Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.496
Filter
Add more filters

Publication year range
1.
Cell ; 184(15): 4090-4104.e15, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34129837

ABSTRACT

The oral mucosa remains an understudied barrier tissue. This is a site of rich exposure to antigens and commensals, and a tissue susceptible to one of the most prevalent human inflammatory diseases, periodontitis. To aid in understanding tissue-specific pathophysiology, we compile a single-cell transcriptome atlas of human oral mucosa in healthy individuals and patients with periodontitis. We uncover the complex cellular landscape of oral mucosal tissues and identify epithelial and stromal cell populations with inflammatory signatures that promote antimicrobial defenses and neutrophil recruitment. Our findings link exaggerated stromal cell responsiveness with enhanced neutrophil and leukocyte infiltration in periodontitis. Our work provides a resource characterizing the role of tissue stroma in regulating mucosal tissue homeostasis and disease pathogenesis.


Subject(s)
Immunity, Mucosal , Mouth Mucosa/cytology , Mouth Mucosa/immunology , Neutrophils/cytology , Adult , Epithelial Cells/cytology , Gene Expression Regulation , Genetic Predisposition to Disease , Gingiva/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Microbiota , Myeloid Cells/cytology , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology , Single-Cell Analysis , Stromal Cells/cytology , T-Lymphocytes/cytology
2.
Immunity ; 46(1): 133-147, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28087239

ABSTRACT

Immuno-surveillance networks operating at barrier sites are tuned by local tissue cues to ensure effective immunity. Site-specific commensal bacteria provide key signals ensuring host defense in the skin and gut. However, how the oral microbiome and tissue-specific signals balance immunity and regulation at the gingiva, a key oral barrier, remains minimally explored. In contrast to the skin and gut, we demonstrate that gingiva-resident T helper 17 (Th17) cells developed via a commensal colonization-independent mechanism. Accumulation of Th17 cells at the gingiva was driven in response to the physiological barrier damage that occurs during mastication. Physiological mechanical damage, via induction of interleukin 6 (IL-6) from epithelial cells, tailored effector T cell function, promoting increases in gingival Th17 cell numbers. These data highlight that diverse tissue-specific mechanisms govern education of Th17 cell responses and demonstrate that mechanical damage helps define the immune tone of this important oral barrier.


Subject(s)
Gingiva/immunology , Immunity, Mucosal/immunology , Immunologic Surveillance/immunology , Mouth Mucosa/immunology , Th17 Cells/immunology , Animals , Flow Cytometry , Gingiva/microbiology , Humans , Mastication , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Mouth Mucosa/microbiology , Real-Time Polymerase Chain Reaction
3.
Histopathology ; 85(1): 40-50, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38497348

ABSTRACT

AIMS: Oral epithelial dysplasia (OED) often exhibits a lymphocytic/lichenoid immune response (LIR), imparting histological resemblance to lichenoid mucositis and rendering diagnosis challenging. The clinical appearances of OED and lichenoid inflammatory processes are generally divergent, presenting as well-demarcated hyperkeratotic plaques and diffuse white and/or red mucosal change with variably prominent Wickham striae, respectively. To date, clinicopathological characterisation of OED with LIR, including clinical/gross appearance, has not been depicted. METHODS AND RESULTS: Cases of solitary OED with LIR for which a clinical photograph was available were identified in the authors' institutional files. Clinical and histological features were documented. In 44 identified cases, dysplasia was mild (19 of 44, 43.2%), moderate (19 of 44, 43.2%) and severe (six of 44, 13.6%). Clinically/grossly, all 44 cases (100.0%), presented as well-demarcated hyperkeratotic plaques lacking diffuse white-and-red mucosal change or Wickham striae. Histologically, OED with LIR exhibited numerous 'lichenoid' features beyond the lymphocytic band in the superficial lamina propria, including: leucocyte transmigration (38 of 44, 86.4%), spongiosis (37 of 44, 84.1%), Civatte/colloid bodies (36 of 44, 81.8%), basal cell degeneration (29 of 45, 65.9%), sawtooth rete ridges (11 of 44, 25.0%) and subepithelial clefting (7 of 44, 15.9%). CONCLUSIONS: Virtually any lichenoid histological feature may be seen in OED with LIR, representing a significant diagnostic pitfall. The typical clinical appearance of OED with LIR is of a well-demarcated hyperkeratotic plaque, characteristic of keratinising dysplasia and devoid of lichenoid features. This suggests that pathologist access to clinical photographs during diagnostic interpretation of biopsied white lesions, which represents opportunity to perform gross examination of the disease process, may reduce interobserver variability and improve diagnostic accuracy in this challenging differential diagnosis.


Subject(s)
Lymphocytes , Humans , Male , Female , Middle Aged , Adult , Aged , Lymphocytes/pathology , Lymphocytes/immunology , Mouth Mucosa/pathology , Mouth Mucosa/immunology , Aged, 80 and over , Young Adult
4.
Immunity ; 43(2): 369-81, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26231115

ABSTRACT

Langerhans cells (LCs) populate the mucosal epithelium, a major entry portal for pathogens, yet their ontogeny remains unclear. We found that, in contrast to skin LCs originating from self-renewing radioresistant embryonic precursors, oral mucosal LCs derive from circulating radiosensitive precursors. Mucosal LCs can be segregated into CD103(+)CD11b(lo) (CD103(+)) and CD11b(+)CD103(-) (CD11b(+)) subsets. We further demonstrated that similar to non-lymphoid dendritic cells (DCs), CD103(+) LCs originate from pre-DCs, whereas CD11b(+) LCs differentiate from both pre-DCs and monocytic precursors. Despite this ontogenetic discrepancy between skin and mucosal LCs, the transcriptomic signature and immunological function of oral LCs highly resemble those of skin LCs but not DCs. These findings, along with the epithelial position, morphology, and expression of the LC-associated phenotype strongly suggest that oral mucosal LCs are genuine LCs. Collectively, in a tissue-dependent manner, murine LCs differentiate from at least three distinct precursors (embryonic, pre-DC, and monocytic) in steady state.


Subject(s)
Cell Differentiation , Dendritic Cells/immunology , Langerhans Cells/immunology , Monocytes/immunology , Mouth Mucosa/immunology , Animals , Antigens, CD/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Blood Circulation , CD11b Antigen/metabolism , Cells, Cultured , Epithelium/immunology , Integrin alpha Chains/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Skin/immunology , Transcriptome/immunology
5.
Inflamm Res ; 73(9): 1477-1492, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39073597

ABSTRACT

OBJECTIVE: We intended to map the single-cell profile of OLP, explore the molecular characteristics of unconventional T cells in OLP tissues. METHODS: Buccal mucosa samples from OLP patients and healthy individuals were used to prepare single-cell suspension. Single-cell RNA sequencing was used to analyze the proportion of all the cells, and the molecular characteristics of unconventional T cells. Immunohistochemical staining was used to detect the expression of unconventional T cells marker genes. RESULTS: The cell clusters from buccal mucosa were categorized into immune cells, fibroblasts, endothelial cells, and epithelial cells. Unconventional T cells with phenotype of CD247+TRDC+NCAM1+ were identified. Immunohistochemical staining revealed higher expression of unconventional T cell marker genes in OLP tissue, predominantly in the lamina propria. In OLP, unconventional T cells are in a unique stress response state, exhibited enhanced NF-κB signaling and apoptosis inhibition, enhanced heat shock protein genes expression, weakened cytotoxic function. A large number of ligand-receptor pairs were found between unconventional T cells and other cells, particularly with fibroblasts and endothelial cells. CONCLUSIONS: This study mapped the single-cell profile of OLP, delineated the molecular characteristics of unconventional T cells in OLP, and uncovered that these unconventional T cells are in a stress response state.


Subject(s)
Lichen Planus, Oral , Mouth Mucosa , Single-Cell Analysis , T-Lymphocytes , Humans , Lichen Planus, Oral/immunology , Lichen Planus, Oral/genetics , Lichen Planus, Oral/metabolism , T-Lymphocytes/immunology , Mouth Mucosa/immunology , Female , Male , Middle Aged , Sequence Analysis, RNA , Adult , NF-kappa B/metabolism , Fibroblasts/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Aged , Epithelial Cells/metabolism , Epithelial Cells/immunology
6.
J Oral Pathol Med ; 53(7): 480-490, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38866540

ABSTRACT

BACKGROUND: Oral lichen planus (OLP) is a common T cell-mediated oral mucosal immune inflammatory disease. Intraepithelial lymphocytes (IELs) are a unique subset of T cells that play an important role in regulating immune response. This study aims to investigate the phenotype and the differentiation mechanism of IELs in OLP. METHODS: The expression of CD4, CD8α, CD8ß, T-helper-inducing POZ/Krueppel-like factor (ThPOK), and RUNX family transcription factor 3 (Runx3) in the epithelium and peripheral blood mononuclear cells (PBMCs) of OLP was determined by immunofluorescence and immunohistochemistry. Then, the correlations among them were analyzed. Naïve CD4+ T cells were sorted from blood of OLP patients and stimulated with retinoic acid (RA) and transforming growth factor-ß1 (TGF-ß1). Then the expression of CD4, CD8α, CD8ß, ThPOK, and Runx3 was investigated by immunocytochemistry. RESULTS: CD8α expression and CD8αα+ cells were upregulated in the epithelium of OLP, whereas they were downregulated in PBMCs of OLP. CD8ß was not expressed in the epithelium of OLP. CD4, CD8α, and Runx3 expression and CD4+CD8α+ cells were increased, whereas ThPOK expression was decreased in the epithelium of OLP. CD8α expression was positively correlated with Runx3 expression, whereas ThPOK expression was negatively correlated with Runx3 expression. After RA and TGF-ß1 stimulation, CD8α and Runx3 expression was upregulated, and ThPOK expression was downregulated in naïve CD4+ T cells. CONCLUSION: CD4+CD8αα+ IELs may be the dominant phenotype of IELs in OLP, and the differentiation of CD4+CD8αα+ IELs in OLP is negatively regulated by ThPOK and positively regulated by Runx3.


Subject(s)
CD8 Antigens , Core Binding Factor Alpha 3 Subunit , Intraepithelial Lymphocytes , Lichen Planus, Oral , Phenotype , Humans , Core Binding Factor Alpha 3 Subunit/metabolism , Lichen Planus, Oral/metabolism , Lichen Planus, Oral/immunology , Lichen Planus, Oral/pathology , Female , Middle Aged , Male , Adult , Intraepithelial Lymphocytes/immunology , CD4 Antigens , Transcription Factors , Aged , CD4-Positive T-Lymphocytes , Mouth Mucosa/metabolism , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Cell Differentiation , DNA-Binding Proteins
7.
J Immunol ; 205(1): 282-289, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32471882

ABSTRACT

The relationship of Porphyromonas gingivalis and oral squamous cell carcinoma (OSCC) has been studied for several years. Previous studies have focused on the direct effect of P. gingivalis on the activities of primary epithelial cells and OSCC cells. However, the immune system is responsible for mediating cancer development, whether P. gingivalis can affect oral cancer immunity has seldom been explored to date. In this study, we investigated the role of P. gingivalis in the immunoevasion of OSCC. We evaluated the effect of P. gingivalis on the phagocytosis of Cal-27 cells (OSCC cell line) by bone marrow-derived macrophages in vitro and studied the effect of P. gingivalis on the growth of OSCC and the polarization of tumor-associated macrophages in vivo. We found that P. gingivalis was able to inhibit the phagocytosis of Cal-27 cells by macrophages, and membrane-component molecules of P. gingivalis, such as proteins, were speculated to be the effector components. In addition, sustained infection with antibiotics-inactivated P. gingivalis promoted OSCC growth in mice and induced the polarization of macrophages into M2 tumor-associated macrophages, which mainly display protumor properties. Transcriptome analysis and quantitative RT-PCR revealed that P. gingivalis infection upregulated the expression of genes encoding protumor molecules in Cal-27 cells (suprabasin, IL-1R2, and CD47) and in macrophages (IL-1α, CCL-3, and CCL-5). Our in vitro and in vivo data suggest that P. gingivalis can promote immunoevasion of oral cancer by protecting cancer from macrophage attack. To our knowledge, the present study reveals a novel mechanism by which P. gingivalis promotes OSCC development.


Subject(s)
Bacteroidaceae Infections/immunology , Macrophages/immunology , Mouth Neoplasms/immunology , Porphyromonas gingivalis/immunology , Squamous Cell Carcinoma of Head and Neck/immunology , Tumor Escape , Animals , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Carcinogenesis/immunology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/immunology , Host-Pathogen Interactions/immunology , Humans , Macrophages/metabolism , Mice , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Mouth Mucosa/pathology , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Phagocytosis/immunology , RNA-Seq , Specific Pathogen-Free Organisms , Squamous Cell Carcinoma of Head and Neck/microbiology , Squamous Cell Carcinoma of Head and Neck/pathology , Up-Regulation , Xenograft Model Antitumor Assays
8.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055069

ABSTRACT

The oral cavity is inhabited by a wide spectrum of microbial species, and their colonization is mostly based on commensalism. These microbes are part of the normal oral flora, but there are also opportunistic species that can cause oral and systemic diseases. Although there is a strong exposure to various microorganisms, the oral mucosa reduces the colonization of microorganisms with high rotation and secretion of various types of cytokines and antimicrobial proteins such as defensins. In some circumstances, the imbalance between normal oral flora and pathogenic flora may lead to a change in the ratio of commensalism to parasitism. Healthy oral mucosa has many important functions. Thanks to its integrity, it is impermeable to most microorganisms and constitutes a mechanical barrier against their penetration into tissues. Our study aims to present the role and composition of the oral cavity microbiota as well as defense mechanisms within the oral mucosa which allow for maintaining a balance between such numerous species of microorganisms. We highlight the specific aspects of the oral mucosa protecting barrier and discuss up-to-date information on the immune cell system that ensures microbiota balance. This study presents the latest data on specific tissue stimuli in the regulation of the immune system with particular emphasis on the resistance of the gingival barrier. Despite advances in understanding the mechanisms regulating the balance on the microorganism/host axis, more research is still needed on how the combination of these diverse signals is involved in the regulation of immunity at the oral mucosa barrier.


Subject(s)
Host Microbial Interactions/immunology , Immunity, Mucosal , Microbiota/immunology , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Age Factors , Animals , Autoimmunity , Biodiversity , Disease Susceptibility , Dysbiosis , Humans , Symbiosis
9.
Carcinogenesis ; 42(1): 127-135, 2021 02 11.
Article in English | MEDLINE | ID: mdl-32621740

ABSTRACT

Dysbiosis of oral microbiome may dictate the progression of oral squamous cell carcinoma (OSCC). Yet, the composition of oral microbiome fluctuates by saliva and distinct sites of oral cavity and is affected by risky behaviors (smoking, drinking and betel quid chewing) and individuals' oral health condition. To characterize the disturbances in the oral microbial population mainly due to oral tumorigenicity, we profiled the bacteria within the surface of OSCC lesion and its contralateral normal tissue from discovery (n = 74) and validation (n = 42) cohorts of male patients with cancers of the buccal mucosa. Significant alterations in the bacterial diversity and relative abundance of specific oral microbiota (most profoundly, an enrichment for genus Fusobacterium and the loss of genus Streptococcus in the tumor sites) were identified. Functional prediction of oral microbiome shown that microbial genes related to the metabolism of terpenoids and polyketides were differentially enriched between the control and tumor groups, indicating a functional role of oral microbiome in formulating a tumor microenvironment via attenuated biosynthesis of secondary metabolites with anti-cancer effects. Furthermore, the vast majority of microbial signatures detected in the discovery cohort was generalized well to the independent validation cohort, and the clinical validity of these OSCC-associated microbes was observed and successfully replicated. Overall, our analyses reveal signatures (a profusion of Fusobacterium nucleatum CTI-2 and a decrease in Streptococcus pneumoniae) and functions (decreased production of tumor-suppressive metabolites) of oral microbiota related to oral cancer.


Subject(s)
Dysbiosis/immunology , Early Detection of Cancer/methods , Microbiota/immunology , Mouth Mucosa/microbiology , Mouth Neoplasms/diagnosis , Squamous Cell Carcinoma of Head and Neck/diagnosis , Adult , Aged , Cohort Studies , DNA, Bacterial/isolation & purification , Disease Progression , Dysbiosis/diagnosis , Dysbiosis/microbiology , Dysbiosis/pathology , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/immunology , Fusobacterium nucleatum/isolation & purification , Humans , Male , Middle Aged , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Mouth Neoplasms/immunology , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Prognosis , RNA, Ribosomal, 16S/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/microbiology , Squamous Cell Carcinoma of Head and Neck/pathology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/isolation & purification , Tumor Microenvironment/immunology
10.
Scand J Immunol ; 93(1): e12972, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32892403

ABSTRACT

Mounting evidence supports the importance of mucosal immunity in the immune response to SARS-CoV-2. Active virus replication in the upper respiratory tract for the first days of infection opens a new perspective in immunological strategies to counteract viral pathogenicity. An effective mucosal innate immune response to SARS-CoV-2 paves the way to an also effective adaptive immune response. A strong local immune response seems to be crucial in the initial contention of the virus by the organism and for triggering the production of the necessary neutralizing antibodies in sera and mucosal secretions. However, if the innate immune response fails to overcome the immune evasion mechanisms displayed by the virus, the infection will progress and the lack of an adaptive immune response will take the patient to an overreactive but ineffective innate immune response. To revert this scenario, an immune strategy based on enhancement of immunity in the first days of infection would be theoretically well come. But serious concerns about cytokine response syndrome prevent us to do so. Fortunately, it is possible to enhance immune system response without causing inflammation through immunomodulation. Immunomodulation of local immune response at the oropharyngeal mucosa could hypothetically activate our mucosal immunity, which could send an early an effective warning to the adaptive immune system. There are studies on immunotherapeutic management of upper respiratory tract infections in children that can place us in the right path to design an immune strategy able to mitigate COVID-19 symptoms and reduce clinical progression.


Subject(s)
COVID-19/immunology , Immunomodulation , Mouth Mucosa/immunology , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , Cytokine Release Syndrome/etiology , Humans , Immunity, Mucosal , Immunosenescence , Polyphenols/therapeutic use
11.
BMC Cancer ; 21(1): 628, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34044801

ABSTRACT

BACKGROUND: Oral Squamous Cell Carcinoma (OSCC) results from a series of genetic alteration in squamous cells. This particular type of cancer considers one of the most aggressive malignancies to control because of its frequent local invasions to the regional lymph node. Although several biomarkers have been reported, the key marker used to predict the behavior of the disease is largely unknown. Here we report Long INterpersed Element-1 (LINE1 or L1) retrotransposon activity in post-operative oral cancer samples. L1 is the only active retrotransposon occupying around 17% of the human genome with an estimated 500,000 copies. An active L1 encodes two proteins (L1ORF1p and L1ORF2p); both of which are critical in the process of retrotransposition. Several studies report that the L1 retrotransposon is highly active in many cancers. L1 activity is generally determined by assaying L1ORF1p because of its high expression and availability of the antibody. However, due to its lower expression and unavailability of a robust antibody, detection of L1ORF2p has been limited. L1ORF2p is the crucial protein in the process of retrotransposition as it provides endonuclease and reverse transcriptase (RT) activity. METHODS: Immunohistochemistry and Western blotting were performed on the post-operative oral cancer samples and murine tissues. RESULTS: Using in house novel antibodies against both the L1 proteins (L1ORF1p and L1ORF2p), we found L1 retrotransposon is extremely active in post-operative oral cancer tissues. Here, we report a novel human L1ORF2p antibody generated using an 80-amino-acid stretch from the RT domain, which is highly conserved among different species. The antibody detects significant L1ORF2p expression in human oral squamous cell carcinoma (OSCC) samples and murine germ tissues. CONCLUSIONS: We report exceptionally high L1ORF1p and L1ORF2p expression in post-operative oral cancer samples. The novel L1ORF2p antibody reported in this study will serve as a useful tool to understand why L1 activity is deregulated in OSCC and how it contributes to the progression of this particular cancer. Cross-species reactivity of L1ORF2p antibody due to the conserved epitope will be useful to study the retrotransposon biology in mice and rat germ tissues.


Subject(s)
Antigens, Neoplasm/immunology , Long Interspersed Nucleotide Elements/genetics , Mouth Neoplasms/genetics , Open Reading Frames/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Amino Acid Sequence/genetics , Animals , Antigens, Neoplasm/genetics , HEK293 Cells , Humans , Mice , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Mouth Mucosa/surgery , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Mouth Neoplasms/surgery , Open Reading Frames/genetics , Rats , Sequence Alignment , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/surgery
12.
Cell Microbiol ; 22(8): e13209, 2020 08.
Article in English | MEDLINE | ID: mdl-32329166

ABSTRACT

The host-microbe relationship is pivotal for oral health as well as for peri-implant diseases. Peri-implant mucosa and commensal biofilm play important roles in the maintenance of host-microbe homeostasis, but little is known about how they interact. We have therefore investigated the early host-microbe interaction between commensal multispecies biofilm (Streptococcus oralis, Actinomyces naeslundii, Veillonella dispar, Porphyromonas gingivalis) and organotypic peri-implant mucosa using our three-dimensional model. After 24 hr, biofilms induced weak inflammatory reaction in the peri-implant mucosa by upregulation of five genes related to immune response and increased secretion of IL-6 and CCL20. Biofilm volume was reduced which might be explained by secretion of ß-Defensins-1, -2, and CCL20. The specific tissue reaction without intrinsic overreaction might contribute to intact mucosa. Thus, a relationship similar to homeostasis and oral health was established within the first 24 hr. In contrast, the mucosa was damaged and the bacterial distribution was altered after 48 hr. These were accompanied by an enhanced immune response with upregulation of additional inflammatory-related genes and increased cytokine secretion. Thus, the homeostasis-like relationship was disrupted. Such profound knowledge of the host-microbe interaction at the peri-implant site may provide the basis to improve strategies for prevention and therapy of peri-implant diseases.


Subject(s)
Biofilms , Fibroblasts/microbiology , Host Microbial Interactions , Models, Anatomic , Mouth Mucosa/microbiology , Actinomyces/physiology , Cytokines/immunology , Fibroblasts/immunology , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Mouth Mucosa/immunology , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/physiology , Veillonella/immunology , Veillonella/physiology
13.
J Immunol ; 202(7): 2044-2056, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30760622

ABSTRACT

Deregulated immune response to a dysbiotic resident microflora within the oral cavity leads to chronic periodontal disease, local tissue destruction, and various systemic complications. To preserve tissue homeostasis, inflammatory signaling pathways involved in the progression of periodontitis must be tightly regulated. A20 (TNFAIP3), a ubiquitin-editing enzyme, has emerged as one of the key regulators of inflammation. Yet, the function of A20 in the oral mucosa and the biological pathways in which A20 mitigates periodontal inflammation remain elusive. Using a combination of in vivo and ex vivo disease models, we report in this study that A20 regulates inflammatory responses to a keystone oral bacterium, Porphyromonas gingivalis, and restrains periodontal inflammation through its effect on NF-κB signaling and cytokine production. Depletion of A20 using gene editing in human macrophage-like cells (THP-1) significantly increased cytokine secretion, whereas A20 overexpression using lentivirus infection dampened the cytokine production following bacterial challenge through modulating NF-κB activity. Similar to human cells, bone marrow-derived macrophages from A20-deficient mice infected with P. gingivalis displayed increased NF-κB activity and cytokine production compared with the cells isolated from A20-competent mice. Subsequent experiments using a murine ligature-induced periodontitis model showed that even a partial loss of A20 promotes an increased inflammatory phenotype and more severe bone loss, further verifying the critical function of A20 in the oral mucosa. Collectively, to our knowledge, these findings reveal the first systematic evidence of a physiological role for A20 in the maintenance of oral tissue homeostasis as a negative regulator of inflammation.


Subject(s)
Inflammation/immunology , Mouth Mucosa/immunology , NF-kappa B/immunology , Periodontitis/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Animals , HEK293 Cells , Humans , Immunity, Mucosal/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mouth Mucosa/metabolism , NF-kappa B/metabolism , Periodontitis/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
14.
Int J Med Sci ; 18(8): 1746-1752, 2021.
Article in English | MEDLINE | ID: mdl-33746591

ABSTRACT

The aim of this study was to examine the acid-electrolyzed functional water (FW)-mediated cytokine release in an oral squamous cell carcinoma-derived cell line (OSCC) following treatment with FW. FW is generated by the electrolysis of a sodium chloride solution and accelerate the burn wound healing. To elucidate the underlying mechanisms, the cytokine/chemokine secretion profile of HSC3 cells was examined using a cytokine array. FW treatment significantly induced interleukin (IL)-1α secretion, which was confirmed by enzyme-linked immunosorbent assay. Subsequently, the HSC3 cells were pre-treated with cycloheximide (CHX) for 1 h prior to FW stimulation to determine whether the augmented IL-1α secretion was due to enhanced protein synthesis. CHX pre-treatment did not affect IL-1α secretion suggesting that the secreted IL-1α might have been derived from intracellular storage sites. The amount of IL-1α in the cell lysate of the FW-treated HSC3 cells was significantly lower than that of the non-treated cells. Immunofluorescence staining using a polyclonal antibody against full-length IL-1α revealed a drastic reduction in IL-1α inside the FW- treated cells. IL-1α is synthesized in its precursor form (pIL-1α) and cleaved to produce pro-piece and mature IL-1α (ppIL-1α and mIL-1α) inside the cells. In the present study, only pIL-1α was detected within the HSC3 cells in its resting state. However, FW stimulation resulted in the release of the 33 kDa and two other smaller forms (about 19 kDa) of the protein. These results indicates that FW treatment induces IL-1α secretion, a typical alarmin, from the intracellular storage in OSCC cells.


Subject(s)
Interleukin-1alpha/metabolism , Intracellular Space/metabolism , Mouth Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/therapy , Water/administration & dosage , Cell Line, Tumor , Electrolysis , Humans , Intracellular Space/immunology , Mouth Mucosa/cytology , Mouth Mucosa/immunology , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , Water/chemistry
15.
Int J Mol Sci ; 22(15)2021 Jul 22.
Article in English | MEDLINE | ID: mdl-34360589

ABSTRACT

The oral mucosa, which is the lining tissue of the oral cavity, is a gateway to the body and it offers first-line protection against potential pathogens, exogenous chemicals, airborne allergens, etc. by means of its physical and microbiological-immune barrier functions. For this reason, oral mucosa is considered as a mirror to the health of the individual as well as a guard or early warning system. It is organized in two main components: a physical barrier, which consists of stratified epithelial cells and cell-cell junctions, and a microbiological-immune barrier that keeps the internal environment in a condition of homeostasis. Different factors, including microorganism, saliva, proteins and immune components, have been considered to play a critical role in disruption of oral epithelial barrier. Altered mucosal structure and barrier functions results in oral pathologies as well as systemic diseases. About 700 kinds of microorganisms exist in the human mouth, constituting the oral microbiota, which plays a significant role on the induction, training and function of the host immune system. The immune system maintains the symbiotic relationship of the host with this microbiota. Crosstalk between the oral microbiota and immune system includes various interactions in homeostasis and disease. In this review, after reviewing briefly the physical barriers of oral mucosa, the fundamentals of oral microbiome and oral mucosal immunity in regard to their barrier properties will be addressed. Furthermore, their importance in development of new diagnostic, prophylactic and therapeutic strategies for certain diseases as well as in the application for personalized medicine will be discussed.


Subject(s)
Homeostasis , Immunity, Mucosal/immunology , Microbiota , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Animals , Humans
16.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34830032

ABSTRACT

The oral mucosa is a site of intense immune activity, where a large variety of immune cells meet to provide a first line of defense against pathogenic organisms. Interestingly, the oral mucosa is exposed to a plethora of antigens from food and commensal bacteria that must be tolerated. The mechanisms that enable this tolerance are not yet fully defined. Many works have focused on active immune mechanisms involving dendritic and regulatory T cells. However, epithelial cells also make a major contribution to tolerance by influencing both innate and adaptive immunity. Therefore, the tolerogenic mechanisms concurring in the oral mucosa are intertwined. Here, we review them systematically, paying special attention to the role of oral epithelial cells.


Subject(s)
Adaptive Immunity , Epithelial Cells/immunology , Immune Tolerance , Immunity, Mucosal , Mouth Mucosa/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans
17.
Bull Exp Biol Med ; 172(2): 158-163, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855089

ABSTRACT

In postnatal ontogeny, the topographic relationships of the tongue glands and lymphoid structures in the thickness of the tongue have clear age-related features. In this article, we discuss the features of the glandular-lymphoid relationship in the thickness of the tongue, which is of particular scientific and practical importance for more precise understanding of the mechanisms providing local immunity in the oral cavity.


Subject(s)
Lymphoid Tissue/immunology , Mouth Mucosa/immunology , Tongue/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Autopsy , Child , Child, Preschool , Female , Humans , Immunity, Innate/physiology , Infant , Infant, Newborn , Lymphoid Tissue/pathology , Male , Middle Aged , Mouth Mucosa/pathology , Salivary Glands/immunology , Salivary Glands/pathology , Tongue/pathology , Young Adult
18.
Eur J Immunol ; 49(1): 133-143, 2019 01.
Article in English | MEDLINE | ID: mdl-30372518

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-17/metabolism , Mouth Mucosa/immunology , Mucosal-Associated Invariant T Cells/immunology , Adult , CD8 Antigens/metabolism , Female , Healthy Volunteers , Humans , Lymphocyte Activation , Lymphocyte Count , Male , Middle Aged , Young Adult
19.
PLoS Pathog ; 14(5): e1007069, 2018 05.
Article in English | MEDLINE | ID: mdl-29782555

ABSTRACT

The opportunistic fungal pathogen Candida albicans frequently causes diseases such as oropharyngeal candidiasis (OPC) in immunocompromised individuals. Although it is well appreciated that the cytokine IL-17 is crucial for protective immunity against OPC, the cellular source and the regulation of this cytokine during infection are still a matter of debate. Here, we directly visualized IL-17 production in the tongue of experimentally infected mice, thereby demonstrating that this key cytokine is expressed by three complementary subsets of CD90+ leukocytes: RAG-dependent αß and γδ T cells, as well as RAG-independent ILCs. To determine the regulation of IL-17 production at the onset of OPC, we investigated in detail the myeloid compartment of the tongue and found a heterogeneous and dynamic mononuclear phagocyte (MNP) network in the infected tongue that consists of Zbtb46-Langerin- macrophages, Zbtb46+Langerin+ dendritic cells (DCs) and Ly6C+ inflammatory monocytes. Of those, the Langerin+ DC population stands out by its unique capacity to co-produce the cytokines IL-1ß, IL-6 and IL-23, all of which promote IL-17 induction in response to C. albicans in the oral mucosa. The critical role of Langerin+ DCs for the innate IL-17 response was confirmed by depletion of this cellular subset in vivo, which compromised IL-17 induction during OPC. In conclusion, our work revealed key regulatory factors and their cellular sources of innate IL-17-dependent antifungal immunity in the oral mucosa.


Subject(s)
Antigens, Surface/immunology , Candida albicans/immunology , Candidiasis, Oral/immunology , Dendritic Cells/immunology , Interleukin-17/biosynthesis , Lectins, C-Type/immunology , Mannose-Binding Lectins/immunology , Mouth Mucosa/immunology , Animals , Candidiasis, Oral/microbiology , Cytokines/immunology , Female , Flow Cytometry , Interleukin-1beta/biosynthesis , Interleukin-23/biosynthesis , Interleukin-23/immunology , Interleukin-6/biosynthesis , Leukocytes/immunology , Male , Mice , Mice, Inbred C57BL , Mononuclear Phagocyte System/immunology , Mouth Mucosa/cytology , Mouth Mucosa/microbiology , Neutrophils/immunology , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/immunology , Thy-1 Antigens/immunology , Tongue/cytology , Tongue/immunology , Tongue/microbiology
20.
Clin Exp Allergy ; 50(8): 954-963, 2020 08.
Article in English | MEDLINE | ID: mdl-32501552

ABSTRACT

BACKGROUND: IgE-mediated food allergy remains a significant and growing worldwide problem. Sublingual immunotherapy (SLIT) shows an excellent safety profile for food allergy, but the clinical efficacy needs to be improved. This study assessed the effects of the Toll-like receptor 4 agonist outer membrane protein (Omp) 16 from Brucella abortus combined with cow´s milk proteins (CMP) through the sublingual route to modulate cow's milk allergy in an experimental model. METHODS: Mice sensitized with cholera toxin and CMP were orally challenged with the allergen to elicit hypersensitivity reactions. Then, mice were treated with a very low amount of CMP along with Omp16 as a mucosal adjuvant, and finally, animals were re-exposed to CMP. Systemic and mucosal immune parameters were assessed in vivo and in vitro. RESULTS: We found that the sublingual administration of Omp16 + CMP induced a buccal Th1 immune response that modulated the intestinal allergic response with the suppression of symptoms, reduction of IgE and IL-5, and up-regulation of IgG2a and IFN-γ. The adoptive transfer of submandibular IFN-γ-producing α4ß7+ CD4+ and CD8+ cells conferred protection against allergic sensitization. The use of Omp16 + CMP promoted enhanced protection compared to CMP alone. CONCLUSION: In conclusion, Omp16 represents a promising mucosal adjuvant that can be used to improve the clinical and immune efficacy of SLIT for food allergy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Allergens/administration & dosage , Bacterial Outer Membrane Proteins/administration & dosage , Cell Cycle Proteins/administration & dosage , Immunity, Mucosal/drug effects , Intestinal Mucosa/drug effects , Milk Hypersensitivity/therapy , Milk Proteins/administration & dosage , Sublingual Immunotherapy , T-Lymphocyte Subsets/drug effects , Administration, Sublingual , Adoptive Transfer , Allergens/immunology , Animals , Bacterial Outer Membrane Proteins/immunology , Cell Cycle Proteins/immunology , Cells, Cultured , Disease Models, Animal , Female , Immunoglobulin E/metabolism , Immunoglobulin G/metabolism , Interferon-gamma/metabolism , Interleukin-5/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Inbred BALB C , Milk Hypersensitivity/immunology , Milk Hypersensitivity/metabolism , Milk Proteins/immunology , Mouth Mucosa/drug effects , Mouth Mucosa/immunology , Mouth Mucosa/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL