51
|
Sanchez-Solares J, Sanchez L, Pablo-Torres C, Diaz-Fernandez C, Sørensen P, Barber D, Gomez-Casado C. Celiac Disease Causes Epithelial Disruption and Regulatory T Cell Recruitment in the Oral Mucosa. Front Immunol 2021; 12:623805. [PMID: 33717129 PMCID: PMC7947325 DOI: 10.3389/fimmu.2021.623805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/13/2021] [Indexed: 01/18/2023] Open
Abstract
Celiac disease (CD) is a chronic autoimmune disease characterized by an immune-triggered enteropathy upon gluten intake. The only current treatment available is lifelong Gluten Free Diet (GFD). Several extraintestinal manifestations have been described in CD, some affecting the oral mucosa. Thus, we hypothesized that oral mucosa could potentially be a target for novel biomarkers and an administration route for CD treatment. Six de novo diagnosed and seven CD patients under GFD for at least 1 year were recruited. Non-celiac subjects (n = 8) were recruited as control group. Two biopsies of the cheek lining were taken from each subject for mRNA analysis and immunohistochemical characterization. We observed a significant decrease in the expression of epithelial junction proteins in all CD patients, indicating that oral mucosa barrier integrity is compromised. FoxP3+ population was greatly increased in CD patients, suggesting that Tregs are recruited to the damaged mucosa, even after avoidance of gluten. Amphiregulin mRNA levels from Peripheral Blood Mononuclear Cells (PBMCs) and epithelial damage in the oral mucosa correlated with Treg infiltration in all the experimental groups, suggesting that recruited Tregs might display a “repair” phenotype. Based on these results, we propose that oral mucosa is altered in CD and, as such, might have diagnostic potential. Furthermore, due to its tolerogenic nature, it could be an important target for oral immunotherapy.
Collapse
Affiliation(s)
- Javier Sanchez-Solares
- Institute of Applied Molecular Medicine, Hospitals Madrid (HM) Group, San Pablo-CEU University, Madrid, Spain
| | - Luis Sanchez
- Service of Gastroenterology, University Hospital San Agustin (HUSA), Aviles, Spain
| | - Carmela Pablo-Torres
- Institute of Applied Molecular Medicine, Hospitals Madrid (HM) Group, San Pablo-CEU University, Madrid, Spain
| | - Celso Diaz-Fernandez
- Department of Otolaryngology Head and Neck Surgery, University Hospital San Agustin (HUSA), Aviels, Spain
| | - Poul Sørensen
- Allero Therapeutics BV, Rotterdam, Netherlands.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Domingo Barber
- Institute of Applied Molecular Medicine, Hospitals Madrid (HM) Group, San Pablo-CEU University, Madrid, Spain.,ARADyAL-RD16/0006/0015, Thematic Network and Cooperative Research Centers, ISCIII, Madrid, Spain
| | - Cristina Gomez-Casado
- Institute of Applied Molecular Medicine, Hospitals Madrid (HM) Group, San Pablo-CEU University, Madrid, Spain.,ARADyAL-RD16/0006/0015, Thematic Network and Cooperative Research Centers, ISCIII, Madrid, Spain
| |
Collapse
|
52
|
Coffey JW, Gaiha GD, Traverso G. Oral Biologic Delivery: Advances Toward Oral Subunit, DNA, and mRNA Vaccines and the Potential for Mass Vaccination During Pandemics. Annu Rev Pharmacol Toxicol 2021; 61:517-540. [PMID: 32466690 PMCID: PMC8057107 DOI: 10.1146/annurev-pharmtox-030320-092348] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oral vaccination enables pain-free and self-administrable vaccine delivery for rapid mass vaccination during pandemic outbreaks. Furthermore, it elicits systemic and mucosal immune responses. This protects against infection at mucosal surfaces, which may further enhance protection and minimize the spread of disease. The gastrointestinal (GI) tract presents a number of prospective mucosal inductive sites for vaccine targeting, including the oral cavity, stomach, and small intestine. However, currently available oral vaccines are effectively limited to live-attenuated and inactivated vaccines against enteric diseases. The GI tract poses a number of challenges,including degradative processes that digest biologics and mucosal barriers that limit their absorption. This review summarizes the approaches currently under development and future opportunities for oral vaccine delivery to established (intestinal) and relatively new (oral cavity, stomach) mucosal targets. Special consideration is given to recent advances in oral biologic delivery that offer promise as future platforms for the administration of oral vaccines.
Collapse
Affiliation(s)
- Jacob William Coffey
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunology, University of Melbourne, Victoria, 3000, Australia
| | - Gaurav Das Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139, USA
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| |
Collapse
|
53
|
Koren N, Zubeidat K, Saba Y, Horev Y, Barel O, Wilharm A, Heyman O, Wald S, Eli-Berchoer L, Shapiro H, Nadler C, Elinav E, Wilensky A, Prinz I, Bercovier H, Hovav AH. Maturation of the neonatal oral mucosa involves unique epithelium-microbiota interactions. Cell Host Microbe 2021; 29:197-209.e5. [PMID: 33412104 DOI: 10.1016/j.chom.2020.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/26/2020] [Accepted: 12/03/2020] [Indexed: 12/19/2022]
Abstract
Postnatal host-microbiota interplay governs mucosal homeostasis and is considered to have life-long health consequences. The intestine monolayer epithelium is critically involved in such early-life processes; nevertheless, the role of the oral multilayer epithelium remains ill defined. We demonstrate that unlike the intestine, the neonate oral cavity is immensely colonized by the microbiota that decline to adult levels during weaning. Neutrophils are present in the oral epithelium prenatally, and exposure to the microbiota postnatally further recruits them to the preamble neonatal epithelium by γδT17 cells. These neutrophils virtually disappear during weaning as the epithelium seals. The neonate and adult epithelium display distinct turnover kinetics and transcriptomic signatures, with neonate epithelium reminiscent of the signature found in germ-free mice. Microbial reduction during weaning is mediated by the upregulation of saliva production and induction of salivary antimicrobial components by the microbiota. Collectively, unique postnatal interactions between the multilayer epithelium and microbiota shape oral homeostasis.
Collapse
Affiliation(s)
- Noam Koren
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Khaled Zubeidat
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yasmin Saba
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yael Horev
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Or Barel
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Anneke Wilharm
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Oded Heyman
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Sharon Wald
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Luba Eli-Berchoer
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Chen Nadler
- Oral Medicine Department, Hebrew University, Hadassah School of Dental Medicine, Jerusalem
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Hillel Bercovier
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Avi-Hai Hovav
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
54
|
Immunological Aspects of Dental Implant Rejection. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7279509. [PMID: 33376734 PMCID: PMC7744195 DOI: 10.1155/2020/7279509] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/29/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Nowadays, dental implants are a prominent therapeutic approach among dentists for replacing missing teeth. Failure in dental implants is a severe challenge recently. The factors which lead to dental implant failure are known. These factors can be categorized into different groups. In this article, we discussed the immunological aspects of implant failure as one of these groups. Cytokines and immune cells have extensive and various functions in peri-implantitis. The equilibrium between pro and anti-inflammatory cytokines and cells, which involve in this orchestra, has a crucial role in implant prognosis. In conclusion, immune cells, especially macrophages and dendritic cells, almost increased in the patients with implant failure. Also, proinflammatory cytokines were proposed as diagnostic factors according to their higher levels in dental implant rejection.
Collapse
|
55
|
Abstract
OBJECTIVES Allergic contact dermatitis is an uncommon but important cause of skin disease in the anogenital region. Relevant allergens are described in women and less commonly in men. The aim of this study was to describe outcomes of patch testing in men and women presenting with anogenital dermatoses. MATERIALS AND METHODS Cases patch tested for anogenital conditions at 2 patch test clinics in Sydney, Australia, from 2002 to 2017 were reviewed. Positive and relevant patch test reactions were recorded. RESULTS Thirty-seven women and 27 men were included. Dermatitis was the most common diagnosis, followed by psoriasis and lichen sclerosus. Thirty percent had a final diagnosis of allergic contact dermatitis. The most frequent relevant allergens were fragrance mix I (9%), patients own products (9%), Myroxylon pereirae (8%), cocamidopropyl betaine (3%), and benzocaine (3%). CONCLUSIONS The top positive and relevant allergens seen were in concordance with other reports from Australia and the rest of the world. Fragrances and medicaments are common allergens, and it is recommended that products used on anogenital skin be fragrance free. Testing patients own products is imperative.
Collapse
|
56
|
Essential role of submandibular lymph node dendritic cells in protective sublingual immunotherapy against murine allergy. Commun Biol 2020; 3:742. [PMID: 33288832 PMCID: PMC7721894 DOI: 10.1038/s42003-020-01466-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 11/04/2020] [Indexed: 01/12/2023] Open
Abstract
While sublingual immunotherapy (SLIT) is known as an allergen-specific treatment for type-1 allergies, how it controls allergic pathogenesis remains unclear. Here, we show the prerequisite role of conventional dendritic cells in submandibular lymph nodes (ManLNs) in the effectiveness of SLIT for the treatment of allergic disorders in mice. Deficiency of conventional dendritic cells or CD4+Foxp3+ regulatory T (Treg) cells abrogates the protective effect of SLIT against allergic disorders. Furthermore, sublingual antigenic application primarily induces antigen-specific CD4+Foxp3+ Treg cells in draining ManLNs, in which it is severely impaired in the absence of cDCs. In ManLNs, migratory CD11b+ cDCs are superior to other conventional dendritic cell subsets for the generation of antigen-specific CD4+Foxp3+ Treg cells, which is reflected by their dominancy in the tolerogenic features to favor this program. Thus, ManLNs are privileged sites in triggering mucosal tolerance mediating protect effect of SLIT on allergic disorders that requires a tolerogenesis of migratory CD11b+ conventional dendritic cells. Noriaki Miyanaga and Hideaki Takagi et al. identify an essential role for migratory dendritic cells in mediating immunotherapy treatment against allergies in mice. They show that submandibular lymph node dendritic cells induce regulatory T cells, and their absence abrogates the effectiveness of immunotherapy treatment.
Collapse
|
57
|
Creighton R, Schuch V, Urbanski AH, Giddaluru J, Costa-Martins AG, Nakaya HI. Network vaccinology. Semin Immunol 2020; 50:101420. [PMID: 33162295 DOI: 10.1016/j.smim.2020.101420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/31/2020] [Indexed: 01/21/2023]
Abstract
The structure and function of the immune system is governed by complex networks of interactions between cells and molecular components. Vaccination perturbs these networks, triggering specific pathways to induce cellular and humoral immunity. Systems vaccinology studies have generated vast data sets describing the genes related to vaccination, motivating the use of new approaches to identify patterns within the data. Here, we describe a framework called Network Vaccinology to explore the structure and function of biological networks responsible for vaccine-induced immunity. We demonstrate how the principles of graph theory can be used to identify modules of genes, proteins, and metabolites that are associated with innate and adaptive immune responses. Network vaccinology can be used to assess specific and shared molecular mechanisms of different types of vaccines, adjuvants, and routes of administration to direct rational design of the next generation of vaccines.
Collapse
Affiliation(s)
- Rachel Creighton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Viviane Schuch
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alysson H Urbanski
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jeevan Giddaluru
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur USP, São Paulo, Brazil
| | - Andre G Costa-Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur USP, São Paulo, Brazil
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur USP, São Paulo, Brazil.
| |
Collapse
|
58
|
Chandavarkar V, Mishra MN, Sangeetha R, Premalatha BR. The Current Understanding on Langerhans' Cells and Its Role in Oral Lesions. Contemp Clin Dent 2020; 11:211-216. [PMID: 33776345 PMCID: PMC7989755 DOI: 10.4103/ccd.ccd_4_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/11/2020] [Accepted: 06/19/2020] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Description of Langerhans' cells (LCs) as an important antigen-presenting cells responsible for detecting the antigens, recruiting T-cells, and thereby initiating the immune response. An adequate response of the mucosal immune system is essential to protect the mucosa against pathological conditions. Hence, a detailed review was planned about this unique antigen-presenting cell. METHODS A literature search of the electronic databases included the MEDLINE, EBSCOHOST, PUBMED, and hand searches of references retrieved were undertaken using the following MeSH terms "Langerhans cells," "LCs in Oral Lichen Planus," "Langerhans cell histiocytosis," "LCs and HIV," "LCs in Periodontitis." RESULTS LCs are present suprabasally in the epithelium of oral mucosa and in the epidermis of the skin. The role played by LCs though not fully elucidated, but several research studies indicate that these cells are involved in the pathogenesis of many oral diseases. In this article, the historical perspective, structure, function, origin, and phenotypic expressions of LCs are discussed in detail. The current understanding on the role of LCs in various oral lesions and its immunological characteristics are discussed. CONCLUSION LCs act as immune mediator cells, tumor cells, vectors of infected cells, and phagocytic cells. Further studies could bolster the knowledge about the role of Langerhans cells in the immune response of various oral diseases and thereby provide diagnostic tools and help for prognostic evaluation. This review illuminates the pivotal role of Langerhans cells and its immune surveillance as a "Sentinels" of the oral mucosa.
Collapse
Affiliation(s)
- Vidyadevi Chandavarkar
- Department of Oral Pathology, School of Dental Sciences, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Mithilesh N. Mishra
- Department of Oral Pathology, School of Dental Sciences, Sharda University, Greater Noida, Uttar Pradesh, India
| | - R. Sangeetha
- Department of Oral Pathology, Dayananda Sagar College of Dental Sciences, Bengaluru, Karnataka, India
| | - B. R. Premalatha
- Department of Oral Pathology and Microbiology, JSS Dental College and Hospital, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| |
Collapse
|
59
|
Heyman O, Horev Y, Koren N, Barel O, Aizenbud I, Aizenbud Y, Brandwein M, Shapira L, Hovav A, Wilensky A. Niche Specific Microbiota-Dependent and Independent Bone Loss around Dental Implants and Teeth. J Dent Res 2020; 99:1092-1101. [DOI: 10.1177/0022034520920577] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Oral mucosal homeostasis is achieved by complex immunologic mechanisms, orchestrating host immunity to adapt to the physiologic functions of the various specialized niches in the oral cavity. Dental implants introduce a novel mucosal niche to the immune system to deal with. Nevertheless, the immune mechanisms engaged toward implants and whether they have broader effects are not well defined. Using a murine model, we found an accumulation of neutrophils and RANKL-expressing T and B lymphocytes in the implant-surrounding mucosa, accompanied by local bone loss. Surprisingly, the presence of implants had an impact on remote periodontal sites, as elevated inflammation and accelerated bone loss were detected in intact distant teeth. This was due to microbial dysbiosis induced by the implants, since antibiotic treatment prevented bone loss around teeth. However, antibiotic treatment failed to prevent the loss of implant-supporting bone, highlighting the distinct mechanisms mediating bone loss at each site. Further analysis revealed that implants induced chronic lymphocyte activation and increased mRNA expression of IFN-α and accumulation of IFN-α–producing plasmacytoid dendritic cells, which we previously reported as bone-destructive immune responses. Collectively, this study demonstrates that implants have a strong and broad impact on oral mucosal homeostasis, inducing periodontal bone loss in a niche-specific manner that is both microbiota dependent and independent.
Collapse
Affiliation(s)
- O. Heyman
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| | - Y. Horev
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| | - N. Koren
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - O. Barel
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - I. Aizenbud
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Y. Aizenbud
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - M. Brandwein
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - L. Shapira
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| | - A.H. Hovav
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - A. Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
60
|
Oral Candidiasis in a Migraine Patient Taking Erenumab and Galcanezumab: a Case Report. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42399-020-00300-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
61
|
Fernandes DT, van Heerden WF, Ribeiro AC, Brandão TB, Mello ES, Rivera C, van Heerden MB, Gondak R, Santos-Silva AR, Vargas PA, Lopes MA. Different methods of cell quantification can lead to different results: a comparison of digital methods using a pilot study of dendritic cells in HIV-positive patients. Med Oral Patol Oral Cir Bucal 2020; 25:e431-e438. [PMID: 32134901 PMCID: PMC7211365 DOI: 10.4317/medoral.23472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/20/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Although new digital pathology tools have improved the positive cell quantification, there is a heterogeneity of the quantification methods in the literature. The aim of this study was to evaluate and propose a novel dendritic cells quantification method in squamous cell carcinoma comparing it with a conventional quantification method. MATERIAL AND METHODS Twenty-six squamous cell carcinomas HIV-positive cases affecting the oropharynx, lips and oral cavity were selected. Immunohistochemistry for CD1a, CD83, and CD207 was performed. The immunohistochemical stains were evaluated by automated examination using a positive pixel count algorithm. A conventional quantification method (unspecific area method; UA) and a novel method (specific area method; SA) were performed obtaining the corresponding density of positive dendritic cells for the intratumoral and peritumoral regions. The Mann-Whitney U test was used to verify the influence of the quantification methods on the positive cell counting according to the evaluated regions. Data were subjected to the ANOVA and Student's t-test to verify the influence of the tumour location, stage, histological grade, and amount of inflammation on the dendritic cells density counting. RESULTS The cell quantification method affected the dendritic cells counting independently of the evaluated region (P-value <0.05). Significant differences between methods were also observed according to the tumour features evaluations. CONCLUSIONS The positive cell quantification method influences the dendritic cells density results. Unlike the conventional method (UA method), the novel SA method avoids non-target areas included in the hotspots improving the reliability and reproducibility of the density cell quantification.
Collapse
Affiliation(s)
- D-T Fernandes
- Faculdade de Odontologia de Piracicaba - UNICAMP Departamento de Diagnóstico Oral - Semiologia Av. Limeira, 901 CEP 13.414-903 Piracicaba, São Paulo, Brasil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Abstract
The terminal differentiation of the epidermis is a complex physiological process. During the past few decades, medical genetics has shown that defects in the stratum corneum (SC) permeability barrier cause a myriad of pathological conditions, ranging from common dry skin to lethal ichthyoses. Contrarily, molecular phylogenetics has revealed that amniotes have acquired a specialized form of cytoprotection cornification that provides mechanical resilience to the SC. This superior biochemical property, along with desiccation tolerance, is attributable to the proper formation of the macromolecular protein-lipid complex termed cornified cell envelopes (CE). Cornification largely depends on the peculiar biochemical and biophysical properties of loricrin, which is a major CE component. Despite its quantitative significance, loricrin knockout (LKO) mice have revealed it to be dispensable for the SC permeability barrier. Nevertheless, LKO mice have brought us valuable lessons. It is also becoming evident that absent loricrin affects skin homeostasis more profoundly in many more aspects than previously expected. Through an extensive review of aggregate evidence, we discuss herein the functional significance of the thiol-rich protein loricrin from a biochemical, genetic, pathological, metabolic, or immunological aspect with some theoretical and speculative perspectives.
Collapse
Affiliation(s)
- Yosuke Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Dennis R. Roop
- Department of Dermatology and Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| |
Collapse
|
63
|
Scott NA, Mann ER. Regulation of mononuclear phagocyte function by the microbiota at mucosal sites. Immunology 2020; 159:26-38. [PMID: 31777068 PMCID: PMC6904663 DOI: 10.1111/imm.13155] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022] Open
Abstract
Mucosal tissues contain distinct microbial communities that differ drastically depending on the barrier site, and as such, mucosal immune responses have evolved to be tailored specifically for their location. Whether protective or regulatory immune responses against invading pathogens or the commensal microbiota occur is controlled by local mononuclear phagocytes (MNPs). Comprising macrophages and dendritic cells (DCs), the functions of these cells are highly dependent on the local environment. For example, the intestine contains the greatest bacterial load of any site in the body, and hence, intestinal MNPs are hyporesponsive to bacterial stimulation. This is thought to be one of the major mechanisms by which harmful immune responses directed against the trillions of harmless bacteria that line the gut lumen are avoided. Regulation of MNP function by the microbiota has been characterized in the most depth in the intestine but there are several mucosal sites that also contain their own microbiota. In this review, we present an overview of how MNP function is regulated by the microbiota at mucosal sites, highlighting recent novel pathways by which this occurs in the intestine, and new studies elucidating these interactions at mucosal sites that have been characterized in less depth, including the urogenital tract.
Collapse
Affiliation(s)
- Nicholas A. Scott
- Lydia Becker Institute of Immunology and InflammationUniversity of ManchesterManchesterUK
- Manchester Collaborative Centre for Inflammation ResearchFaculty of Biology, Medicine and HealthManchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Elizabeth R. Mann
- Lydia Becker Institute of Immunology and InflammationUniversity of ManchesterManchesterUK
- Manchester Collaborative Centre for Inflammation ResearchFaculty of Biology, Medicine and HealthManchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
64
|
DYNAMICS OF ATP-POSITIVE DENDRITIC CELLS IN RAT’S OROPHARYNGEAL SUBMUCOSA AFTER ANTENATAL ANTIGEN ADMINISTRATION. WORLD OF MEDICINE AND BIOLOGY 2020. [DOI: 10.26724/2079-8334-2020-3-73-164-168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
65
|
Silva LC, Fonseca FP, Almeida OP, Mariz BA, Lopes MA, Radhakrishnan R, Sharma M, Kowalski LP, Vargas PA. CD1a+ and CD207+ cells are reduced in oral submucous fibrosis and oral squamous cell carcinoma. Med Oral Patol Oral Cir Bucal 2020; 25:e49-e55. [PMID: 31880289 PMCID: PMC6982989 DOI: 10.4317/medoral.23177] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/16/2019] [Indexed: 12/26/2022] Open
Abstract
Background The objective of this study investigated the distribution of immature dendritic cells (DCs), Langerhans cells and plasmacytoid DCs in oral submucous fibrosis (OSMF), OSMF associated with oral squamous cell carcinoma (OSMF-OSCC), oral leukoplakia (OL), and oral squamous cell carcinoma (OSCC).
Material and Methods Fourteen cases of OSMF, 9 of OSMF-OSCC, 8 of OL¸ 45 of OSCC and 8 of normal epithelium were retrospectively retrieved and their diagnoses confirmed. Immunoreactions against CD1a, CD207 e CD303 were performed and the number of positive cells quantified.
Results A significant decrease of CD1a+ was found in OSMF (p≤0.05), OSMF-OSCC (p ≤ 0.01), and OSCC (p ≤ 0.001) when compared to normal epithelium. For CD207+ the significance decrease was observed in OSMF-OSCC (p ≤ 0.05), and OSCC (p ≤ 0.01) when compared with normal epithelium, and in OSMF when compared with OL (p ≤ 0.05). There was no significant difference for CD303, but increased in CD303+ was observed in OSCC when compared with normal epithelium.
Conclusions The decrease in the number of CD1a+ and CD207+ cells may be associate to the development of oral OSCC, and in OPMDs they might be indicators of malignant transformation. Key words:Premalignant lesions, oral submucous fibrosis, oral squamous cell carcinoma, immune response.
Collapse
Affiliation(s)
- L-C Silva
- Department of Oral Diagnosis - Oral Pathology Piracicaba Dental School, University of Campinas Av Limeira, 901 - 13414-016 Piracicaba, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Molero-Abraham M, Sanchez-Trincado JL, Gomez-Perosanz M, Torres-Gomez A, Subiza JL, Lafuente EM, Reche PA. Human Oral Epithelial Cells Impair Bacteria-Mediated Maturation of Dendritic Cells and Render T Cells Unresponsive to Stimulation. Front Immunol 2019; 10:1434. [PMID: 31316504 PMCID: PMC6611079 DOI: 10.3389/fimmu.2019.01434] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/07/2019] [Indexed: 01/03/2023] Open
Abstract
The oral mucosa is a first line of defense against pathogenic organisms and yet tolerates food antigens and resident bacteria. Mucosal epithelial cells are emerging as important regulators of innate and adaptive immune responses. However, the contribution of oral epithelial cells (OECs) determining oral immunity is understudied. Here, we evaluated the ability of H413 and TR146 cells, two OEC lines derived from human oral squamous cell carcinomas, and primary OECs to modulate immune responses to a cocktail of Gram+ and Gram− bacteria known as MV130. OECs expressed CD40 constitutively and class II major histocompatibility complex (MHC II) molecules when stimulated with IFNγ, but not CD80 or CD86. Dendritic cells (DCs) treated with bacteria in co-culture with OECs did not fully mature, as judged by the expression of MHC II, CD80 and CD86, and barely released IL-12 and TNFα, compared to control DCs. Furthermore, in the presence of OECs, DCs were unable to stimulate allogenic naive CD4 T cells to produce IFNγ and TNFα. Similarly, OECs in culture with total CD4 T cells or Th1 cells stimulated with anti-CD3 and anti-CD28 antibodies abrogated CD25 and CD69 expression, T cell proliferation and the release of IFNγ and TNFα. The inhibition on T cell activation by OECs was cell-contact dependent, TGFβ independent and largely irreversible. Overall, this behavior of OECs is likely key to avoid immune system over-reaction against resident bacteria.
Collapse
Affiliation(s)
| | - Jose L Sanchez-Trincado
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Marta Gomez-Perosanz
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Alvaro Torres-Gomez
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | - Esther M Lafuente
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Pedro A Reche
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
67
|
Tanaka Y, Fukumoto S, Sugawara S. Mechanisms underlying the induction of regulatory T cells by sublingual immunotherapy. J Oral Biosci 2019; 61:73-77. [DOI: 10.1016/j.job.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/04/2019] [Accepted: 02/15/2019] [Indexed: 12/30/2022]
|
68
|
Pandiyan P, Bhaskaran N, Zou M, Schneider E, Jayaraman S, Huehn J. Microbiome Dependent Regulation of T regs and Th17 Cells in Mucosa. Front Immunol 2019; 10:426. [PMID: 30906299 PMCID: PMC6419713 DOI: 10.3389/fimmu.2019.00426] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
Mammals co-exist with resident microbial ecosystem that is composed of an incredible number and diversity of bacteria, viruses and fungi. Owing to direct contact between resident microbes and mucosal surfaces, both parties are in continuous and complex interactions resulting in important functional consequences. These interactions govern immune homeostasis, host response to infection, vaccination and cancer, as well as predisposition to metabolic, inflammatory and neurological disorders. Here, we discuss recent studies on direct and indirect effects of resident microbiota on regulatory T cells (Tregs) and Th17 cells at the cellular and molecular level. We review mechanisms by which commensal microbes influence mucosa in the context of bioactive molecules derived from resident bacteria, immune senescence, chronic inflammation and cancer. Lastly, we discuss potential therapeutic applications of microbiota alterations and microbial derivatives, for improving resilience of mucosal immunity and combating immunopathology.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Natarajan Bhaskaran
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Elizabeth Schneider
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sangeetha Jayaraman
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
69
|
Jones AT, Shen X, Walter KL, LaBranche CC, Wyatt LS, Tomaras GD, Montefiori DC, Moss B, Barouch DH, Clements JD, Kozlowski PA, Varadarajan R, Amara RR. HIV-1 vaccination by needle-free oral injection induces strong mucosal immunity and protects against SHIV challenge. Nat Commun 2019; 10:798. [PMID: 30778066 PMCID: PMC6379385 DOI: 10.1038/s41467-019-08739-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/25/2019] [Indexed: 02/08/2023] Open
Abstract
The oral mucosa is an attractive site for mucosal vaccination, however the thick squamous epithelium limits antigen uptake. Here we utilize a modified needle-free injector to deliver immunizations to the sublingual and buccal (SL/B) tissue of rhesus macaques. Needle-free SL/B vaccination with modified vaccinia Ankara (MVA) and a recombinant trimeric gp120 protein generates strong vaccine-specific IgG responses in serum as well as vaginal, rectal and salivary secretions. Vaccine-induced IgG responses show a remarkable breadth against gp70-V1V2 sequences from multiple clades of HIV-1. In contrast, topical SL/B immunizations generates minimal IgG responses. Following six intrarectal pathogenic SHIV-SF162P3 challenges, needle-free but not topical immunization results in a significant delay of acquisition of infection. Delay of infection correlates with non-neutralizing antibody effector function, Env-specific CD4+ T-cell responses, and gp120 V2 loop specific antibodies. These results demonstrate needle-free MVA/gp120 oral vaccination as a practical and effective route to induce protective immunity against HIV-1.
Collapse
Affiliation(s)
- Andrew T Jones
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, Georgia, 30329, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710, USA
| | - Korey L Walter
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Celia C LaBranche
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Linda S Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710, USA
| | - David C Montefiori
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - John D Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 8638, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Raghavan Varadarajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Rama Rao Amara
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA.
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, Georgia, 30329, USA.
| |
Collapse
|
70
|
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
|
71
|
Elewa YHA, Mizoguchi T, Ichii O, Nakamura T, Kon Y. Morphofunctional analysis of antigen uptake mechanisms following sublingual immunotherapy with beads in mice. PLoS One 2018; 13:e0201330. [PMID: 30571699 PMCID: PMC6301667 DOI: 10.1371/journal.pone.0201330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/03/2018] [Indexed: 11/19/2022] Open
Abstract
Background Recently, sublingual immunotherapy (SLIT) has been used as a safe and efficient method for the treatment of and immunization against asthma and various allergies. However, the routes of antigen/allergen (particulate antigen) uptake through the mucosa of the oral cavity remain incompletely understood, as do the roles of sex and age in the process. For this purpose, to elucidate the mechanism and efficacy of SLIT among different sexes and ages, microbeads were dripped into the sublingual region to mimic particulate antigen uptake by the sublingual mucosa. Methods Twenty microliters of either phosphate buffered saline (PBS) or fluorescently labelled microbeads (latex and silica beads) were placed under the tongue of both male and female C57BL/6 mice at young (3 months) and old (6 months) ages. The lower jaw was examined 30 min after administration, and beads were detected with a fluorescence stereomicroscope. Morphological observations of the mucosa of the fluorescent areas were made with a scanning electron microscope (SEM) and an all-in-one light fluorescence microscope (LM). Fluorescence intensity was compared between both sexes and ages. Results Stereomicroscopic observation revealed fluorescent illuminations in three compartments of the sublingual mucosa: the sublingual caruncles (SC), the oral rostral mucosa (OR) and the buccal mucosa (BM). Interestingly, the fluorescence intensity tended to be higher among females than among males in the SC region in particular. However, there were no significant age-related differences. SEM and LM revealed beads in the lumina of both mandibular ducts and sublingual ducts (Sd). Additionally, the apical cytoplasm of some Sd cells contained silica beads. However, there was no specification in the OR mucosa or BM. Conclusions This study reveals the major role Sd plays in local immunity via the antigen uptake mechanisms. Furthermore, our data suggest that the efficacy of SLIT in humans could be affected by sex.
Collapse
Affiliation(s)
- Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Faculty of Veterinary Medicine, Basic Veterinary Sciences, Laboratory of Anatomy, Hokkaido University, Sapporo, Japan
- * E-mail: ,
| | - Tatsuya Mizoguchi
- Faculty of Veterinary Medicine, Basic Veterinary Sciences, Laboratory of Anatomy, Hokkaido University, Sapporo, Japan
| | - Osamu Ichii
- Faculty of Veterinary Medicine, Basic Veterinary Sciences, Laboratory of Anatomy, Hokkaido University, Sapporo, Japan
| | - Teppei Nakamura
- Faculty of Veterinary Medicine, Basic Veterinary Sciences, Laboratory of Anatomy, Hokkaido University, Sapporo, Japan
- Section of Biological Science, Chitose Laboratory, Japan Food Research Laboratories, Chitose, Japan
| | - Yasuhiro Kon
- Faculty of Veterinary Medicine, Basic Veterinary Sciences, Laboratory of Anatomy, Hokkaido University, Sapporo, Japan
| |
Collapse
|
72
|
Ahsan H. Biomolecules and biomarkers in oral cavity: bioassays and immunopathology. J Immunoassay Immunochem 2018; 40:52-69. [DOI: 10.1080/15321819.2018.1550423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Haseeb Ahsan
- Department of Biochemistry, Faculty of Dentistry, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
73
|
Sobkowiak MJ, Davanian H, Heymann R, Gibbs A, Emgård J, Dias J, Aleman S, Krüger-Weiner C, Moll M, Tjernlund A, Leeansyah E, Sällberg Chen M, Sandberg JK. Tissue-resident MAIT cell populations in human oral mucosa exhibit an activated profile and produce IL-17. Eur J Immunol 2018; 49:133-143. [PMID: 30372518 PMCID: PMC6519349 DOI: 10.1002/eji.201847759] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/21/2018] [Accepted: 10/26/2018] [Indexed: 12/31/2022]
Abstract
Mucosa‐associated invariant T (MAIT) cells are unconventional T lymphocytes defined by their innate‐like characteristics and broad antimicrobial responsiveness. Whether MAIT cells are part of the tissue‐resident defense in the oral mucosal barrier is unknown. Here, we found MAIT cells present in the buccal mucosa, with a tendency to cluster near the basement membrane, and located in both epithelium and the underlying connective tissue. Overall MAIT cell levels were similar in the mucosa compared to peripheral blood, in contrast to conventional T cells that showed an altered representation of CD4+ and CD8+ subsets. The major mucosal MAIT cell subset displayed a tissue‐resident and activated profile with high expression of CD69, CD103, HLA‐DR, and PD‐1, as well as a skewed subset distribution with higher representation of CD4–/CD8– double‐negative cells and CD8αα+ cells. Interestingly, tissue‐resident MAIT cells had a specialized polyfunctional response profile with higher IL‐17 levels, as assessed by polyclonal stimulus and compared to tissue nonresident and circulating populations. Furthermore, resident buccal MAIT cells were low in perforin. Together, these data indicate that MAIT cells form a part of the oral mucosal T cell compartment, where they exhibit a tissue‐resident‐activated profile biased toward IL‐17 production.
Collapse
Affiliation(s)
- Michał J Sobkowiak
- Center for Infection Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Haleh Davanian
- Division of Clinical Diagnostics and Surgery, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert Heymann
- Division of Clinical Diagnostics and Surgery, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden.,Trauma and Reparative Medicine, PO Craniofacial diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Gibbs
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Johanna Emgård
- Center for Infection Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Joana Dias
- Center for Infection Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Soo Aleman
- Division of Infectious Diseases, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Carina Krüger-Weiner
- Division of Clinical Diagnostics and Surgery, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Markus Moll
- Center for Infection Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Annelie Tjernlund
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Edwin Leeansyah
- Center for Infection Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Margaret Sällberg Chen
- Division of Clinical Diagnostics and Surgery, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan K Sandberg
- Center for Infection Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
74
|
Heyman O, Koren N, Mizraji G, Capucha T, Wald S, Nassar M, Tabib Y, Shapira L, Hovav AH, Wilensky A. Impaired Differentiation of Langerhans Cells in the Murine Oral Epithelium Adjacent to Titanium Dental Implants. Front Immunol 2018; 9:1712. [PMID: 30158922 PMCID: PMC6103475 DOI: 10.3389/fimmu.2018.01712] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/12/2018] [Indexed: 11/13/2022] Open
Abstract
Peri-implantitis is a destructive inflammatory process affecting tissues surrounding dental implants and it is considered a new global health concern. Human studies have suggested that the frequencies of Langerhans cells (LCs), the main antigen-presenting cells (APCs) of the oral epithelium, are dysregulated around the implants. Since LCs play a role in regulating oral mucosal homeostasis, we studied the impact of dental titanium implants on LC differentiation using a novel murine model. We demonstrate that whereas the percentage of LC precursors (CD11c+MHCII+) increased in the peri-implant epithelium, the frequencies of LCs (CD11c+MHCII+EpCAM+langerin+) were significantly reduced. Instead, a population of partially developed LCs expressing CD11c+MHCII+EpCAM+ but not langerin evolved in the peri-implant mucosa, which was also accompanied by a considerable leukocyte infiltrate. In line with the increased levels of LC precursors, expression of CCL2 and CCL20, chemokines mediating their translocation to the epithelium, was elevated in the peri-implant epithelium. However, expression of TGF-β1, the major cytokine driving final differentiation of LCs, was reduced in the epithelium. Further analysis revealed that while the expression of the TGF-β1 canonical receptor activing-like kinase (ALK)5 was upregulated, expression of its non-canonical receptor ALK3 was decreased. Since titanium ions releasing from implants were proposed to alter APC function, we next analyzed the impact of such ions on TGF-β1-induced LC differentiation cultures. Concurring with the in vivo studies, the presence of titanium ions resulted in the generation of partially developed LCs that express CD11c+MHCII+EpCAM+ but failed to upregulate langerin expression. Collectively, these findings suggest that titanium dental implants have the capacity to impair the development of oral LCs and might subsequently dysregulate immunity in the peri-implant mucosa.
Collapse
Affiliation(s)
- Oded Heyman
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Noam Koren
- Faculty of Dental Medicine, The Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Gabriel Mizraji
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel.,Faculty of Dental Medicine, The Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Tal Capucha
- Faculty of Dental Medicine, The Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Sharon Wald
- Faculty of Dental Medicine, The Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Maria Nassar
- Faculty of Dental Medicine, The Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Yaara Tabib
- Faculty of Dental Medicine, The Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Lior Shapira
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Avi-Hai Hovav
- Faculty of Dental Medicine, The Institute of Dental Sciences, Hebrew University, Jerusalem, Israel
| | - Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
75
|
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
|
76
|
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
|
77
|
Soria I, López-Relaño J, Viñuela M, Tudela JI, Angelina A, Benito-Villalvilla C, Díez-Rivero CM, Cases B, Manzano AI, Fernández-Caldas E, Casanovas M, Palomares O, Subiza JL. Oral myeloid cells uptake allergoids coupled to mannan driving Th1/Treg responses upon sublingual delivery in mice. Allergy 2018; 73:875-884. [PMID: 29319882 PMCID: PMC5947296 DOI: 10.1111/all.13396] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Polymerized allergoids coupled to nonoxidized mannan (PM-allergoids) may represent novel vaccines targeting dendritic cells (DCs). PM-allergoids are better captured by DCs than native allergens and favor Th1/Treg cell responses upon subcutaneous injection. Herein we have studied in mice the in vivo immunogenicity of PM-allergoids administered sublingually in comparison with native allergens. METHODS Three immunization protocols (4-8 weeks long) were used in Balb/c mice. Serum antibody levels were tested by ELISA. Cell responses (proliferation, cytokines, and Tregs) were assayed by flow cytometry in spleen and lymph nodes (LNs). Allergen uptake was measured by flow cytometry in myeloid sublingual cells. RESULTS A quick antibody response and higher IgG2a/IgE ratio were observed with PM-allergoids. Moreover, stronger specific proliferative responses were seen in both submandibular LNs and spleen cells assayed in vitro. This was accompanied by a higher IFNγ/IL-4 ratio with a quick IL-10 production by submandibular LN cells. An increase in CD4+ CD25high FOXP3+ Treg cells was detected in LNs and spleen of mice treated with PM-allergoids. These allergoids were better captured than native allergens by antigen-presenting (CD45+ MHC-II+ ) cells obtained from the sublingual mucosa, including DCs (CD11b+ ) and macrophages (CD64+ ). Importantly, all the differential effects induced by PM-allergoids were abolished when using oxidized instead of nonoxidized PM-allergoids. CONCLUSION Our results demonstrate for the first time that PM-allergoids administered through the sublingual route promote the generation of Th1 and FOXP3+ Treg cells in a greater extent than native allergens by mechanisms that might well involve their better uptake by oral antigen-presenting cells.
Collapse
Affiliation(s)
- I. Soria
- Inmunotek; Alcalá de Henares Spain
| | - J. López-Relaño
- Inmunotek; Alcalá de Henares Spain
- Immunology-Experimental Unit; Hospital Clínico Universitario San Carlos; Madrid Spain
- Department of Immunology; School of Medicine; Complutense University of Madrid; Madrid Spain
| | - M. Viñuela
- Immunology-Experimental Unit; Hospital Clínico Universitario San Carlos; Madrid Spain
| | | | - A. Angelina
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | - C. Benito-Villalvilla
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | | | - B. Cases
- Inmunotek; Alcalá de Henares Spain
| | | | | | | | - O. Palomares
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | | |
Collapse
|
78
|
Wang Y, Shang S, Sun Q, Chen J, Du G, Nie H, Han X, Tang G. Increased infiltration of CD11 c +/CD123 + dendritic cell subsets and upregulation of TLR/IFN-α signaling participate in pathogenesis of oral lichen planus. Oral Surg Oral Med Oral Pathol Oral Radiol 2017; 125:459-467.e2. [PMID: 29429903 DOI: 10.1016/j.oooo.2017.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 11/22/2017] [Accepted: 12/03/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Investigation of dendritic cell (DC) subsets and expression patterns of Toll-like receptors (TLRs) was conducted to understand the pathogenesis in oral lichen planus (OLP). STUDY DESIGN Blood, OLP lesion, and control samples were collected. Four DC subsets (CD11c+CD123-myeloid DC1 [mDC1], CD141+mDC2, CD11c-CD123+plasmacytoid DC [pDC], and CD1a+CD207+Langerhans cells [LC]) were investigated via flow cytometry (FCM) and immunohistochemical staining. Expression patterns of TLRs and their downstream molecules were analyzed via quantitative real-time polymerase chain reaction and immunohistochemistry in situ. RESULTS Thirty-two samples were collected (9 controls and 23 OLP patients). FCM results found that the percentages of LC, mDC1, mDC2, and pDC in situ were 0.0119 ± 0.0251%, 0.0064 ± 0.0134%, 0.0005 ± 0.0011%, and 0.0022 ± 0.0019% in control mucosa, respectively. The mDC1 (0.0300 ± 0.0276%) and pDC (0.0204 ± 0.0186%) subsets were significantly increased in OLP lesions (P < .01). No marked differences were evident, when comparing all 4 DC subsets from blood, between control and OLP groups. Significant upregulation of TLR7, TLR8, and TLR9 were disclosed in OLP (P < .01), along with their downstream interferon-α (IFN-α) signaling molecules (IRF7 and IFN-α, P < .01). CONCLUSION Our findings of increased infiltration of pDC and mDC1, along with upregulation of TLR/IFN-α signaling, provide valuable information for further understanding the immunity in OLP.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Oral Mucosal Diseases, Ninth People's Hospital, College of Stomatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China; National Clinical Research Center of Stomatology, Shanghai, China
| | - Shu Shang
- Shanghai University of Medicine and Health Science, Shanghai, China
| | - Qianqian Sun
- Department of Oral Mucosal Diseases, Ninth People's Hospital, College of Stomatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China; National Clinical Research Center of Stomatology, Shanghai, China
| | - Junjun Chen
- Department of Oral Mucosal Diseases, Ninth People's Hospital, College of Stomatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China; National Clinical Research Center of Stomatology, Shanghai, China
| | - Guanhuan Du
- Department of Oral Mucosal Diseases, Ninth People's Hospital, College of Stomatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China; National Clinical Research Center of Stomatology, Shanghai, China
| | - Hong Nie
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaozhe Han
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, USA; Harvard School of Dental Medicine, Department of Oral Medicine, Infection and Immunity, Boston, MA, USA
| | - Guoyao Tang
- Department of Oral Mucosal Diseases, Ninth People's Hospital, College of Stomatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China; National Clinical Research Center of Stomatology, Shanghai, China.
| |
Collapse
|
79
|
Fakhry C, Qeadan F, Gilman RH, Yori P, Kosek M, Patterson N, Eisele DW, Gourin CG, Chitguppi C, Marks M, Gravitt P. Oral sampling methods are associated with differences in immune marker concentrations. Laryngoscope 2017; 128:E214-E221. [PMID: 29171655 DOI: 10.1002/lary.27002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/13/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To determine whether the concentration and distribution of immune markers in paired oral samples were similar. STUDY TYPE Clinical research. STUDY DESIGN Cross-sectional study. METHODS Paired saliva and oral secretions (OS) samples were collected. The concentration of immune markers was estimated using Luminex multiplex assay (Thermo Fisher Scientific, Waltham, MA). For each sample, the concentration of respective immune markers was normalized to total protein present and log-transformed. Median concentrations of immune markers were compared between both types of samples. Intermarker correlation in each sampling method and across sampling methods was evaluated. RESULTS There were 90 study participants. Concentrations of immune markers in saliva samples were significantly different from concentrations in OS samples. Oral secretions samples showed higher concentrations of immunoregulatory markers, whereas the saliva samples contained proinflammatory markers in higher concentration. CONCLUSION The immune marker profile in saliva samples is distinct from the immune marker profile in paired OS samples. LEVEL OF EVIDENCE 2b. Laryngoscope, 128:E214-E221, 2018.
Collapse
Affiliation(s)
- Carole Fakhry
- Department of Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Fares Qeadan
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Robert H Gilman
- Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pablo Yori
- Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Margaret Kosek
- Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - David W Eisele
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine G Gourin
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Morgan Marks
- Johns Hopkins Hospital and Health System, Baltimore, Maryland
| | - Patti Gravitt
- Milken Institute of Public Health, George Washington University, Washington, District of Columbia, U.S.A
| |
Collapse
|
80
|
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
|
81
|
Retamozo S, Flores-Chavez A, Consuegra-Fernández M, Lozano F, Ramos-Casals M, Brito-Zerón P. Cytokines as therapeutic targets in primary Sjögren syndrome. Pharmacol Ther 2017; 184:81-97. [PMID: 29092775 DOI: 10.1016/j.pharmthera.2017.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Primary Sjögren syndrome (SjS) is a systemic autoimmune disease that may affect 1 in 1000 people (overwhelmingly women) and that can be a serious disease with excess mortality due to severe organ-specific involvements and the development of B cell lymphoma; systemic involvement clearly marks the disease prognosis, and strongly suggests the need for closer follow-up and more robust therapeutic management. Therapy is established according to the organ involved and severity. As a rule, the management of systemic SjS should be organ-specific, with glucocorticoids and immunosuppressive agents limited to potentially-severe involvements; unfortunately, the limited evidence available for these drugs, together with the potential development of serious adverse events, makes solid therapeutic recommendations difficult. The emergence of biological therapies has increased the therapeutic armamentarium available to treat primary SjS. Biologics currently used in SjS patients are used off-label and are overwhelmingly agents targeting B cells, but the most recent studies are moving on into the evaluation of targeting specific cytokines involved in the SjS pathogenesis. The most recent etiopathogenic advances in SjS are shedding some light in the search for new highly-selective biological therapies without the adverse effects of the standard drugs currently used (corticosteroids and immunosuppressant drugs). This review summarizes the potential pharmacotherapeutic options targeting the main cytokine families involved in the etiopathogenesis of primary SjS and analyzes potential insights for developing new therapies.
Collapse
Affiliation(s)
- Soledad Retamozo
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Hospital Privado Universitario de Córdoba, Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina; Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Córdoba, Consejo Nacional de Investigaciones Científicas y Técnicas (INICSA-UNC-CONICET), Córdoba, Argentina; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| | - Alejandra Flores-Chavez
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Biomedical Research Unit 02, Clinical Epidemiology Research Unit, UMAE, Specialties Hospital, Western Medical Center, Mexican Institute for Social Security (IMSS), Guadalajara, Mexico; Postgraduate Program of Medical Science, University Center for Biomedical Research (CUIB), University of Colima, Colima, Mexico; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| | - Marta Consuegra-Fernández
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Servei d'Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain; Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.
| | - Manuel Ramos-Casals
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain.
| | - Pilar Brito-Zerón
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA-Sanitas, Barcelona, Spain; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| |
Collapse
|
82
|
Marcussen M, Skrubbeltrang C, Bødker JS, Christiansen I, Bøgsted M, Dybkær K, Bergmann OJ, Johnsen HE. A systematic review of molecular responses to cancer therapy in normal human mucosa. Oral Surg Oral Med Oral Pathol Oral Radiol 2017; 124:355-366. [PMID: 29042035 DOI: 10.1016/j.oooo.2017.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 07/24/2017] [Accepted: 08/01/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Cancer therapy-induced inflammation of oral and gastrointestinal mucosae affects patients nonuniformly. Preventive strategies are limited; no biomarker exists for pretreatment identification of patients likely to be severely affected. Animal models are preferred for studying molecular responses in mucosae during chemotherapy, but translation into clinical practice is difficult. We performed a systematic review to retrieve articles that described molecular changes in human mucosae during cancer therapy. STUDY DESIGN We searched MEDLINE and Ovid Embase searches for studies reported in the English language literature from January 1990 to November 2016 and studies referenced in selected articles, which analyzed mucosae from patients at risk of developing mucositis during cancer therapy. Two authors extracted data according to predefined data fields, including study quality indicators. RESULTS We identified 17 human studies on chemotherapy (n = 9) and radiotherapy (n = 8), but no studies on targeted therapy. Studies were heterogeneous with regard to patient cohorts, analysis methods, cancer treatments, biopsy timings, and correlations to clinical mucositis. Consequently, a meta-analysis was not feasible. CONCLUSIONS Few human studies described the molecular responses of the normal mucosa to cancer therapy. Studies were heterogeneous and had sparse correlations to clinical mucositis. We proposed a model for acquiring data on treatment- and disease-specific phenotypes and transcriptomes for predictive or preventive initiatives.
Collapse
Affiliation(s)
- Mette Marcussen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | | | - Julie Støve Bødker
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Ilse Christiansen
- Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | - Karen Dybkær
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | - Olav Jonas Bergmann
- School of Dentistry, Faculty of Health Science, Aarhus University; Aarhus, Denmark
| | - Hans Erik Johnsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
83
|
Tissue-Specific Immunity at the Oral Mucosal Barrier. Trends Immunol 2017; 39:276-287. [PMID: 28923364 DOI: 10.1016/j.it.2017.08.005] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/05/2017] [Accepted: 08/11/2017] [Indexed: 02/07/2023]
Abstract
The oral mucosal barrier is constantly exposed to a plethora of triggers requiring immune control, including a diverse commensal microbiome, ongoing damage from mastication, and dietary and airborne antigens. However, how these tissue-specific cues participate in the training of immune responsiveness at this site is minimally understood. Moreover, the mechanisms mediating homeostatic immunity at this interface are not yet fully defined. Here we present basic aspects of the oral mucosal barrier and discuss local cues that may modulate and train local immune responsiveness. We particularly focus on the immune cell network mediating immune surveillance at a specific oral barrier, the gingiva - a constantly stimulated and dynamic environment where homeostasis is often disrupted, resulting in the common inflammatory disease periodontitis.
Collapse
|
84
|
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
|
85
|
Free immunoglobulin light chain (FLC) promotes murine colitis and colitis-associated colon carcinogenesis by activating the inflammasome. Sci Rep 2017; 7:5165. [PMID: 28701727 PMCID: PMC5507933 DOI: 10.1038/s41598-017-05468-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 05/31/2017] [Indexed: 01/10/2023] Open
Abstract
Numerous studies have demonstrated that free Ig light chain (FLC), a novel inflammation mediator, participates in many inflammatory diseases by activating mast cells and extending the survival of neutrophils. However, it remains unclear whether FLC is involved in colitis and colitis-associated colon carcinogenesis (CAC). In this study, we found a significant increase in FLC in murine models of DSS (Dextran Sulfate Sodium Salt)-induced colitis and CAC compared to controls. Peptide F991, a functional blocker of FLC, significantly attenuated colitis progression, which included abrogating the development of diarrhea and tumor burden, elevating survival rate, greatly reducing the infiltration of inflammatory cells (such as ROS+ active neutrophils), especially reducing tumorigenesis in CAC. Furthermore, we demonstrated that F991 inhibited the activation of the inflammasome by reducing the expression of cleaved caspase-1 and the maturation of IL-1β and IL-18. Altogether, our findings demonstrate that FLC can promote the pathogenesis of colitis and CAC and may be used as novel biomarker for the diagnosis of inflammatory bowel disease. Additionally, F991 may become a potential therapeutic option for colitis or colorectal cancer.
Collapse
|
86
|
Sichien D, Lambrecht BN, Guilliams M, Scott CL. Development of conventional dendritic cells: from common bone marrow progenitors to multiple subsets in peripheral tissues. Mucosal Immunol 2017; 10:831-844. [PMID: 28198365 DOI: 10.1038/mi.2017.8] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/14/2017] [Indexed: 02/04/2023]
Abstract
Our understanding of conventional dendritic cell (cDC) development and the functional specializations of distinct subsets in the peripheral tissues has increased greatly in recent years. Here, we review cDC development from the distinct progenitors in the bone marrow through to the distinct cDC subsets found in barrier tissues, providing an overview of the different subsets described in each location. In addition, we detail the transcription factors and local signals that have been proposed to control this developmental process. Importantly, despite these significant advances, numerous questions remain to be answered regarding cDC development. For example, it remains unclear whether the different subsets described, such as the CD103+CD11b+ and CD103-CD11b+ cDCs in the intestines, truly represent different populations or rather distinct developmental or activation stages. Furthermore, whether distinct progenitors exist for these cDC subsets remains to be determined. Thus in the last part of this review we discuss what we believe will be the main questions facing the field for the coming years.
Collapse
Affiliation(s)
- D Sichien
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - B N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, The Netherlands
| | - M Guilliams
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - C L Scott
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
87
|
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that play a pivotal role in the pathogenesis of periodontitis. The use of animal models to study the role of DCs in periodontitis has been limited by lack of a method for sustained depletion of DCs. Hence, the objectives of this study were to validate the zDC-DTR knockin mouse model of conventional DCs (cDCs) depletion, as well as to investigate whether this depletion could be sustained long enough to induce alveolar bone loss in this model. zDC-DTR mice were treated with different dose regimens of diphtheria toxin (DT) to determine survival rate. A loading DT dose of 20ng/bw, followed and maintained with doses of 10ng/bm every 3days for up to 4weeks demonstrated 80% survival. Animals were weighed weekly and peripheral blood was obtained to confirm normal neutrophil counts. Five animals per group were euthanized at baseline, 24h, 1 and 4weeks. Bone marrow (BM), spleen (SP) and gingival tissue (GT) were harvested, and cells were isolated, separated and stained for Pre-DCs precursors (CD45R-MHCII+CD11c+Flt3+CD172a+) in BM, cDCs (CD11c+MHCII+CD209+) in spleen, and DCs in GT (CD45R+MHCII+CD11c+ DC-SIGN/CD209+). Pre-DCs in BM were significantly depleted at 24h and depletion maintained for up to 4weeks, as compared to blank (PBS) controls. Circulating cDCs in spleen demonstrated a non-significant trend to deplete in 1week with high variability among mice. GT also showed a similar non-significant trend to deplete in 24h. The zDC-DTR model seems to be viable for evaluating the role of DCs immune homeostasis disruption and alveolar bone loss pathogenesis in response to long-term oral infection.
Collapse
|
88
|
Gutowska-Owsiak D, Ogg GS. Therapeutic vaccines for allergic disease. NPJ Vaccines 2017; 2:12. [PMID: 29263869 PMCID: PMC5604746 DOI: 10.1038/s41541-017-0014-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/03/2017] [Accepted: 03/20/2017] [Indexed: 12/14/2022] Open
Abstract
Allergic diseases are highly prevalent worldwide and affect all age groups, contributing to a high personal and socioeconomic burden. Treatment with an “allergy vaccine” or allergen immunotherapy aims to provide long-lasting benefits by inducing unresponsiveness to the relevant antigen. The consequences of the therapy are considered disease modifying and range from dampening of the immediate immune responses to the reduction of secondary tissue remodeling. Furthermore, allergen immunotherapy interventions have a potential to slow or cease the development of additional allergic manifestations with a long-term overall effect on morbidity and quality of life. Here, we review proposed mechanisms underlying the therapeutic effects of immunotherapy for allergic diseases. Further, we discuss both standard and novel approaches and possible future directions in the development of allergen immunotherapy.
Collapse
Affiliation(s)
- Danuta Gutowska-Owsiak
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Graham S Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
89
|
Kelsen JR, Albenberg L. Does Poor Oral Health Protect Against Inflammatory Bowel Disease? Clin Gastroenterol Hepatol 2017; 15:532-534. [PMID: 28043930 DOI: 10.1016/j.cgh.2016.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023]
Affiliation(s)
- Judith R Kelsen
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Lindsey Albenberg
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
90
|
Aran K, Chooljian M, Paredes J, Rafi M, Lee K, Kim AY, An J, Yau JF, Chum H, Conboy I, Murthy N, Liepmann D. An oral microjet vaccination system elicits antibody production in rabbits. Sci Transl Med 2017; 9:eaaf6413. [PMID: 28275153 DOI: 10.1126/scitranslmed.aaf6413] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 08/16/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
Noninvasive immunization technologies have the potential to revolutionize global health by providing easy-to-administer vaccines at low cost, enabling mass immunizations during pandemics. Existing technologies such as transdermal microneedles are costly, deliver drugs slowly, and cannot generate mucosal immunity, which is important for optimal immunity against pathogens. We present a needle-free microjet immunization device termed MucoJet, which is a three-dimensional microelectromechanical systems-based drug delivery technology. MucoJet is administered orally, placed adjacent to the buccal tissue within the oral cavity, and uses a self-contained gas-generating chemical reaction within its two-compartment plastic housing to produce a high-pressure liquid jet of vaccine. We show that the vaccine jet ejected from the MucoJet device is capable of penetrating the buccal mucosal layer in silico, in porcine buccal tissue ex vivo, and in rabbits in vivo. Rabbits treated with ovalbumin by MucoJet delivery have antibody titers of anti-ovalbumin immunoglobulins G and A in blood serum and buccal tissue, respectively, that are three orders of magnitude higher than rabbits receiving free ovalbumin delivered topically by a dropper in the buccal region. MucoJet has the potential to accelerate the development of noninvasive oral vaccines, given its ability to elicit antibody production that is detectable locally in the buccal tissue and systemically via the circulation.
Collapse
Affiliation(s)
- Kiana Aran
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA 91711, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Marc Chooljian
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| | - Jacobo Paredes
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
- Center of Studies and Technical Research of Gipuzkoa and Tecnun (Technological Campus of the University of Navarra), 20018 San Sebastián, Spain
| | - Mohammad Rafi
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Kunwoo Lee
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
| | - Allison Y Kim
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Jeanny An
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Jennifer F Yau
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Helen Chum
- Office of Laboratory Animal Care, UC Berkeley, Berkeley, CA 94720, USA
| | - Irina Conboy
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Niren Murthy
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| | - Dorian Liepmann
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| |
Collapse
|
91
|
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
|
92
|
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
|
93
|
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
|
94
|
Mašek J, Lubasová D, Lukáč R, Turánek-Knotigová P, Kulich P, Plocková J, Mašková E, Procházka L, Koudelka Š, Sasithorn N, Gombos J, Bartheldyová E, Hubatka F, Raška M, Miller AD, Turánek J. Multi-layered nanofibrous mucoadhesive films for buccal and sublingual administration of drug-delivery and vaccination nanoparticles - important step towards effective mucosal vaccines. J Control Release 2017; 249:183-195. [DOI: 10.1016/j.jconrel.2016.07.036] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/22/2016] [Accepted: 07/23/2016] [Indexed: 12/12/2022]
|
95
|
Wilensky A, Mizraji G, Tabib Y, Sharawi H, Hovav AH. Analysis of Leukocytes in Oral Mucosal Tissues. Methods Mol Biol 2017; 1559:267-278. [PMID: 28063050 DOI: 10.1007/978-1-4939-6786-5_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The oral mucosa is constantly exposed to an immense amount of microorganisms, while some colonize the various anatomical niches existing in the oral cavity. To deal with such a complex challenge, the oral mucosal immune system must tolerate commensal microorganisms but prevent invasion of pathogens. Such activity is likely to be achieved by a wide range of mechanisms that could be similar or different to those employed by other mucosal tissues. The dental biofilm represents a unique challenge to the mucosal immune system, and inadequate immune responses might lead to periodontal diseases and the associated adverse systemic complications. It is thus crucial to study the mechanisms by which the oral mucosal immune system maintains homeostasis, and also induces protective immune responses against pathogens. To facilitate probing oral mucosal immunity, we describe here methods allowing immunological analysis of murine oral tissues using flow cytometry and immunofluorescence techniques.
Collapse
Affiliation(s)
- Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Gabriel Mizraji
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel.,Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, 12272, Jerusalem, 91120, Israel
| | - Yaara Tabib
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, 12272, Jerusalem, 91120, Israel
| | - Hafez Sharawi
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, 12272, Jerusalem, 91120, Israel
| | - Avi-Hai Hovav
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, 12272, Jerusalem, 91120, Israel.
| |
Collapse
|
96
|
Straka M, Polák Š, Straková Trapezanlidis M, Varga I. What we know about the cellular microenvironment of clinically healthy human gingiva? An immunohistochemical and histological study. Biologia (Bratisl) 2017. [DOI: 10.1515/biolog-2017-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
97
|
Oral CD103 -CD11b + classical dendritic cells present sublingual antigen and induce Foxp3 + regulatory T cells in draining lymph nodes. Mucosal Immunol 2017; 10:79-90. [PMID: 27166558 DOI: 10.1038/mi.2016.46] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 04/07/2016] [Indexed: 02/04/2023]
Abstract
Sublingual immunotherapy (SLIT) is a safe and efficient treatment for type 1 allergies; however, the underlying immunological mechanisms, particularly the phenotype of oral antigen-presenting cells (APCs) responsible for the induction of regulatory T (Treg) cells, remain unclear. We show here that the sublingual application of ovalbumin (OVA) induced antigen-specific Foxp3+ Treg cells in draining submandibular lymph nodes (ManLNs). Oral APCs were classified into macrophages, classical dendritic cells (cDCs), and Langerhans cells by flow cytometry. A major subset of oral cDCs with the CD103-CD11b+ phenotype showed retinoic acid (RA)-producing activity and converted naive CD4+ T cells to Foxp3+ Treg cells in a transforming growth factor-β- and RA-dependent manner in vitro. In the ManLNs, migratory CD103-CD11b+ cDCs also showed RA-producing activity. After the sublingual application of fluorescent OVA, fluorescence was detected in oral macrophages in tissues, followed by migratory CD103-CD11b+ cDCs in ManLNs and migratory CD103-CD11b+ cDCs were the main APCs responsible for the induction of sublingual antigen-specific Treg cells. The transfer of OVA-SLIT-induced Treg cells suppressed the OVA-induced hypersensitivity response. These results suggest that oral CD103-CD11b+ cDCs transport sublingual antigens to draining ManLNs and induce antigen-specific Foxp3+ Treg cells, and, thus, provide a rationale for developing cDC-based therapeutic approaches in SLIT.
Collapse
|
98
|
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
|
99
|
Gorelik M, Frischmeyer-Guerrerio PA. Innate and adaptive dendritic cell responses to immunotherapy. Curr Opin Allergy Clin Immunol 2016; 15:575-80. [PMID: 26509662 DOI: 10.1097/aci.0000000000000213] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW In allergic disease, dendritic cells play a critical role in orchestrating immune responses to innate stimuli and promoting the formation of T helper 2 (TH2) effector versus T-regulatory cells. Here, we review recent advances in our understanding of how current forms of immunotherapy modulate dendritic cell responses. (Figure is included in full-text article.) RECENT FINDINGS Sublingual immunotherapy (SLIT) and oral immunotherapy (OIT) for peanut allergy alter the expression of costimulatory molecules on dendritic cells, which leads to reduced expression of TH2 effector cytokines in an antigen-nonspecific manner. SLIT and OIT also modulate dendritic cell innate immune responses to Toll-like receptor agonists, including enhanced production of interferon α and reduced expression of proinflammatory cytokines that may serve to promote the development of tolerance. Dendritic cells isolated from patients post-OIT promoted hypomethylation of the FOXP3 locus in effector T cells. Reduced methylation of the FOXP3 locus has been associated with more persistent clinical desensitization following OIT. Recent studies have additionally highlighted a role for B cells in inducing tolerogenic dendritic cell populations and T-regulatory cells during immunotherapy. Epicutaneous immunotherapy may also elicit immunosuppressive populations of cutaneous dendritic cells, although in some cases, antigen exposure through the skin can lead to sensitization. Finally, efforts have focused on identifying pharmacologic and/or antigen-independent strategies of altering dendritic cell function to enhance the immunosuppressive effects of immunotherapy. SUMMARY Dendritic cells are a critical target of immunotherapy. Alterations in both adaptive and innate immunity likely underlie the immunosuppressive effects of this treatment.
Collapse
Affiliation(s)
- Mark Gorelik
- aDepartment of Pediatrics, Division of Allergy and Immunology, Johns Hopkins University School of Medicine, Baltimore bLaboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | | |
Collapse
|
100
|
Bittner-Eddy PD, Fischer LA, Kaplan DH, Thieu K, Costalonga M. Mucosal Langerhans Cells Promote Differentiation of Th17 Cells in a Murine Model of Periodontitis but Are Not Required for Porphyromonas gingivalis-Driven Alveolar Bone Destruction. THE JOURNAL OF IMMUNOLOGY 2016; 197:1435-46. [PMID: 27402698 DOI: 10.4049/jimmunol.1502693] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/07/2016] [Indexed: 11/19/2022]
Abstract
Periodontitis is a chronic oral inflammatory disease affecting one in five individuals that can lead to tooth loss. CD4(+) Th cells activated by a microbial biofilm are thought to contribute to the destruction of alveolar bone surrounding teeth by influencing osteoclastogenesis through IL-17A and receptor activator for NF-κB ligand effects. The relative roles of mucosal Ag presentation cells in directing Th cell immune responses against oral pathogens and their contribution to destruction of alveolar bone remain unknown. We tested the contribution of mucosal Langerhans cells (LCs) to alveolar bone homeostasis in mice following oral colonization with a well-characterized human periodontal pathogen, Porphyromonas gingivalis We found that oral mucosal LCs did not protect from or exacerbate crestal alveolar bone destruction but were responsible for promoting differentiation of Th17 cells specific to P. gingivalis. In mice lacking LCs the Th17 response was suppressed and a Th1 response predominated. Bypassing LCs with systemic immunization of P. gingivalis resulted in a predominantly P. gingivalis-specific Th1 response regardless of whether LCs were present. Interestingly, we find that in vivo clonal expansion of P. gingivalis-specific Th cells and induced regulatory T cells does not depend on mucosal LCs. Furthermore, destruction of crestal alveolar bone induced by P. gingivalis colonization occurred regardless of the presence of mucosal LCs or P. gingivalis-specific Th17 cells. Our data indicate that both LCs and Th17 cells are redundant in contributing to alveolar bone destruction in a murine model of periodontitis.
Collapse
Affiliation(s)
- Peter D Bittner-Eddy
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis MN 55455; and
| | - Lori A Fischer
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis MN 55455; and
| | - Daniel H Kaplan
- Department of Dermatology, Medical School, University of Minnesota, Minneapolis MN 55455
| | - Kathleen Thieu
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis MN 55455; and
| | - Massimo Costalonga
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis MN 55455; and
| |
Collapse
|