively, these results demonstrate a previously unappreciated role and a distinct developmental pathway for Foxp3+ Treg cells in the oral mucosa, which are essential to control local tissue immunity.
Collapse
|
59
|
Rosace D, Gomez-Casado C, Fernandez P, Perez-Gordo M, Dominguez MDC, Vega A, Belver MT, Ramos T, Vega F, Marco G, de Pedro M, Sanchez L, Arnas MDLM, Santaolalla M, Saez MÁ, Benedé S, Fernandez-Rivas M, Blanco C, Alvarado MI, Escribese MM, Barber D. Profilin-mediated food-induced allergic reactions are associated with oral epithelial remodeling. J Allergy Clin Immunol 2018; 143:681-690.e1. [PMID: 29705246 DOI: 10.1016/j.jaci.2018.03.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 02/26/2018] [Accepted: 03/16/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND In areas of high exposure to grass pollen, allergic patients are frequently sensitized to profilin, and some experience severe profilin-mediated food-induced reactions. This specific population of patients is ideal to study the relationship between respiratory and food allergies. OBJECTIVE We sought to determine the role of oral mucosal epithelial barrier integrity in profilin-mediated allergic reactions. METHODS Thirty-eight patients with profilin allergy stratified into mild or severe according to their clinical history and response to a profilin challenge test and 6 nonallergic subjects were recruited. Oral mucosal biopsies were used for measurement of CD11c, CD3, CD4, tryptase, claudin-1, occludin, E-cadherin, and vascular endothelial growth factor A levels; Masson trichrome staining; and POSTN, IL33, TPSAB, TPSB, and CMA gene expression analysis by using quantitative RT-PCR. Blood samples were used for basophil activation tests. RESULTS Distinct features of the group with severe allergy included the following: (1) impaired epithelial integrity with reduced expression of claudin-1, occludin, and E-cadherin and decreased numbers of epithelial cells, which is indicative of acanthosis, higher collagen deposition, and angiogenesis; (2) inflammatory immune response in the mucosa, with an increased number of CD11c+ and CD4+ infiltrates and increased expression of the cytokine genes POSTN and IL33; and (3) a 10-fold increased sensitivity of basophils to profilin. CONCLUSIONS Patients with profilin allergy present with significant damage to the oral mucosal epithelial barrier, which might allow profilin penetration into the oral mucosa and induction of local inflammation. Additionally, severely allergic patients presented with increased sensitivity of effector cells.
Collapse
Affiliation(s)
- Domenico Rosace
- Instituto de Medicina Molecular Aplicada, Grupo Hospital de Madrid, Universidad San Pablo-CEU, Madrid, Spain
| | - Cristina Gomez-Casado
- Instituto de Medicina Molecular Aplicada, Grupo Hospital de Madrid, Universidad San Pablo-CEU, Madrid, Spain
| | - Paloma Fernandez
- Instituto de Medicina Molecular Aplicada, Grupo Hospital de Madrid, Universidad San Pablo-CEU, Madrid, Spain
| | - Marina Perez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, Madrid, Spain
| | | | - Angel Vega
- Hospital Virgen del Puerto, Plasencia, Cáceres, Spain
| | - María Teresa Belver
- Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Tania Ramos
- Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Francisco Vega
- Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | | | | | | | | | | | | | - Sara Benedé
- Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Carlos Blanco
- Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | | | - María M Escribese
- Instituto de Medicina Molecular Aplicada, Grupo Hospital de Madrid, Universidad San Pablo-CEU, Madrid, Spain; Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, Madrid, Spain.
| | - Domingo Barber
- Instituto de Medicina Molecular Aplicada, Grupo Hospital de Madrid, Universidad San Pablo-CEU, Madrid, Spain
| |
Collapse
|
60
|
Sanchiz A, Cuadrado C, Dieguez MC, Ballesteros I, Rodríguez J, Crespo JF, de las Cuevas N, Rueda J, Linacero R, Cabanillas B, Novak N. Thermal processing effects on the IgE-reactivity of cashew and pistachio. Food Chem 2018; 245:595-602. [DOI: 10.1016/j.foodchem.2017.10.132] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 10/18/2022]
|
61
|
Affiliation(s)
- Ahmed S. Sultan
- Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, Maryland, United States of America
| | - Eric F. Kong
- Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, Maryland, United States of America
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
- Graduate Program in Life Sciences, Molecular Microbiology and Immunology Program, University of Maryland, Baltimore, Maryland, United States of America
| | - Alexandra M. Rizk
- Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, Maryland, United States of America
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, Maryland, United States of America
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
62
|
Fábián T, Gótai L, Beck A, Fábián G, Fejérdy P. The Role of Molecular Chaperones (Hspas/Hsp70S) in Oral Health and Oral Inflammatory Diseases: A Review. EUR J INFLAMM 2017. [DOI: 10.1177/1721727x0900700201] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Heat shock proteins of the 70kDa family (HSPAs/HSP70s) are major molecular chaperones and cytokines of most cells and microbes, extracellular and interstitial fluids, blood, synovial fluids and secretory body fluids like saliva. The induction of human HSPAs plays an important role at cellular level under most stress conditions; whereas microbial HSPAs improve microbial tolerance to environmental changes, and improve virulence and resistance against antimicrobial peptides. Extracellular HSPAs reveal cytoprotective properties and are involved in numerous physiological and pathological events, including modulation of cytokine release and immunity. Accordingly, HSPAs play a role in the maintenance of pulpal health, and the repair of injured dental hard tissues. HSPAs also play a role in stress adaptation of periodontal tissues, and in the maintenance of periodontal and mucosal health including defense against microbes, prevention of mucosal allergic reactions, and facilitation of healing of ulcers and wounds. Despite their advantageous effects maintaining health of several oral tissues, HSPAs are likely to play a role in the disadvantageous amplification of pulpal inflammatory response to bacteria, and in the formation of several periapical inflammatory lesions. HSPAs may also induce gingivitis under certain conditions, and play a role in the progression of periodontal bone defects. HSPAs may also play a role in atopic-type allergic reactions, autoimmune disorders, and haptenation in certain cases. Based on the above data, it can be assumed that HSPAs play an important role in oral defense under healthy conditions; however, their role is somewhat “Janus-faced” under pathological conditions.
Collapse
Affiliation(s)
- T.K. Fábián
- Semmelweis University Budapest, Faculty of Dentistry, Clinic of Prosthetic Dentistry, Budapest
| | - L. Gótai
- Semmelweis University Budapest, Faculty of Dentistry, Clinic of Prosthetic Dentistry, Budapest
| | - A. Beck
- Semmelweis University Budapest, Faculty of Dentistry, Clinic of Prosthetic Dentistry, Budapest
| | - G. Fábián
- Semmelweis University Budapest, Faculty of Dentistry, Clinic of Pediatric Dentistry and Orthodontics, Budapest, Hungary, EU
| | - P. Fejérdy
- Semmelweis University Budapest, Faculty of Dentistry, Clinic of Prosthetic Dentistry, Budapest
| |
Collapse
|
63
|
Shen S, Yang J, Carvajal RD. Mucosal melanoma: epidemiology, biology, management and the role of immunotherapy. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1399122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Sherry Shen
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Jessica Yang
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Richard D. Carvajal
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
64
|
Bittner-Eddy PD, Fischer LA, Tu AA, Allman DA, Costalonga M. Discriminating between Interstitial and Circulating Leukocytes in Tissues of the Murine Oral Mucosa Avoiding Nasal-Associated Lymphoid Tissue Contamination. Front Immunol 2017; 8:1398. [PMID: 29163479 PMCID: PMC5666297 DOI: 10.3389/fimmu.2017.01398] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022] Open
Abstract
Periodontitis is a chronic inflammatory response to a microbial biofilm that destroys bone and soft tissues supporting the teeth. Murine models of periodontitis based on Porphyromonas gingivalis (Pg) colonization have shown that extravasation of leukocytes into oral tissue is critical to driving alveolar bone destruction. Identifying interstitial leukocytes is key to understanding the immunopathogenesis of periodontitis. Here, we describe a robust flow cytometry assay based on intravenous FITC-conjugated anti-mouse CD45 mAb that distinguishes interstitial leukocytes in the oral mucosa of mice from those circulating within the vasculature or in post-dissection contaminating blood. Unaccounted circulating leukocytes skewed the relative frequency of B cells and granulocytes and inflated the numbers of all leukocyte cell types. We also describe a dissection technique that avoids contamination of oral mucosal tissues with nasal-associated lymphoid tissues (NALT), a B cell rich organ that can inflate leukocyte numbers at least 10-fold and skew the assessment of interstitial CD4 T cell phenotypes. Unlike circulating CD4 T cells, interstitial CD4 T cells were almost exclusively antigen-experienced cells (CD44hi). We report for the first time the presence of antigen-experienced Pg-specific CD4 T cells in NALT following oral feeding of mice with Pg. This new combined flow cytometry and dissection approach allows identification of leukocytes infiltrating the connective tissues of the murine oral mucosa and avoids confounding analyses of leukocytes not recruited to inflamed oral mucosal tissues in disease conditions like periodontitis, candidiasis, or sialadenitis.
Collapse
Affiliation(s)
- Peter D Bittner-Eddy
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Lori A Fischer
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Andy A Tu
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Daniel A Allman
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Massimo Costalonga
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
65
|
Kosten IJ, van de Ven R, Thon M, Gibbs S, de Gruijl TD. Comparative phenotypic and functional analysis of migratory dendritic cell subsets from human oral mucosa and skin. PLoS One 2017; 12:e0180333. [PMID: 28704477 PMCID: PMC5509153 DOI: 10.1371/journal.pone.0180333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 06/14/2017] [Indexed: 12/13/2022] Open
Abstract
Antigen exposure to oral mucosa is generally thought to lead to immune tolerance induction. However, very little is known about the subset composition and function of dendritic cells (DC) migrating from human oral mucosa. Here we show that migratory DC from healthy human gingival explants consist of the same phenotypic subsets in the same frequency distribution as DC migrating from human skin. The gingival CD1a+ Langerhans cell and interstitial DC subsets lacked CXCR4 expression in contrast to their cutaneous counterparts, pointing to different migration mechanisms, consistent with previous observations in constructed skin and gingival equivalents. Remarkably, without any exogenous conditioning, gingival explants released higher levels of inflammatory cytokines than human skin explants, resulting in higher DC migration rates and a superior ability of migrated DC to prime allogeneic T cells and to induce type-1 effector T cell differentiation. From these observations we conclude that rather than an intrinsic ability to induce T cell tolerance, DC migrating from oral mucosa may have a propensity to induce effector T cell immunity and maintain a high state of alert against possible pathogenic intruders in the steady state. These findings may have implications for oral immunization strategies.
Collapse
Affiliation(s)
| | - Rieneke van de Ven
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Maria Thon
- Department of Dermatology, VU University Medical Center, Amsterdam, the Netherlands
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam, the Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
66
|
Gurumurthy S, Iyer G, Srinivasan B, Agarwal S, Angayarkanni N. Ocular surface cytokine profile in chronic Stevens-Johnson syndrome and its response to mucous membrane grafting for lid margin keratinisation. Br J Ophthalmol 2017; 102:169-176. [PMID: 28689166 DOI: 10.1136/bjophthalmol-2017-310373] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/04/2017] [Accepted: 05/01/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND To study the tear cytokine and the conjunctival and oral mucosal marker profile in chronic ocular Stevens-Johnson syndrome (SJS) and their alteration following mucous membrane grafting (MMG) for lid margin keratinisation (LMK). METHODS In a 1-year prospective study, SJS cases (n=25) and age-matched/sex-matched healthy controls (n=25) were recruited. Tear specimen (Schirmer's strip), conjunctival and oral mucosal imprints were collected from controls and SJS cases pre-MMG and post-MMG (at first follow-up, n=17). Tear cytokines were profiled using 27-bioplex array. Transforming growth factor-beta (TGF-β)-mediated extracellular matrix changes in conjunctival and oral mucosal cells were analysed by gene expression studies. 30 RESULTS: Tear cytokine profiling of chronic SJS cases at pre-MMG stage revealed significant upregulation of cytokines granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-8, IL-1β, monocyte chemoattractant protein-1, IL-15, IL-2, IL-17A and basic fibroblast growth factor (bFGF) with downregulation of IP-10 (interferon gamma-induced protein 10), tumour necrosis factor-α, interferon-γ, IL-10, vascular endothelial growth factor, regulated upon activation normal T-cell expressed and secreted (RANTES), IL-7, IL-12p70 and IL-13, with maximal increase in GM-CSF and maximal downregulation of IP-10, respectively. Of these, IL-2, IL-15, bFGF and IL-17A showed significant correlation with disease severity, pre-MMG. Conjunctival cells pre-MMG showed increase in TGF-β1, TGF-βRII, connective tissue growth factor and collagen-III gene expression by 10, 67, 173 and 184 folds, respectively, which dropped to 1.3, 11, 13.5 and 19 folds correspondingly, post-MMG. However, their expressions in oral mucosa were negligible. CONCLUSION A proinflammatory, profibrotic, antiapoptotic ocular surface milieu characterises chronic ocular SJS. IP-10, an antifibrotic cytokine was noted to be maximally downregulated, unlike in other forms of chronic dry eye disease. The alterations in the ocular surface are seen to reverse largely with MMG for LMK.
Collapse
Affiliation(s)
- Srividya Gurumurthy
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO Block, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Geetha Iyer
- C.J. Shah Cornea Services, Dr G Sitalakshmi Memorial Clinic for Ocular Disorders, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Bhaskar Srinivasan
- C.J. Shah Cornea Services, Dr G Sitalakshmi Memorial Clinic for Ocular Disorders, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Shweta Agarwal
- C.J. Shah Cornea Services, Dr G Sitalakshmi Memorial Clinic for Ocular Disorders, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Narayanasamy Angayarkanni
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO Block, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| |
Collapse
|
67
|
Bankvall M, Östberg AK, Jontell M, Wold A, Östman S. The engagement of oral-associated lymphoid tissues during oral versus gastric antigen administration. Immunology 2017; 149:98-110. [PMID: 27288650 DOI: 10.1111/imm.12633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 05/31/2016] [Accepted: 06/03/2016] [Indexed: 12/12/2022] Open
Abstract
The role of oral-associated lymphoid tissues during induction of oral tolerance still remains elusive. Therefore, the aim was to compare T-cell activation and induction of tolerance to ovalbumin (OVA) presented through either of two routes; deposited into the oral cavity, or the stomach, thereby bypassing the oral cavity. OVA was administered by the oral or gastric route to BALB/c mice that had received OVA-specific DO11.10+ CD4(+) T cells, stained with CellTrace(™) Violet dye, through intravenous injection. Proliferating OVA-specific T cells were detected in the nose-associated lymphoid tissues (NALT) and the cervical, mesenteric and peripheral lymph nodes at different time-points following OVA exposure. OVA-specific T-cell proliferation was initially observed in the NALT 1 hr after oral, but not gastric, administration. However, at day 1, proliferation at this site was also detected after gastric administration and profound proliferation was observed at all sites by day 4. For the oral route the degree of proliferation observed was lower in the peripheral lymph nodes by day 4 compared with the other sites. These results demonstrate a similar activation pattern achieved by the two routes. However, the NALT distinguishes itself as a site of rapid T-cell activation towards fed antigens irrespective of feeding regimen. To evaluate induction of tolerance a semi-effective OVA dose was used, to detect differences in the degree of tolerance achieved. This was performed in a model of OVA-induced airway hypersensitivity. No differences in tolerance induction were observed between the two administration routes.
Collapse
Affiliation(s)
- Maria Bankvall
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Anna-Karin Östberg
- Department of Oral Microbiology and Immunology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Mats Jontell
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Agnes Wold
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Sofia Östman
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
68
|
Park JY, Chung H, Choi Y, Park JH. Phenotype and Tissue Residency of Lymphocytes in the Murine Oral Mucosa. Front Immunol 2017; 8:250. [PMID: 28337201 PMCID: PMC5340784 DOI: 10.3389/fimmu.2017.00250] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/20/2017] [Indexed: 12/15/2022] Open
Abstract
The oral mucosa is a critical barrier tissue that harbors a series of distinct immune cell subsets. Immune surveillance in the oral mucosa is important for both local and systemic immunity because the oral cavity is a heavily utilized route of pathogen entry and also serves as site of pathogen propagation. Nonetheless, composition and phenotype of the lymphocyte pool in the oral mucosa have remained poorly characterized. Utilizing a newly established protocol for mucosal immune cell isolation, here, we report that the oral mucosa features a unique cellular composition of immune cells, which differed not only from secondary lymphoid organs but also from mucosal tissues in the gut and lung. We observed profound accumulation of CD11b+Ly6Clo monocytes in the oral mucosa that were maintained independently of T- and B-lymphocytes. Unlike the gut mucosa, the oral mucosa neither contained CD8αα T cells nor was it enriched for CD103+CD69+ tissue-resident memory CD8 T cells. In fact, a major fraction of T cells circulated and trafficked through the mucosa as revealed by treatment with the S1P1 receptor antagonist, FTY720, a potent inhibitor of lymphocyte migration. Collectively, these results provide a comprehensive picture of immune cells in the oral mucosa as an active site of lymphocyte recruitment and surveillance.
Collapse
Affiliation(s)
- Joo-Young Park
- Experimental Immunology Branch, National Cancer Institute, NIH , Bethesda, MD , USA
| | - Hyunsoo Chung
- Experimental Immunology Branch, National Cancer Institute, NIH , Bethesda, MD , USA
| | - Youngnim Choi
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University , Seoul , South Korea
| | - Jung-Hyun Park
- Experimental Immunology Branch, National Cancer Institute, NIH , Bethesda, MD , USA
| |
Collapse
|
69
|
Heron SE, Elahi S. HIV Infection and Compromised Mucosal Immunity: Oral Manifestations and Systemic Inflammation. Front Immunol 2017; 8:241. [PMID: 28326084 PMCID: PMC5339276 DOI: 10.3389/fimmu.2017.00241] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/20/2017] [Indexed: 12/26/2022] Open
Abstract
Mucosal surfaces account for the vast majority of HIV transmission. In adults, HIV transmission occurs mainly by vaginal and rectal routes but rarely via oral route. By contrast, pediatric HIV infections could be as the result of oral route by breastfeeding. As such mucosal surfaces play a crucial role in HIV acquisition, and spread of the virus depends on its ability to cross a mucosal barrier. HIV selectively infects, depletes, and/or dysregulates multiple arms of the human immune system particularly at the mucosal sites and causes substantial irreversible damage to the mucosal barriers. This leads to microbial products translocation and subsequently hyper-immune activation. Although introduction of antiretroviral therapy (ART) has led to significant reduction in morbidity and mortality of HIV-infected patients, viral replication persists. As a result, antigen presence and immune activation are linked to “inflammaging” that attributes to a pro-inflammatory environment and the accelerated aging process in HIV patients. HIV infection is also associated with the prevalence of oral mucosal infections and dysregulation of oral microbiota, both of which may compromise the oral mucosal immunity of HIV-infected individuals. In addition, impaired oral immunity in HIV infection may predispose the patients to periodontal diseases that are associated with systemic inflammation and increased risk of cardiovascular diseases. The purpose of this review is to examine existing evidence regarding the role of innate and cellular components of the oral cavity in HIV infection and how HIV infection may drive systemic hyper-immune activation in these patients. We will also discuss current knowledge on HIV oral transmission, HIV immunosenescence in relation to the oral mucosal alterations during the course of HIV infection and periodontal disease. Finally, we discuss oral manifestations associated with HIV infection and how HIV infection and ART influence the oral microbiome. Therefore, unraveling how HIV compromises the integrity of the oral mucosal tissues and innate immune components of the oral cavity and its association with induction of chronic inflammation are critical for the development of effective preventive interventions and therapeutic strategies.
Collapse
Affiliation(s)
- Samantha E Heron
- Faculty of Medicine and Dentistry, Department of Dentistry, University of Alberta , Edmonton, AB , Canada
| | - Shokrollah Elahi
- Faculty of Medicine and Dentistry, Department of Dentistry, University of Alberta, Edmonton, AB, Canada; Faculty of Medicine and Dentistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
70
|
Heron SE, Elahi S. HIV Infection and Compromised Mucosal Immunity: Oral Manifestations and Systemic Inflammation. Front Immunol 2017; 8:241. [PMID: 28326084 DOI: 10.3389/fimmu.2017.00241doi|] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/20/2017] [Indexed: 05/25/2023] Open
Abstract
Mucosal surfaces account for the vast majority of HIV transmission. In adults, HIV transmission occurs mainly by vaginal and rectal routes but rarely via oral route. By contrast, pediatric HIV infections could be as the result of oral route by breastfeeding. As such mucosal surfaces play a crucial role in HIV acquisition, and spread of the virus depends on its ability to cross a mucosal barrier. HIV selectively infects, depletes, and/or dysregulates multiple arms of the human immune system particularly at the mucosal sites and causes substantial irreversible damage to the mucosal barriers. This leads to microbial products translocation and subsequently hyper-immune activation. Although introduction of antiretroviral therapy (ART) has led to significant reduction in morbidity and mortality of HIV-infected patients, viral replication persists. As a result, antigen presence and immune activation are linked to "inflammaging" that attributes to a pro-inflammatory environment and the accelerated aging process in HIV patients. HIV infection is also associated with the prevalence of oral mucosal infections and dysregulation of oral microbiota, both of which may compromise the oral mucosal immunity of HIV-infected individuals. In addition, impaired oral immunity in HIV infection may predispose the patients to periodontal diseases that are associated with systemic inflammation and increased risk of cardiovascular diseases. The purpose of this review is to examine existing evidence regarding the role of innate and cellular components of the oral cavity in HIV infection and how HIV infection may drive systemic hyper-immune activation in these patients. We will also discuss current knowledge on HIV oral transmission, HIV immunosenescence in relation to the oral mucosal alterations during the course of HIV infection and periodontal disease. Finally, we discuss oral manifestations associated with HIV infection and how HIV infection and ART influence the oral microbiome. Therefore, unraveling how HIV compromises the integrity of the oral mucosal tissues and innate immune components of the oral cavity and its association with induction of chronic inflammation are critical for the development of effective preventive interventions and therapeutic strategies.
Collapse
Affiliation(s)
- Samantha E Heron
- Faculty of Medicine and Dentistry, Department of Dentistry, University of Alberta , Edmonton, AB , Canada
| | - Shokrollah Elahi
- Faculty of Medicine and Dentistry, Department of Dentistry, University of Alberta, Edmonton, AB, Canada; Faculty of Medicine and Dentistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
71
|
Shima K, Koya T, Tsukioka K, Sakagami T, Hasegawa T, Fukano C, Ohashi-Doi K, Watanabe S, Suzuki E, Kikuchi T. Effects of sublingual immunotherapy in a murine asthma model sensitized by intranasal administration of house dust mite extracts. Allergol Int 2017; 66:89-96. [PMID: 27397923 DOI: 10.1016/j.alit.2016.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/07/2016] [Accepted: 05/21/2016] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Sublingual immunotherapy (SLIT) has received attention as a method for allergen immunotherapy. However, the mechanism of SLIT has not yet been fully investigated. Therefore, we evaluated the effects of SLIT in a murine asthma model, sensitized by intranasal administration of house dust mite (HDM) extracts. METHODS Female BALB/c mice were intranasally exposed to HDM for either 3 or 5 weeks (5 consecutive days per week). Mice were administered either low-dose (0.5 mg/day) or high-dose (5 mg/day) sublingual HDM extracts for 2 weeks, followed by an additional week of intranasal exposure. Airway hyperresponsiveness (AHR), bronchoalveolar lavage fluid (BALF) cell count, cytokine levels in the BALF and lymph node cell culture supernatants, and allergen-specific antibodies were measured. Lung histology was also investigated. RESULTS In mice sensitized for 5 weeks, high-dose SLIT ameliorated AHR, airway eosinophilia and goblet cell metaplasia. In mice sensitized for 3 weeks, even low dose SLIT ameliorated AHR and airway eosinophilia. Th2 cytokine levels in culture supernatants of submandibular lymph node cells in high-dose SLIT mice decreased, whereas IL-10 levels increased. Total IgA in BALF increased in mice sensitized for 3 or 5 weeks, and high-dose SLIT also increased allergen-specific IgG2a in mice sensitized for 5 weeks. CONCLUSIONS These data suggest that earlier induction of SLIT in HDM-sensitized mice provides superior suppression of AHR and goblet cell metaplasia. The modulation of allergen specific IgG2a and local IgA might play a role in the amelioration of AHR and airway inflammation.
Collapse
|
72
|
Jonsdottir S, Svansson V, Stefansdottir SB, Mäntylä E, Marti E, Torsteinsdottir S. Oral administration of transgenic barley expressing a Culicoides
allergen induces specific antibody response. Equine Vet J 2016; 49:512-518. [DOI: 10.1111/evj.12655] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/04/2016] [Indexed: 01/22/2023]
Affiliation(s)
- S. Jonsdottir
- Institute for Experimental Pathology; Biomedical Center; University of Iceland; Keldur Iceland
| | - V. Svansson
- Institute for Experimental Pathology; Biomedical Center; University of Iceland; Keldur Iceland
| | - S. B. Stefansdottir
- Institute for Experimental Pathology; Biomedical Center; University of Iceland; Keldur Iceland
| | - E. Mäntylä
- Faculty of Pharmacy; University of Iceland; Reykjavik Iceland
- ORF Genetics Ltd; Kopavogur Iceland
| | - E. Marti
- Department of Clinical Research and Veterinary Public Health; Vetsuisse Faculty; University of Berne; Berne Switzerland
| | - S. Torsteinsdottir
- Institute for Experimental Pathology; Biomedical Center; University of Iceland; Keldur Iceland
| |
Collapse
|
73
|
Aissa S, Ben Jazia R, Ayachi J, Ben Salem C, Hayouni A, Abdelghani A, Ben Saad H, Boussarsar M. Critical appraisal of the clinical utility of sublingual immunotherapy in allergy. Contemp Clin Trials Commun 2016; 4:1-8. [PMID: 29736465 PMCID: PMC5935903 DOI: 10.1016/j.conctc.2016.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 11/30/2022] Open
Abstract
Since it was introduced by Noon in 1911, allergen-specific immunotherapy or desensitization has been widely prescribed in the management of allergic diseases. Aimed at the etiology, it represents the only effective treatment for allergy. The basic mechanisms of immunotherapy are becoming better understood and allow us to improve this technique in the future. The sublingual immunotherapy as an alternative to subcutaneous route has been widely studied. Several clinical trials confirmed that sublingual immunotherapy is efficient in reducing allergic respiratory symptoms. The sublingual immunotherapy reduces the risk of developing serious side effects due to desensitization. We performed a literature review in order to remind the mechanisms of action and to demonstrate efficacy and tolerability of the sublingual immunotherapy in the treatment of allergic rhinoconjunctivitis and asthma and its impact on the quality of life.
Collapse
Affiliation(s)
- S. Aissa
- Pulmonology Department, Farhat Hached University Hospital, Sousse, 4000, Tunisia
| | - R. Ben Jazia
- Pulmonology Department, Farhat Hached University Hospital, Sousse, 4000, Tunisia
| | - J. Ayachi
- Medical Intensive Care Unit, Farhat Hached University Hospital, Sousse, Tunisia
| | - C. Ben Salem
- Department of Clinical Pharmacology, Faculty of Medicine of Sousse, Tunisia
| | - A. Hayouni
- Pulmonology Department, Farhat Hached University Hospital, Sousse, 4000, Tunisia
| | - A. Abdelghani
- Pulmonology Department, Farhat Hached University Hospital, Sousse, 4000, Tunisia
| | - H. Ben Saad
- Laboratory of Physiology, Farhat Hached University Hospital, Sousse, Tunisia
| | - M. Boussarsar
- Medical Intensive Care Unit, Farhat Hached University Hospital, Sousse, Tunisia
| |
Collapse
|
74
|
Würtzen PA, Gupta S, Brand S, Andersen PS. Grass pollen immunotherapy: where are we now. Immunotherapy 2016; 8:399-411. [PMID: 26973122 DOI: 10.2217/imt.16.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
During allergen immunotherapy (AIT), the allergic patient is exposed to the disease-inducing antigens (allergens) in order to induce clinical and immunological tolerance and obtain disease modification. Large trials of grass AIT with highly standardized subcutaneous and sublingual tablet vaccines have been conducted to document the clinical effect. Induction of blocking antibodies as well as changes in the balance between T-cell phenotypes, including induction of regulatory T-cell subtypes, have been demonstrated for both treatment types. These observations increase the understanding of the immunological mechanism behind the clinical effect and may make it possible to use the immunological changes as biomarkers of clinical effect. The current review describes the recent mechanistic findings for subcutaneous immunotherapy and sublingual immunotherapy/tablet treatment and discusses how the observed immunological changes translate into a scientific foundation for the observed clinical effects of grass pollen immunotherapy and lead to new treatment strategies for grass AIT.
Collapse
Affiliation(s)
- Peter A Würtzen
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| | - Shashank Gupta
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| | - Stephanie Brand
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| | - Peter S Andersen
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| |
Collapse
|
75
|
Tortajada-Girbés M, Mesa Del Castillo M, Larramona H, Lucas JM, Álvaro M, Tabar AI, Jerez MJ, Martínez-Cañavate A. Evidence in immunotherapy for paediatric respiratory allergy: Advances and recommendations. Allergol Immunopathol (Madr) 2016; 44 Suppl 1:1-32. [PMID: 27776895 DOI: 10.1016/j.aller.2016.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/05/2016] [Indexed: 01/26/2023]
Abstract
Allergic respiratory diseases are major health problems in paediatric population due their high level of prevalence and chronicity, and to their relevance in the costs and quality of life. One of the most important risk factors for the development of airway diseases in children and adolescents is atopy. The mainstays for the treatment of these diseases are avoiding allergens, controlling symptoms, and preventing them through sustained desensitization by allergen immunotherapy (AIT). AIT is a treatment option that consists in the administration of increasing amounts of allergens to modify the biological response to them, inducing long-term tolerance even after treatment has ended. This treatment approach has shown to decrease symptoms and improve quality of life, becoming cost effective for a large number of patients. In addition, it is considered the only treatment that can influence the natural course of the disease by targeting the cause of the allergic inflammatory response. The aim of this publication is to reflect the advances of AIT in the diagnosis and treatment of allergic respiratory diseases in children and adolescents reviewing articles published since 2000, establishing evidence categories to support the strength of the recommendations based on evidence. The first part of the article covers the prerequisite issues to understand how AIT is effective, such as the correct etiologic and clinical diagnosis of allergic respiratory diseases. Following this, the article outlines the advancements in understanding the mechanisms by which AIT achieve immune tolerance to allergens. Administration routes, treatment regimens, dose and duration, efficacy, safety, and factors associated with adherence are also reviewed. Finally, the article reviews future advances in the research of AIT.
Collapse
Affiliation(s)
- M Tortajada-Girbés
- Paediatric Allergology and Pulmonology Unit, Dr. Peset University Hospital, Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain.
| | - M Mesa Del Castillo
- Paediatric Allergology and Neumology Unit, Hospital El Escorial, Madrid, Spain
| | - H Larramona
- Paediatric Allergology and Pulmonology Unit, Department of Paediatrics, University Autonoma of Barcelona, and Corporacio Sanitaria Parc Tauli, Hospital of Sabadell, Barcelona, Spain
| | - J M Lucas
- Pediatric Allergy and Immunology Unit, Virgen Arrixaca Clinic Universitary Hospital, Murcia, Spain
| | - M Álvaro
- Allergy and Clinical Immunology Section, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - A I Tabar
- Servicio de Alergología. Complejo Hospitalario de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), RETIC de Asma, Reacciones adversas y Alérgicas (ARADYAL), Pamplona, Spain
| | - M J Jerez
- Publications Office of the European Union, Luxembourg
| | - A Martínez-Cañavate
- Paediatric Allergology and Neumology Unit, Complejo Hospitalario Universitario de Granada, Spain
| |
Collapse
|
76
|
Coronel R, Jesus DM, Dalle Ore L, Mymryk JS, Hertel L. Activation of Langerhans-Type Dendritic Cells Alters Human Cytomegalovirus Infection and Reactivation in a Stimulus-Dependent Manner. Front Microbiol 2016; 7:1445. [PMID: 27683575 PMCID: PMC5021960 DOI: 10.3389/fmicb.2016.01445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/30/2016] [Indexed: 12/18/2022] Open
Abstract
Oral mucosal Langerhans cells (LC) are likely to play important roles in host defense against infection by human cytomegalovirus (CMV). We previously showed that in vitro-differentiated immature LC (iLC) populations contain smaller amounts of infected cells but produce higher yields than mature LC (mLC) cultures, obtained by iLC stimulation with fetal bovine serum (FBS), CD40 ligand (CD40L) and lipopolysaccharide (LPS). Here, we sought to determine if exposure to select stimuli can improve LC permissiveness to infection, if specific components of the mLC cocktail are responsible for lowering viral yields, if this is due to defects in progeny production or release, and if these restrictions are also effective against reactivated virus. None of the stimuli tested extended the proportion of infected cells to 100%, suggesting that the block to infection onset cannot be fully removed. While CD40L and FBS exerted positive effects on viral progeny production per cell, stimulation with LPS alone or in combination with CD40L was detrimental. Reductions in viral titers were not due to defects in progeny release, and the permissive or restrictive intracellular environment established upon exposure to each stimulus appeared to act in a somewhat similar way toward lytic and latent infections.
Collapse
Affiliation(s)
- Roxanne Coronel
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute Oakland, CA, USA
| | - Desyree M Jesus
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute Oakland, CA, USA
| | - Lucia Dalle Ore
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute Oakland, CA, USA
| | - Joe S Mymryk
- Department of Microbiology and Immunology and Department of Oncology, The University of Western Ontario London, ON, Canada
| | - Laura Hertel
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute Oakland, CA, USA
| |
Collapse
|
77
|
Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Mechanisms of Aeroallergen Immunotherapy: Subcutaneous Immunotherapy and Sublingual Immunotherapy. Immunol Allergy Clin North Am 2016; 36:71-86. [PMID: 26617228 DOI: 10.1016/j.iac.2015.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Allergen immunotherapy (AIT) is an effective way to treat allergic disorders, targeting the underlying mechanisms and altering the disease course by inducing a long-lasting clinical and immune tolerance to allergens. Although sublingual and subcutaneous routes are used in daily practice, many novel ways to decrease side effects and duration and increase efficacy have been pursued. Further studies are needed to develop biomarkers for the identification of AIT responder patients and also to use the developed knowledge in allergy prevention studies. Future directions in AIT include treatments for autoimmune diseases, chronic infections, organ transplantation, and breaking immune tolerance to cancer cells.
Collapse
Affiliation(s)
- Cevdet Ozdemir
- Department of Pediatric Allergy, Memorial Atasehir Hospital, Memorial Health Group, Vedat Gunyol Cad. 28-30, Istanbul 34758, Turkey
| | - Umut Can Kucuksezer
- Department of Immunology, Institute of Experimental Medicine (DETAE), Vakif Gureba Cad, Istanbul University, Istanbul 34093, Turkey
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Obere Strasse, CH-7270, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Obere Strasse, CH-7270, Davos, Switzerland.
| |
Collapse
|
78
|
Davis EM. Gene Sequence Analyses of the Healthy Oral Microbiome in Humans and Companion Animals. J Vet Dent 2016; 33:97-107. [DOI: 10.1177/0898756416657239] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
It has long been accepted that certain oral bacterial species are responsible for the development of periodontal disease. However, the focus of microbial and immunological research is shifting from studying the organisms associated with disease to examining the indigenous microbial inhabitants that are present in health. Microbiome refers to the aggregate genetic material of all microorganisms living in, or on, a defined habitat. Recent developments in gene sequence analysis have enabled detection and identification of bacteria from polymicrobial samples, including subgingival plaque. Diversity surveys utilizing this technology have demonstrated that bacterial culture techniques have vastly underestimated the richness and diversity of microorganisms in vivo, since only certain bacteria grow in vitro. Surveys using gene sequence analysis have demonstrated that the healthy oral microbiome is composed of an unexpectedly high number of diverse species, including putative pathogens. These findings support the view that coevolution microorganisms and macroscopic hosts has occurred in which certain microorganisms have adapted to survive in the oral cavity and host immune tolerance has allowed the establishment of a symbiotic relationship in which both parties receive benefits (mutualism). This review describes gene sequence analysis as an increasingly common, culture-independent tool for detecting bacteria in vivo and describes the results of recent oral microbiome diversity surveys of clinically healthy humans, dogs, and cats. Six bacterial phyla consistently dominated the healthy oral microbiome of all 3 host species. Previous hypotheses on etiology of periodontitis are reviewed in light of new scientific findings. Finally, the consideration that clinically relevant periodontal disease occurs when immune tolerance of the symbiotic oral microbiome is altered to a proinflammatory response will be discussed.
Collapse
Affiliation(s)
- Eric M. Davis
- Animal Dental Specialists of Upstate New York, Fayetteville, NY, USA
| |
Collapse
|
79
|
Cortez VS, Colonna M. Diversity and function of group 1 innate lymphoid cells. Immunol Lett 2016; 179:19-24. [PMID: 27394699 DOI: 10.1016/j.imlet.2016.07.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/05/2016] [Indexed: 12/17/2022]
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous population of cells with diverse roles in immune responses. Three major groups of ILCs have been defined on the basis of similarity in their production of signature cytokines, developmental requirements, and phenotypic markers. Group 1 ILCs produce IFN-γ, express the T-box transcription factors (TF) T-bet and/or Eomesodermin (Eomes), group 2 ILCs secrete IL-5 and IL-13 and express the TF GATA-3, while group 3 ILCs produce IL-22 and IL-17 and express the TF RORgt. In this review, we will briefly overview each group in terms of phenotype, function and development and then focus more extensively on group 1 ILCs, expanding on their emerging diversity, their disparate functions and the differences between NK cells and ILC1.
Collapse
Affiliation(s)
- Victor S Cortez
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
80
|
Donati C, Zolfo M, Albanese D, Tin Truong D, Asnicar F, Iebba V, Cavalieri D, Jousson O, De Filippo C, Huttenhower C, Segata N. Uncovering oral Neisseria tropism and persistence using metagenomic sequencing. Nat Microbiol 2016; 1:16070. [PMID: 27572971 DOI: 10.1038/nmicrobiol.2016.70] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 04/19/2016] [Indexed: 12/18/2022]
Abstract
Microbial epidemiology and population genomics have previously been carried out near-exclusively for organisms grown in vitro. Metagenomics helps to overcome this limitation, but it is still challenging to achieve strain-level characterization of microorganisms from culture-independent data with sufficient resolution for epidemiological modelling. Here, we have developed multiple complementary approaches that can be combined to profile and track individual microbial strains. To specifically profile highly recombinant neisseriae from oral metagenomes, we integrated four metagenomic analysis techniques: single nucleotide polymorphisms in the clade's core genome, DNA uptake sequence signatures, metagenomic multilocus sequence typing and strain-specific marker genes. We applied these tools to 520 oral metagenomes from the Human Microbiome Project, finding evidence of site tropism and temporal intra-subject strain retention. Although the opportunistic pathogen Neisseria meningitidis is enriched for colonization in the throat, N. flavescens and N. subflava populate the tongue dorsum, and N. sicca, N. mucosa and N. elongata the gingival plaque. The buccal mucosa appeared as an intermediate ecological niche between the plaque and the tongue. The resulting approaches to metagenomic strain profiling are generalizable and can be extended to other organisms and microbiomes across environments.
Collapse
Affiliation(s)
- Claudio Donati
- Computational Biology Unit, Research and Innovation Centre, Fondazione Edmund Mach, Via Edmund Mach 1, 38010 San Michele All'adige, Italy
| | - Moreno Zolfo
- Centre for Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Davide Albanese
- Computational Biology Unit, Research and Innovation Centre, Fondazione Edmund Mach, Via Edmund Mach 1, 38010 San Michele All'adige, Italy
| | - Duy Tin Truong
- Centre for Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Francesco Asnicar
- Centre for Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Valerio Iebba
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci Bolognetti Foundation, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Duccio Cavalieri
- Department of Biology, University of Florence, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Firenze, Italy.,Institute of Biometeorology, National Research Council (IBIMET-CNR), Via Caproni 8, 50145 Firenze, Italy
| | - Olivier Jousson
- Centre for Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Carlotta De Filippo
- Institute of Biometeorology, National Research Council (IBIMET-CNR), Via Caproni 8, 50145 Firenze, Italy
| | - Curtis Huttenhower
- Biostatistics Department, Harvard School of Public Health, Boston, Massachusetts 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Nicola Segata
- Centre for Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
81
|
Reinartz SM, van Tongeren J, van Egmond D, de Groot EJJ, Fokkens WJ, van Drunen CM. Dendritic Cell Subsets in Oral Mucosa of Allergic and Healthy Subjects. PLoS One 2016; 11:e0154409. [PMID: 27166951 PMCID: PMC4864364 DOI: 10.1371/journal.pone.0154409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/24/2016] [Indexed: 12/24/2022] Open
Abstract
Immunohistochemistry was used to identify, enumerate, and describe the tissue distribution of Langerhans type (CD1a and CD207), myeloid (CD1c and CD141), and plasmacytoid (CD303 and CD304) dendritic cell subsets in oral mucosa of allergic and non-allergic individuals. Allergic individuals have more CD141+ myeloid cells in epithelium and more CD1a+ Langerhans cells in the lamina propria compared to healthy controls, but similar numbers for the other DC subtypes. Our data are the first to describe the presence of CD303+ plasmacytoid DCs in human oral mucosa and a dense intraepithelial network of CD141+ DCs. The number of Langerhans type DCs (CD1a and CD207) and myeloid DCs (CD1c), was higher in the oral mucosa than in the nasal mucosa of the same individual independent of the atopic status.
Collapse
Affiliation(s)
- Susanne M. Reinartz
- Department of Otorhinolaryngology, Academic Medical Center, Amsterdam, the Netherlands
| | - Joost van Tongeren
- Department of Otorhinolaryngology, Academic Medical Center, Amsterdam, the Netherlands
- * E-mail:
| | - Danielle van Egmond
- Department of Otorhinolaryngology, Academic Medical Center, Amsterdam, the Netherlands
| | - Esther J. J. de Groot
- Department of Otorhinolaryngology, Academic Medical Center, Amsterdam, the Netherlands
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology, Academic Medical Center, Amsterdam, the Netherlands
| | | |
Collapse
|
82
|
de Steenhuijsen Piters WAA, Sanders EAM, Bogaert D. The role of the local microbial ecosystem in respiratory health and disease. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0294. [PMID: 26150660 DOI: 10.1098/rstb.2014.0294] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Respiratory tract infections are a major global health concern, accounting for high morbidity and mortality, especially in young children and elderly individuals. Traditionally, highly common bacterial respiratory tract infections, including otitis media and pneumonia, were thought to be caused by a limited number of pathogens including Streptococcus pneumoniae and Haemophilus influenzae. However, these pathogens are also frequently observed commensal residents of the upper respiratory tract (URT) and form-together with harmless commensal bacteria, viruses and fungi-intricate ecological networks, collectively known as the 'microbiome'. Analogous to the gut microbiome, the respiratory microbiome at equilibrium is thought to be beneficial to the host by priming the immune system and providing colonization resistance, while an imbalanced ecosystem might predispose to bacterial overgrowth and development of respiratory infections. We postulate that specific ecological perturbations of the bacterial communities in the URT can occur in response to various lifestyle or environmental effectors, leading to diminished colonization resistance, loss of containment of newly acquired or resident pathogens, preluding bacterial overgrowth, ultimately resulting in local or systemic bacterial infections. Here, we review the current body of literature regarding niche-specific upper respiratory microbiota profiles within human hosts and the changes occurring within these profiles that are associated with respiratory infections.
Collapse
Affiliation(s)
- Wouter A A de Steenhuijsen Piters
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Elisabeth A M Sanders
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Debby Bogaert
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
83
|
Boks MA, Gunput STG, Kosten I, Gibbs S, van Vliet SJ, Ligtenberg AJM, van Kooyk Y. The Human Glycoprotein Salivary Agglutinin Inhibits the Interaction of DC-SIGN and Langerin with Oral Micro-Organisms. J Innate Immun 2016; 8:350-61. [PMID: 27082983 DOI: 10.1159/000443016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 12/03/2015] [Indexed: 11/19/2022] Open
Abstract
Salivary agglutinin (SAG), also known as gp340 or SALSA, is a glycoprotein encoded by the Deleted in Malignant Brain Tumours 1 gene and is abundantly present in human saliva. SAG aggregates bacteria and viruses, thereby promoting their clearance from the oral cavity. The mucosa lining the oral cavity contains dendritic cells (DC) and Langerhans cells (LC), which express the C-type lectin receptors (CLR) DC-SIGN and Langerin, respectively. Both DC-SIGN and Langerin recognise mannose and fucose carbohydrate structures on pathogens and self-glycoproteins to regulate immunity and homeostasis. The purpose of this study was to investigate whether SAG interacts with these CLR and whether this interferes with the binding to oral pathogens. We show that whole parotid saliva and SAG, when coated to microplates, strongly interact with DC-SIGN and Langerin, probably via mannose and fucose structures. Also, primary human DC and LC bind parotid saliva and SAG via DC-SIGN and Langerin, respectively. Furthermore, SAG binding to DC-SIGN or Langerin prevented binding to the micro-organisms Candida albicans and Escherichia coli which express mannose and fucose-containing glycan structures. Thus, binding of saliva glycoprotein SAG to DC-SIGN and Langerin may inhibit pathogen-DC/LC interactions, and could prove to be a new immunomodulatory mechanism of SAG.
Collapse
Affiliation(s)
- Martine A Boks
- Department of Molecular Cell Biology and Immunology, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
84
|
Elkassaby A, Kotb M. Management of anterior urethral strictures with buccal mucosa: Our pioneering experience. AFRICAN JOURNAL OF UROLOGY 2016. [DOI: 10.1016/j.afju.2016.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
85
|
Masuyama K, Goto M, Takeno S, Ohta N, Okano M, Kamijo A, Suzuki M, Terada T, Sakurai D, Horiguchi S, Honda K, Matsune S, Yamada T, Sakashita M, Yuta A, Fuchiwaki T, Miyanohara I, Nakayama T, Okamoto Y, Fujieda S. Guiding principles of sublingual immunotherapy for allergic rhinitis in Japanese patients. Auris Nasus Larynx 2015; 43:1-9. [PMID: 26615715 DOI: 10.1016/j.anl.2015.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/03/2015] [Accepted: 08/26/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Sublingual immunotherapy (SLIT) appears to offer practical advantages for the treatment of allergic rhinitis (AR). Based on a review of the scientific literature, we present recommendations as guiding principles to administer SLIT safely. METHODS Clinical questions concerning SLIT were prepared. Literature published between January 2003 and December 2012 was searched from PubMed, the Cochrane Library, and Japana Centra Revuo Medicina. Qualified studies were analyzed and the results were evaluated, consolidated, and codified. We answered 17 clinical questions and, based on this, presented evidence-based recommendations. RESULTS Sublingual immunotherapy improved symptoms (e.g., quality of life [QOL]) and reduced medication scores in seasonal AR and perennial AR. Most SLIT-induced adverse effects were local oral reactions, although systemic adverse effects such as gastrointestinal symptoms, urticaria, and asthma are occasionally reported. There have been no reports of lethal anaphylactic reactions by SLIT. When SLIT is continued for 3-4 years, its effect persists long after discontinuation. CONCLUSION A correct diagnosis of AR and sufficient informed consent from patients are required before initiating SLIT. Sublingual immunotherapy should be continued for 3 years or longer. The initial administration of SLIT during the uptitration of an allergen vaccine and the general condition of patients are critical for the safe performance of SLIT.
Collapse
Affiliation(s)
- Keisuke Masuyama
- Department of Otorhinolaryngology, University of Yamanashi, Japan
| | - Minoru Goto
- Department of Otorhinolaryngology - Head and Neck Surgery, Nippon Medical School, Japan
| | - Sachio Takeno
- Department of Otolaryngology - Head and Neck Surgery, Hiroshima University, Japan
| | - Nobuo Ohta
- Department of Otolaryngology - Head and Neck Surgery, Yamagata University, Japan
| | - Mitsuhiro Okano
- Department of Otolaryngology - Head and Neck Surgery, Okayama University, Okayama, Japan
| | - Atsushi Kamijo
- Otorhinolaryngology/Allergy Center, Saitama Medical University, Japan
| | - Motohiko Suzuki
- Department of Otolaryngology - Head and Neck Surgery, Nagoya City University, Japan
| | - Tetsuya Terada
- Department of Otorhinolaryngology, Osaka Medical University, Japan
| | - Daiju Sakurai
- Department of Otolaryngology - Head and Neck Surgery, Chiba University, Japan
| | | | - Kohei Honda
- Department of Otorhinolaryngology - Head and Neck Surgery, Akita University, Japan
| | - Shoji Matsune
- Department of Otolaryngology Nippon Medical School, Musashikosugi Hospital, Japan
| | - Takechiyo Yamada
- Department of Otolaryngology - Head and Neck Surgery, University of Fukui, Japan
| | - Masafumi Sakashita
- Department of Otolaryngology - Head and Neck Surgery, University of Fukui, Japan
| | | | | | - Ikuyo Miyanohara
- Department of Otolaryngology - Head and Neck Surgery, Kagoshima University, Japan
| | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Japan
| | - Yoshitaka Okamoto
- Department of Otolaryngology - Head and Neck Surgery, Chiba University, Japan
| | - Shigeharu Fujieda
- Department of Otolaryngology - Head and Neck Surgery, University of Fukui, Japan.
| |
Collapse
|
86
|
Gingiva Equivalents Secrete Negligible Amounts of Key Chemokines Involved in Langerhans Cell Migration Compared to Skin Equivalents. J Immunol Res 2015; 2015:627125. [PMID: 26539556 PMCID: PMC4619927 DOI: 10.1155/2015/627125] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/05/2015] [Indexed: 11/18/2022] Open
Abstract
Both oral mucosa and skin have the capacity to maintain immune homeostasis or regulate immune responses upon environmental assault. Whereas much is known about key innate immune events in skin, little is known about oral mucosa. Comparative studies are limited due to the scarce supply of oral mucosa for ex vivo studies. Therefore, we used organotypic tissue equivalents (reconstructed epithelium on fibroblast-populated collagen hydrogel) to study cross talk between cells. Oral mucosa and skin equivalents were compared regarding secretion of cytokines and chemokines involved in LC migration and general inflammation. Basal secretion, representative of homeostasis, and also secretion after stimulation with TNFα, an allergen (cinnamaldehyde), or an irritant (SDS) were assessed. We found that proinflammatory IL-18 and chemokines CCL2, CCL20, and CXCL12, all involved in LC migration, were predominantly secreted by skin as compared to gingiva. Furthermore, CCL27 was predominantly secreted by skin whereas CCL28 was predominantly secreted by gingiva. In contrast, general inflammatory cytokines IL-6 and CXCL8 were secreted similarly by skin and gingiva. These results indicate that the cytokines and chemokines triggering innate immunity and LC migration are different in skin and gingiva. This differential regulation should be figured into novel therapy or vaccination strategies in the context of skin versus mucosa.
Collapse
|
87
|
de Steenhuijsen Piters WAA, Sanders EAM, Bogaert D. The role of the local microbial ecosystem in respiratory health and disease. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140294. [PMID: 26150660 PMCID: PMC4528492 DOI: 10.1098/rstb.2014.0294;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory tract infections are a major global health concern, accounting for high morbidity and mortality, especially in young children and elderly individuals. Traditionally, highly common bacterial respiratory tract infections, including otitis media and pneumonia, were thought to be caused by a limited number of pathogens including Streptococcus pneumoniae and Haemophilus influenzae. However, these pathogens are also frequently observed commensal residents of the upper respiratory tract (URT) and form-together with harmless commensal bacteria, viruses and fungi-intricate ecological networks, collectively known as the 'microbiome'. Analogous to the gut microbiome, the respiratory microbiome at equilibrium is thought to be beneficial to the host by priming the immune system and providing colonization resistance, while an imbalanced ecosystem might predispose to bacterial overgrowth and development of respiratory infections. We postulate that specific ecological perturbations of the bacterial communities in the URT can occur in response to various lifestyle or environmental effectors, leading to diminished colonization resistance, loss of containment of newly acquired or resident pathogens, preluding bacterial overgrowth, ultimately resulting in local or systemic bacterial infections. Here, we review the current body of literature regarding niche-specific upper respiratory microbiota profiles within human hosts and the changes occurring within these profiles that are associated with respiratory infections.
Collapse
Affiliation(s)
- Wouter A. A. de Steenhuijsen Piters
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Elisabeth A. M. Sanders
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands,Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Debby Bogaert
- Department of Paediatric Immunology and Infectious Diseases, The Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands,e-mail:
| |
Collapse
|
88
|
Vacher G, Sublet E, Gurny R, Borchard G. Establishment and first characterization of a sublingual epithelial and immune cell co-culture model. Int J Pharm 2015; 482:61-7. [PMID: 25448555 DOI: 10.1016/j.ijpharm.2014.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/11/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
We describe here the establishment and first characterization of a co-culture model of human epithelial sublingual cells (HO-1-u-1 cell line) and human dendritic cells derived from human peripheral blood monocytes (PBMC). Cell culture conditions for HO-1-u-1 cells were optimized. First characterization of phenotypic features by electron microscopy and fluorescence imaging revealed resemblance to sublingual tissue specimen from healthy donors. Successful co-culturing of epithelial and dendritic cells (DCs) was confirmed by confocal laser scanning microscopy. Stimulation of HO-1-u1 cells alone and the epithelial/DC co-culture by incubation with liposomes, virosomes and influenza virus lead reproducibly to the release of inflammatory cytokine GM-CSF. This co-culture model may be suitable for elucidation of mechanisms involved in the immune response at the sublingual epithelium as well as for the evaluation of novel topical vaccines, potentially replacing cumbersome ex vivo and in vivo methods currently in place.
Collapse
Affiliation(s)
- Gaëlle Vacher
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland
| | - Emmanuelle Sublet
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland
| | - Robert Gurny
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland.
| |
Collapse
|
89
|
Dynamics of Human Cytomegalovirus Infection in CD34+ Hematopoietic Cells and Derived Langerhans-Type Dendritic Cells. J Virol 2015; 89:5615-32. [PMID: 25762731 DOI: 10.1128/jvi.00305-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/02/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Acquisition of human cytomegalovirus (CMV) usually occurs by contact between contaminated bodily fluids, such as urine and saliva, and host mucosal cells. Langerhans-type dendritic cells (LC) are the only type of immune cells found in the outermost layers of the oral mucosae, where they not only provide a first line of defense against CMV but can easily be targeted by orally administered vaccines, while their bone marrow resident progenitors are important sites of virus latency. In this work, we tracked the progress of infection in CD34(+) progenitor cells, immature LC (iLC), and mature LC (mLC) exposed to the clinical-like strain TB40-BAC4 or to the vaccine strain AD169varATCC, prior to their long-term maintenance under either immature or mature conditions. We show that the genomes of both strains are efficiently maintained in CD34(+) cells during their differentiation into iLC, although this requires the presence of larger amounts of input AD169varATCC DNA. Lipopolysaccharide- and CD40 ligand-induced maturation of iLC derived from latently infected progenitors was not associated with robust viral genome replication and progeny production, while maturation of directly infected iLC increased and prolonged expression of the viral immediate early proteins. While effective replication of viral genomes from both strains occurred only in mLC, both iLC and mLC produced viral progeny, suggesting that both types of LC may contribute to CMV horizontal transmission in vivo. IMPORTANCE Human CMV is usually acquired via the oral and nasal mucosae. Langerhans-type dendritic cells (LC) are the only type of immune cells found in the outermost layers of these tissues. Understanding how CMV interacts with LC and their hematopoietic progenitors is thus essential to develop innovative means of defense against this virus. Here we show that the genomes of a virulent and an attenuated strain of CMV are maintained in hematopoietic progenitor cells during their differentiation into immature LC and that maturation of these cells by exposure to lipopolysaccharide and CD40 ligand is not sufficient to trigger virus reactivation. While the extents of viral protein expression and genome replication were broadest in directly infected mature LC populations, similar amounts of viral progeny were detected in the supernatants of immature and mature LC, suggesting that these immune cells of the oral mucosa are likely to be important for CMV transmission within the human population.
Collapse
|
90
|
Effect of a protein-free diet in the development of food allergy and oral tolerance in BALB/c mice. Br J Nutr 2015; 113:935-43. [PMID: 25759975 DOI: 10.1017/s0007114515000173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aim of the present study was to investigate the effect of a protein-free diet in the induction of food allergy and oral tolerance in BALB/c mice. The experimental model used was mice that were fed, since weaning up to adulthood, a balanced diet in which all dietary proteins were replaced by amino acid diet (Aa). The absence of dietary proteins did not prevent the development of food allergy to ovalbumin (OVA) in these mice. However, Aa-fed mice produced lower levels of IgE, secretory IgA and cytokines. In addition, when compared with mice from control group, Aa-fed mice had a milder aversive reaction to the allergen measured by consumption of OVA-containing solution and weight loss during food allergy development. In addition, mice that did not have dietary proteins in their diets were less susceptible to induction of oral tolerance. One single oral administration was not enough to suppress specific serum Ig and IgG1 levels in the Aa-fed group, although it was efficient to induce suppression in the control group. The present results indicate that the stimulation by dietary proteins alters both inflammatory reactivity and regulatory immune reactivity in mice probably due to their effect in the maturation of the immune system.
Collapse
|
91
|
Muris J, Goossens A, Gonçalo M, Bircher AJ, Giménez-Arnau A, Foti C, Rustemeyer T, Feilzer AJ, Kleverlaan CJ. Sensitization to palladium and nickel in Europe and the relationship with oral disease and dental alloys. Contact Dermatitis 2015; 72:286-96. [DOI: 10.1111/cod.12327] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/01/2014] [Accepted: 11/15/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Joris Muris
- Department of Dental Materials Science; Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam; Gustav Mahlerlaan 3004 1081 LA Amsterdam The Netherlands
| | - An Goossens
- Contact Allergy Unit, Department of Dermatology; K.U. Leuven University Hospital; Kapucijnenvoer 33 3000 Leuven Belgium
| | - Margarida Gonçalo
- Department of Dermatology; Faculty of Medicine, University Hospital, University of Coimbra; Praceta Mota Pinto 3000-075 Coimbra Portugal
| | - Andreas J. Bircher
- Allergy Unit, Department of Dermatology; University Hospital Basel; Spitalstrasse 26 4031 Basel Switzerland
| | - Ana Giménez-Arnau
- Department of Dermatology; Hospital del Mar. Parc de Salut Mar, Universitat Autònoma de Barcelona; Passeig Maritim 25-29 08003 Barcelona Spain
| | - Caterina Foti
- Department of Biomedical Science and Human Oncology; Dermatological Clinic, University of Bari; Policlinico di Bari, Piazza Giulio Cesare 11 70124 Bari Italy
| | - Thomas Rustemeyer
- Department of Dermatology; VU University Medical Centre; De Boelelaan 1117 1081 HV Amsterdam The Netherlands
| | - Albert J. Feilzer
- Department of Dental Materials Science; Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam; Gustav Mahlerlaan 3004 1081 LA Amsterdam The Netherlands
| | - Cornelis J. Kleverlaan
- Department of Dental Materials Science; Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam; Gustav Mahlerlaan 3004 1081 LA Amsterdam The Netherlands
| |
Collapse
|
92
|
Zhang C, Ohno T, Kang S, Takai T, Azuma M. Repeated antigen painting and sublingual immunotherapy in mice convert sublingual dendritic cell subsets. Vaccine 2014; 32:5669-76. [PMID: 25168308 DOI: 10.1016/j.vaccine.2014.08.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 07/21/2014] [Accepted: 08/08/2014] [Indexed: 12/24/2022]
Abstract
The sublingual mucosa (SLM) is utilized as the site for sublingual immunotherapy (SLIT) to induce tolerance against allergens. The contribution of SLM-dendritic cells (SLM-DCs) has not been clarified. The aim of this study was to examine the dynamics and phenotype of SLM-DCs after topical antigen painting and SLIT. SLM-DCs were histologically evaluated after FITC painting. A novel murine Japanese cedar pollinosis (JCP) model was generated and change in SLM-DCs after SLIT was examined. The density of SLM-DCs was clearly lower compared with the buccal mucosa and dorsal surface of the tongue. Topical FITC painting on the SLM induced maximal recruitment of submucosal DCs (smDCs) at 6h, but most smDCs had vanished at 24h. Repeated painting on the SLM induced exhaustion and conversion of the smDC phenotype. CD206(high)CD11c(low) round-type cells with fewer dendrites and less lymph node migration capacity became dominant. In the murine model of JCP, SLIT efficiently inhibited clinical symptoms and allergen-mediated immunological responses. SLIT markedly reduced the number of SLM-DCs, converted to the round-type dominant phenotype and inhibited the activation of regional lymph node DCs. Topical antigen painting on the SLM induced rapid exhaustion and conversion of smDCs. The unique dynamics of SLM-DCs may contribute to tolerance induction in SLIT.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan
| | - Tatsukuni Ohno
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan
| | - Siwen Kang
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan
| | - Toshiro Takai
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan.
| |
Collapse
|
93
|
Frischmeyer-Guerrerio PA, Keet CA, Guerrerio AL, Chichester KL, Bieneman AP, Hamilton RG, Wood RA, Schroeder JT. Modulation of dendritic cell innate and adaptive immune functions by oral and sublingual immunotherapy. Clin Immunol 2014; 155:47-59. [PMID: 25173802 DOI: 10.1016/j.clim.2014.08.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 08/16/2014] [Accepted: 08/19/2014] [Indexed: 10/24/2022]
Abstract
Sublingual (SLIT) and oral immunotherapy (OIT) are promising treatments for food allergy, but underlying mechanisms are poorly understood. Dendritic cells (DCs) induce and maintain Th2-type allergen-specific T cells, and also regulate innate immunity through their expression of Toll-like receptors (TLRs). We examined how SLIT and OIT influenced DC innate and adaptive immune responses in children with IgE-mediated cow's milk (CM) allergy. SLIT, but not OIT, decreased TLR-induced IL-6 secretion by myeloid DCs (mDCs). SLIT and OIT altered mDC IL-10 secretion, a potent inhibitor of FcεRI-dependent pro-inflammatory responses. OIT uniquely augmented IFN-α and decreased IL-6 secretion by plasmacytoid DCs (pDCs), which was associated with reduced TLR-induced IL-13 release in pDC-T cell co-cultures. Both SLIT and OIT decreased Th2 cytokine secretion to CM in pDC-T, but not mDC-T, co-cultures. Therefore, SLIT and OIT exert unique effects on DC-driven innate and adaptive immune responses, which may inhibit allergic inflammation and promote tolerance.
Collapse
Affiliation(s)
- Pamela A Frischmeyer-Guerrerio
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Corinne A Keet
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Anthony L Guerrerio
- Division of Gastroenterology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Kristin L Chichester
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Anja P Bieneman
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Robert G Hamilton
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Robert A Wood
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - John T Schroeder
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
94
|
Wu RQ, Zhang DF, Tu E, Chen QM, Chen W. The mucosal immune system in the oral cavity-an orchestra of T cell diversity. Int J Oral Sci 2014; 6:125-32. [PMID: 25105816 PMCID: PMC4170154 DOI: 10.1038/ijos.2014.48] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2014] [Indexed: 02/05/2023] Open
Abstract
The mucosal immune system defends against a vast array of pathogens, yet it exhibits limited responses to commensal microorganisms under healthy conditions. The oral-pharyngeal cavity, the gateway for both the gastrointestinal and respiratory tracts, is composed of complex anatomical structures and is constantly challenged by antigens from air and food. The mucosal immune system of the oral-pharyngeal cavity must prevent pathogen entry while maintaining immune homeostasis, which is achieved via a range of mechanisms that are similar or different to those utilized by the gastrointestinal immune system. In this review, we summarize the features of the mucosal immune system, focusing on T cell subsets and their functions. We also discuss our current understanding of the oral-pharyngeal mucosal immune system.
Collapse
Affiliation(s)
- Rui-Qing Wu
- 1] Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, USA [2] State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dun-Fang Zhang
- 1] Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, USA [2] State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Eric Tu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, USA
| | - Qian-Ming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, USA
| |
Collapse
|
95
|
Beswick EJ, Johnson JR, Saada JI, Humen M, House J, Dann S, Qiu S, Brasier AR, Powell DW, Reyes VE, Pinchuk IV. TLR4 activation enhances the PD-L1-mediated tolerogenic capacity of colonic CD90+ stromal cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:2218-29. [PMID: 25070848 DOI: 10.4049/jimmunol.1203441] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Signaling via programmed death ligand-1 (PD-L1) and PD-L2 is crucial for maintaining peripheral tolerance. CD90(+) myofibroblasts/fibroblasts (CMFs) are major programmed cell death-1 (PD-1) ligand-expressing cells in normal human colonic mucosa. CMFs suppress activated CD4(+) T cell proliferation via PD-1 ligands. It is not known whether signaling through TLRs contribute to the regulation PD-1 ligands on CMFs upon colonic mucosal tolerance. In this study, we demonstrated that stimulation of TLR4 on human CMFs upregulates PD-L1, but not PD-L2, and reinforces CMF-mediated suppression of CD4(+) T cell proliferation and IFN-γ production. TLR4-mediated upregulation of PD-L1 on CMFs involved NF-κB pathways and was JAK2 and MyD88 dependent. MyD88-dependent stimulation of TLR1/2 and TLR5 also upregulated PD-L1 expression on CMFs in culture. PD-L1 expression was drastically decreased in vivo in the colonic mucosa of mice devoid of MyD88. Induction of MyD88 deficiency in CMFs in fibroblast-specific MyD88 conditional knockout mice resulted in a strong increase in a mucosal IFN-γ expression concomitantly with the abrogation of PD-L1 expression in CMFs under homeostasis and epithelial injury induced by dextran sodium sulfate. Together, these data suggest that MyD88-dependent TLR stimulation of CMFs in the normal colonic mucosa may reinforce these cells' anti-inflammatory capacity and thus contribute to the maintenance of mucosal tolerance.
Collapse
Affiliation(s)
- Ellen J Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131
| | - Jameel R Johnson
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Jamal I Saada
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Martin Humen
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555
| | - Jenifer House
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555
| | - Sara Dann
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Suimin Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; Institute of Translational Science, University of Texas Medical Branch, Galveston, TX 77555
| | - Don W Powell
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; Institute of Translational Science, University of Texas Medical Branch, Galveston, TX 77555; Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Victor E Reyes
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Irina V Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; Institute of Translational Science, University of Texas Medical Branch, Galveston, TX 77555; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
96
|
Baron-Bodo V, Horiot S, Lautrette A, Chabre H, Drucbert AS, Danzé PM, Sénéchal H, Peltre G, Galvain S, Zeldin RK, Horak F, Moingeon P. Heterogeneity of antibody responses among clinical responders during grass pollen sublingual immunotherapy. Clin Exp Allergy 2014; 43:1362-73. [PMID: 24261946 DOI: 10.1111/cea.12187] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/17/2013] [Accepted: 08/15/2013] [Indexed: 01/03/2023]
Abstract
BACKGROUND During allergen-specific sublingual immunotherapy (SLIT), the relevance of changes in specific IgE and IgG antibody titres to treatment efficacy remains to be evaluated at an individual patient level. OBJECTIVE To investigate whether antibody responses can be used as biomarkers for SLIT efficacy. METHODS Comprehensive quantitative, qualitative and functional analyses of allergen-specific IgA, IgE, IgG1-4 and IgM responses were performed using purified Phl p 1 to 12 allergens in sera, saliva and nasal secretions from 82 grass pollen allergic patients. These patients were enrolled in a randomized, double-blind placebo-controlled study and assessed in an allergen challenge chamber (ClinicalTrials.gov NCT00619827). Antibody responses were monitored in parallel to clinical responses before and after daily sublingual treatment for 4 months with either a grass pollen or a placebo tablet. RESULTS A significant mean improvement (i.e. 33-40.6%) in rhinoconjunctivitis total symptom scores was observed in SLIT recipients, irrespective of their baseline patterns of IgE sensitization (i.e. narrow, intermediate, broad) to grass pollen allergens. SLIT did not induce any de novo IgE sensitization. Clinical responders encompassed both immunoreactive patients who exhibited strong increases in titres, affinity and/or blocking activity of grass-pollen-specific IgGs (representing 17% of treated patients), as well as patients with no detectable antibody responses distinguishing them from the placebo group. No significant changes were detected in antibody titres in saliva and nasal washes, even in clinical responders. CONCLUSIONS AND CLINICAL RELEVANCE Sublingual immunotherapy with a grass pollen tablet is efficacious irrespective of the patients' baseline sensitization to either single or multiple grass pollen allergens. Seric IgG responses may contribute to SLIT-induced clinical tolerance in a fraction (i.e. 17%) of patients, but additional immune mechanisms are involved in most patients. Consequently, antibody responses cannot be used as a marker of SLIT efficacy at an individual patient level.
Collapse
|
97
|
Zaura E, Nicu EA, Krom BP, Keijser BJF. Acquiring and maintaining a normal oral microbiome: current perspective. Front Cell Infect Microbiol 2014; 4:85. [PMID: 25019064 PMCID: PMC4071637 DOI: 10.3389/fcimb.2014.00085] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/08/2014] [Indexed: 12/15/2022] Open
Abstract
The oral microbiota survives daily physical and chemical perturbations from the intake of food and personal hygiene measures, resulting in a long-term stable microbiome. Biological properties that confer stability in the microbiome are important for the prevention of dysbiosis—a microbial shift toward a disease, e.g., periodontitis or caries. Although processes that underlie oral diseases have been studied extensively, processes involved in maintaining of a normal, healthy microbiome are poorly understood. In this review we present our hypothesis on how a healthy oral microbiome is acquired and maintained. We introduce our view on the prenatal development of tolerance for the normal oral microbiome: we propose that development of fetal tolerance toward the microbiome of the mother during pregnancy is the major factor for a successful acquisition of a normal microbiome. We describe the processes that influence the establishment of such microbiome, followed by our perspective on the process of sustaining a healthy oral microbiome. We divide microbiome-maintenance factors into host-derived and microbe-derived, while focusing on the host. Finally, we highlight the need and directions for future research.
Collapse
Affiliation(s)
- Egija Zaura
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam Amsterdam, Netherlands
| | - Elena A Nicu
- Department of Periodontology, Academic Centre for Dentistry Amsterdam Amsterdam, Netherlands
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam Amsterdam, Netherlands
| | - Bart J F Keijser
- Microbiology and Systems Biology, TNO Earth, Environmental and Life Sciences Zeist, Netherlands ; Top Institute Food and Nutrition Wageningen, Netherlands
| |
Collapse
|
98
|
Hertel L. Human cytomegalovirus tropism for mucosal myeloid dendritic cells. Rev Med Virol 2014; 24:379-95. [PMID: 24888709 DOI: 10.1002/rmv.1797] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/01/2014] [Accepted: 05/07/2014] [Indexed: 02/01/2023]
Abstract
Human CMV infections are a serious source of morbidity and mortality for immunocompromised patients and for the developing fetus. Because of this, the development of new strategies to prevent CMV acquisition and transmission is a top priority. Myeloid dendritic cells (DC) residing in the oral and nasal mucosae are among the first immune cells to encounter CMV during entry and greatly contribute to virus dissemination, reactivation from latency, and horizontal spread. Albeit affected by the immunoevasive tactics of CMV, mucosal DC remain potent inducers of cellular and humoral immune responses against this virus. Their natural functions could thus be exploited to generate long-lasting protective immunity against CMV by vaccination via the oronasal mucosae. Although related, epithelial Langerhans-type DC and dermal monocyte-derived DC interact with CMV in dramatically different ways. Whereas immature monocyte-derived DC are fully permissive to infection, for instance, immature Langerhans-type DC are completely resistant. Understanding these differences is essential to design innovative vaccines and new antiviral compounds to protect these cells from CMV infection in vivo.
Collapse
Affiliation(s)
- Laura Hertel
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute, Oakland, CA, 94609, USA
| |
Collapse
|
99
|
Li F, Jin X, Liu B, Zhuang W, Scalabrin D. Follow-up formula consumption in 3- to 4-year-olds and respiratory infections: an RCT. Pediatrics 2014; 133:e1533-40. [PMID: 24843061 DOI: 10.1542/peds.2013-3598] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Children are vulnerable to diet inadequacies, which may affect immune function. Our objective was to determine if a follow-up formula (FUF) containing DHA, the prebiotics PDX and GOS, and yeast β-glucan affects incidence of respiratory infections and diarrheal disease in healthy children. METHODS In a double-blind, randomized, controlled, prospective trial, 3-4 year old children were fed 3 servings per day of either a FUF with 25 mg DHA, 1.2 g PDX/GOS, and 8.7 mg yeast β-glucan per serving or an unfortified, cow's milk-based beverage (control) for 28 weeks. Fecal and blood samples were collected to assess immune markers and iron/zinc status. Incidence of acute respiratory infections (ARI), diarrheal disease, and antibiotic treatment were obtained from medical records. RESULTS The FUF group had fewer episodes and shorter duration of ARI (mean days [SE]; control = 4.3 [0.2]; FUF = 3.5 [0.2]; P = .007), less antibiotic use (n [%]; control = 21 [14%]; FUF = 8 [5%]; P = .01), and fewer missed days of day care due to illness. No diarrheal disease was diagnosed in either group. The FUF group had higher interleukin-10 and white blood cell count at the end of the study. There were no differences in hemoglobin, serum ferritin and zinc, or fecal secretory immunoglobulin A. CONCLUSIONS Daily consumption of a FUF was associated with fewer episodes and shorter duration of ARI, as well as less antibiotic use. The children who consumed the FUF had increased interleukin-10 and white blood cells, suggesting an antiinflammatory mechanism and/or an increase of effector immune cells.
Collapse
Affiliation(s)
- Fei Li
- Developmental and Behavioral Pediatric Department, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China; and
| | - Xingming Jin
- Developmental and Behavioral Pediatric Department, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China; and
| | - Bryan Liu
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, Indiana
| | - Weihong Zhuang
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, Indiana
| | - Deolinda Scalabrin
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, Indiana
| |
Collapse
|
100
|
Feliciano RP, Meudt JJ, Shanmuganayagam D, Krueger CG, Reed JD. Ratio of "A-type" to "B-type" proanthocyanidin interflavan bonds affects extra-intestinal pathogenic Escherichia coli invasion of gut epithelial cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:3919-3925. [PMID: 24215458 DOI: 10.1021/jf403839a] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Gut colonization by extra-intestinal pathogenic Escherichia coli (ExPEC) increases the risk of subsequent infections, including urinary tract infection and septicemia. Previous work suggests that cranberry proanthocyanidins (PAC) interact with bacterial surface factors, altering bacterial interaction with host cells. Methods were developed to determine if ratios of "A-type" to "B-type" interflavan bonds in PAC affect ExPEC agglutination and invasion of enterocytes. In cranberries, 94.5% of PAC contain one or more "A-type" bonds, whereas in apples, 88.3% of PAC contain exclusively "B-type" bonds. Results show that cranberry "A-type" PAC have greater bioactivity than apple "B-type" PAC for increasing ExPEC agglutination and decreasing ExPEC epithelial cell invasion.
Collapse
Affiliation(s)
- Rodrigo P Feliciano
- Department of Food Science, University of Wisconsin-Madison , 1605 Linden Drive, Madison, Wisconsin 53706, United States
| | | | | | | | | |
Collapse
|