1
|
Ahmed HS. The Impact of Prevotella on Neurobiology in Aging: Deciphering Dendritic Cell Activity and Inflammatory Dynamics. Mol Neurobiol 2024; 61:9240-9251. [PMID: 38613648 DOI: 10.1007/s12035-024-04156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Prevotella species, notably Prevotella copri, significantly populate the human gut. In particular, P. copri is prevalent among non-Western populations with diets high in fiber. These species show complex relationships with diverse health aspects, associating with beneficial outcomes, including reduced visceral fat and improved glucose tolerance. Studies implicate various Prevotella species in specific diseases. Prevotella nigrescens and Porphyromonas gingivalis were linked to periodontal disease, promoting immune responses and influencing T helper type 17 (Th17) cells. Prevotella bivia was associated with bacterial vaginosis and a specific increase in activated cells in the vaginal mucosa. In contrast, they have shown substantial potential for inducing connective tissue degradation and alveolar bone resorption. Prevotella's role in neuroinflammatory disorders and autoinflammatory conditions such as Alzheimer's disease and Parkinson's disease has also been noted. The complex relationship between Prevotella and age-related conditions further extends to neurobiological changes in aging, with varying associations with Alzheimer's, Parkinson's, and other inflammatory conditions. Studies have also identified Prevotella to be implicated in cognitive decline in middle aged and the elderly. Future directions in this research area are anticipated to explore Prevotella-associated inflammatory mechanisms and therapeutic interventions. Investigating specific drug targets and immunomodulatory measures could lead to novel therapeutic strategies. Understanding how Prevotella-induced inflammation interacts with aging diseases would offer promising insights for treatments and interventions. This review urges ongoing research to discover therapeutic targets and mechanisms for moderating Prevotella-associated inflammation to further enhance our understanding and improve health outcomes.
Collapse
Affiliation(s)
- H Shafeeq Ahmed
- Bangalore Medical College and Research Institute, K.R Road, Bangalore, 560002, Karnataka, India.
| |
Collapse
|
2
|
Zhou Y, Qin Y, Ma J, Li Z, Heng W, Zhang L, Liu H, Li R, Zhang M, Peng Q, Ye P, Duan N, Liu T, Wang W, Wang X. Heat-killed Prevotella intermedia promotes the progression of oral squamous cell carcinoma by inhibiting the expression of tumor suppressors and affecting the tumor microenvironment. Exp Hematol Oncol 2024; 13:33. [PMID: 38515216 PMCID: PMC10956211 DOI: 10.1186/s40164-024-00500-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Oral microbial dysbiosis contributes to the development of oral squamous cell carcinoma (OSCC). Our previous study showed that Prevotella intermedia (P. intermedia) were enriched in the oral mucosal surface, plaque, and saliva of patients with OSCC. Intratumoral microbiome could reshape the immune system and influence the development of various tumors. However, the invasion status of human OSCC tissues by P. intermedia and the pathway through which intratumoral P. intermedia potentiates tumor progression remain unexplored. METHODS P. intermedia in human OSCC or normal tissues was detected by FISH. A mouse OSCC cell line SCC7 was adopted to investigate the effects of heat-killed P. intermedia treatment on cell proliferation, invasion, and cytokine release by using CCK-8 assay, transwell invasion assay and ELISA. Moreover, we established a mouse transplanted tumor model by using SCC7 cells, injected heat-killed P. intermedia into tumor tissues, and investigated the effects of heat-killed P. intermedia on tumor growth, invasion, cytokine levels, immune cell infiltrations, and expression levels by using gross observation, H&E staining, ELISA, immunohistochemistry, mRNA sequencing, and transcriptomic analysis. RESULTS Our results indicated that P. intermedia were abundant in OSCC and surrounding muscle tissues. Heat-killed P. intermedia promoted SCC7 cell proliferation, invasion and proinflammatory cytokine secretions, accelerated transplanted tumor growth in mice, exacerbate muscle and perineural invasion of OSCC, elevated the serum levels of IL-17A, IL-6, TNF-α, IFN-γ, and PD-L1, induced Treg cells M2 type macrophages in mouse transplanted tumors. The data of transcriptomic analysis revealed that heat-killed P. intermedia increased the expression levels of inflammatory cytokines and chemokines while reduced the expression levels of some tumor suppressor genes in mouse transplanted tumors. Additionally, IL-17 signaling pathway was upregulated whereas GABAergic system was downregulated by heat-killed P. intermedia treatment. CONCLUSIONS Taken together, our results suggest that P. intermedia could inhibit the expression of tumor suppressors, alter the tumor microenvironment, and promote the progression of OSCC.
Collapse
Affiliation(s)
- Yifan Zhou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Yao Qin
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Jingjing Ma
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Zhiyuan Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Weiwei Heng
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Lei Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Hong Liu
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Ruowei Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Miaomiao Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Qiao Peng
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Pei Ye
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Ning Duan
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Ting Liu
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.
| | - Wenmei Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.
| | - Xiang Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.
| |
Collapse
|
3
|
Wu L, Luo Z, Chen Y, Yan Z, Fu J, Jiang Y, Xu J, Liu Y. Butyrate Inhibits Dendritic Cell Activation and Alleviates Periodontitis. J Dent Res 2023; 102:1326-1336. [PMID: 37775917 DOI: 10.1177/00220345231187824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023] Open
Abstract
Dendritic cells (DCs) can mediate inflammation-related bone resorption that is crucial in the development of periodontitis. Butyrate is a critical by-product of microbes with antibacterial and anti-inflammatory properties. Here, we found that butyrate inhibited the activation of lipopolysaccharide (LPS)-induced DCs and generation of inflammatory cytokines by DCs. Moreover, butyrate regulated glycolysis in LPS-induced DCs via the G-protein-coupled receptor/hypoxia-inducible factor-1α pathway. In addition, butyrate inhibited the maturation of CD11c+MHC-II+ DCs in vivo, suppressing local inflammatory infiltration and ultimately alleviating bone resorption in a periodontitis model. Our results imply that butyrate suppresses the activation of LPS-induced DCs by modulating their metabolism, highlighting its potential as a therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- L Wu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Z Luo
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Y Chen
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Z Yan
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - J Fu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Y Jiang
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - J Xu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Y Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| |
Collapse
|
4
|
de Vasconcelos Gurgel BC, Peixe PG, Queiroz SIML, de Almeida Freitas R, de Aquino Martins ARL, Duarte PM. Comparison of immunoexpression of dendritic cells, mast cells and blood vessels in periodontal disease between adults and elderly. Clin Oral Investig 2023; 27:6823-6833. [PMID: 37814161 DOI: 10.1007/s00784-023-05297-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
OBJECTIVE The aim of this study was to compare, in adults and elderly individuals, the immunoexpression of immature and mature dendritic cells (DCs), mast cells, and blood vessels in healthy and diseased gingival tissues. MATERIALS AND METHODS The expressions of immunohistochemical markers, including CD1a (immature dendritic cells), CD83 (mature dendritic cells), tryptase (mast cells) and CD34 (blood vessels), were analyzed in gingival biopsies from elderly (n = 27) and adult (n = 127) patients presenting health, gingivitis and periodontitis. Positive cells for each specimen and marker were counted. RESULTS There were no differences in the immunostaining of DCs, mast cells and the amount of blood vessels among gingival biopsies with health, gingivitis and periodontitis in adult and elderly subjects (p > 0.05). Immature DCs were more frequent in tissues with gingivitis and periodontitis in elderly patients, when compared to adults (p < 0.05). Furthermore, degranulated mast cell counts were higher, whereas the number of microvessels was lower in gingivitis in the elderly, when compared to adults (p < 0.05). CONCLUSIONS Diseased periodontal sites in the elderly present an overall significant overexpression of immature DCs and degranulated mast cells, in relation to those of adults. Furthermore, gingivitis in elderly is associated with decreased microvessel growth. These immunoinflammatory differences between elderly and adults may have implications in periodontal tissue breakdown in the late adulthood. Further studies should be performed to elucidate this hypothesis. CLINICAL RELEVANCE Understading the relationship between aging and changes in immune cells during periodontal inflammation may lead to therapeutic targets for the future management of periodontal diseases.
Collapse
Affiliation(s)
- Bruno Cesar de Vasconcelos Gurgel
- Department of Dentistry, Federal University of Rio Grande do Norte, 1787, Senador Salgado Filho Ave, Lagoa Nova, Natal, Rio Grande do Norte, CEP: 59056-000, Brazil.
| | - Patrícia Guerra Peixe
- Department of Dentistry, Federal University of Rio Grande do Norte, 1787, Senador Salgado Filho Ave, Lagoa Nova, Natal, Rio Grande do Norte, CEP: 59056-000, Brazil
| | - Salomão Israel Monteiro Lourenço Queiroz
- Department of Dentistry, Federal University of Rio Grande do Norte, 1787, Senador Salgado Filho Ave, Lagoa Nova, Natal, Rio Grande do Norte, CEP: 59056-000, Brazil
| | - Roseana de Almeida Freitas
- Department of Dentistry, Federal University of Rio Grande do Norte, 1787, Senador Salgado Filho Ave, Lagoa Nova, Natal, Rio Grande do Norte, CEP: 59056-000, Brazil
| | - Ana Rafaela Luz de Aquino Martins
- Department of Dentistry, Federal University of Rio Grande do Norte, 1787, Senador Salgado Filho Ave, Lagoa Nova, Natal, Rio Grande do Norte, CEP: 59056-000, Brazil
| | - Poliana Mendes Duarte
- Department of Periodontology, University of Florida, 1395 Center Dr, Gainesville, FL, 32610, USA
| |
Collapse
|
5
|
Zhu Y, Tao C, Goh C, Shrestha A. Innovative biomaterials for the treatment of periodontal disease. FRONTIERS IN DENTAL MEDICINE 2023; 4:1163562. [PMID: 39916927 PMCID: PMC11797777 DOI: 10.3389/fdmed.2023.1163562] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/03/2023] [Indexed: 02/09/2025] Open
Abstract
Periodontitis is a multifactorial disease that involves the destruction of hard and soft tissues surrounding the tooth. Routine periodontal treatment includes mechanical debridement (surgical and non-surgical) and the systemic administration of antibiotics. In contrast, severe and chronic periodontitis involves aggressive tissue destruction and bone resorption, and the damage is usually irreversible. In these severe cases, bone grafts, the delivery of growth hormones, and guided tissue regeneration can all be used to stimulate periodontal regeneration. However, these approaches do not result in consistent and predictable treatment outcomes. As a result, advanced biomaterials have evolved as an adjunctive approach to improve clinical performance. These novel biomaterials are designed to either prolong the release of antibacterial agents or osteogenic molecules, or to act as immunomodulators to promote healing. The first half of this review briefly summarizes the key immune cells and their underlying cellular pathways implicated in periodontitis. Advanced biomaterials designed to promote periodontal regeneration will be highlighted in the second half. Finally, the limitations of the current experimental design and the challenges of translational science will be discussed.
Collapse
Affiliation(s)
- Yi Zhu
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Chen Tao
- Stomatological Hospital of Chongqing, Key Laboratory of Oral Diseases and Biomaterial Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Cynthia Goh
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON, Canada
| | - Annie Shrestha
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Dentistry, Mt. Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
6
|
Renu S, Deblais L, Patil V, Schrock J, Kathayat D, Srivastava V, Feliciano-Ruiz N, Han Y, Ramesh A, Lakshmanappa YS, Ghimire S, Dhakal S, Rajashekara G, Renukaradhya GJ. Gut Microbiota of Obese Children Influences Inflammatory Mucosal Immune Pathways in the Respiratory Tract to Influenza Virus Infection: Optimization of an Ideal Duration of Microbial Colonization in a Gnotobiotic Pig Model. Microbiol Spectr 2022; 10:e0267421. [PMID: 35579462 PMCID: PMC9241774 DOI: 10.1128/spectrum.02674-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/15/2022] [Indexed: 11/20/2022] Open
Abstract
The impact of obesity on the human microbiota, immune maturation, and influenza virus infection has not been yet established in natural host animal models of influenza. In this study, gnotobiotic (Gn) pigs were colonized with human fecal microbiota (HFM) of obese (oHFM) or healthy lean (hHFM) children and infected at different periods (2-, 3-, and 5-weeks post-transplantation) using a zoonotic influenza virus strain. The infected oHFM pigs were characterized by lower levels of Firmicutes (Lactococcus, Lactobacillus, Turicibacter, and Streptococcus) and Actinobacteria (Bifidobacterium), which was associated with higher levels of Proteobacteria (Klebsiella), Bacteroidetes, and Verrucomicrobia (Akkermansia) compared with the infected hHFM group (P < 0.01). Furthermore, these genera significantly correlated with the expression of immune effectors, immune regulators, and inflammatory mediators, and displayed opposite trends between oHFM and hHFM groups (P < 0.01). The lymphoid and myeloid immune cell frequencies were differently modulated by the oHFM and hHFM colonization, especially apparent in the 5-weeks HFM colonized piglets. In addition, oHFM group had higher pro-inflammatory cytokines (IL-6, IL-12, TNF-α, and IFNγ) gene expression in the respiratory tract compared with the hHFM colonized pigs was detected. In conclusion, pigs colonized for longer duration, established oHFM increased the immune maturation favoring the activation of inflammatory mediators, however, the influenza virus load remained comparable with the hHFM group. Further, a longer duration of microbial colonization (5 weeks) may be required to reveal the impact of microbiome on the host immune maturation and susceptibility to influenza virus infection in the humanized Gn pig model. IMPORTANCE The diversity of gut microbiome of obese people differs markedly from that of lean healthy individuals which, in turn, influences the severity of inflammatory diseases because of differential maturation of immune system. The mouse model provides crucial insights into the mechanism(s) regulating the immune systems mediated by the gut microbiota but its applicability to humans is questionable because immune cells in mice are poorly activated in microbiota humanized mice. Several important strains of Bifidobacterium, Lactobacillus, and Clostridium fails to colonize the murine gut. Thus, understanding the role of certain important commensal gut bacterial species influences upon health and disease, a suitable large animal model like pig that supports the growth and colonization of most of the important human gut bacteria and possess comparable immunology and physiology to humans is beneficial to improve health.
Collapse
Affiliation(s)
- Sankar Renu
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Loic Deblais
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Veerupaxagouda Patil
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Jennifer Schrock
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Dipak Kathayat
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Vishal Srivastava
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Ninoshkaly Feliciano-Ruiz
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Yi Han
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Anikethana Ramesh
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Yashavanth S. Lakshmanappa
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Shristi Ghimire
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Santosh Dhakal
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Gireesh Rajashekara
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| | - Gourapura J. Renukaradhya
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, Ohio, USA
| |
Collapse
|
7
|
Liu Y, Lin Q, Huang X, Jiang G, Li C, Zhang X, Liu S, He L, Liu Y, Dai Q, Huang X. Effects of Dietary Ferulic Acid on the Intestinal Microbiota and the Associated Changes on the Growth Performance, Serum Cytokine Profile, and Intestinal Morphology in Ducks. Front Microbiol 2021; 12:698213. [PMID: 34326826 PMCID: PMC8313987 DOI: 10.3389/fmicb.2021.698213] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/09/2021] [Indexed: 01/11/2023] Open
Abstract
The present study investigated the effects of ferulic acid (FA) on the growth performance, serum cytokine profile, intestinal morphology, and intestinal microbiota in ducks at the growing stage. 300 female Linwu ducks at 28 days of age with similar body weights were randomly divided into five groups. Each group contained six replicates of 10 birds. The dietary treatments were corn-soybean-based diet supplemented with FA at the concentrations of 0 (control), 100, 200, 400, and 800 mg/kg diet. The results demonstrated that dietary FA at the levels of 200, 400, and 800 mg/kg increased the average daily gain (P = 0.01), 400 and 800 mg/kg FA increased the final body weight (P = 0.02), 100, 200, and 800 mg/kg FA increased the serum glutathione (P = 0.01), and 100, 400, and 800 mg/kg FA increased the glutathione peroxidase activities in birds (P < 0.01). Additionally, 200, 400, and 800 mg/kg dietary FA lowered the serum levels of interleukin-2 (P = 0.02) and interleukin-6 (P = 0.04). Moreover, the morphometric study of the intestines indicated that 400 mg/kg FA decreased the crypt depth in jejunum (P = 0.01) and caecum (P = 0.04), and increased the ratio of villus height to crypt depth in jejunum (P = 0.02). Significant linear and/or quadratic relationships were found between FA concentration and the measured parameters. 16S rRNA sequencing revealed that dietary FA increased the populations of genera Faecalibacterium, Paludicola, RF39, and Faecalicoccus in the cecum (P < 0.05), whereas decreased the populations of Anaerofilum and UCG-002 (P < 0.05). The Spearman correlation analysis indicated that phylum Proteobacteria were negatively, but order Oscillospirales, and family Ruminococcaceae were positively related to the parameters of the growth performance. Phylum Bacteroidetes, class Negativicutes and family Rikenellaceae were negatively associated with the parameters of the antioxidative capability. And phylum Cyanobacteria, Elusimicrobia, and Bacteroidetes, class Bacilli, family Rikenellaceae, and genus Prevotella were positively associated with the parameters of the immunological capability. Thus, it was concluded that the supplementations of 400 mg/kg FA in diet was able to improve the growth performance, antioxidative and immunological capabilities, intestinal morphology, and modulated the gut microbial construction of Linwu ducks at the growing stage.
Collapse
Affiliation(s)
- Yang Liu
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China.,Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Qian Lin
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Xuan Huang
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Guitao Jiang
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Chuang Li
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Xu Zhang
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Shengli Liu
- Shandong Lonct Enzymes Co., Ltd., Linyi, China
| | - Lingyun He
- Animal Husbandry and Fisheries Affairs Center, Huaihua, China
| | - Yali Liu
- Hunan Perfly Biotech Co., Ltd., Changsha, China
| | - Qiuzhong Dai
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Xingguo Huang
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| |
Collapse
|
8
|
Aziz J, Rahman MT, Vaithilingam RD. Dysregulation of metallothionein and zinc aggravates periodontal diseases. J Trace Elem Med Biol 2021; 66:126754. [PMID: 33831799 DOI: 10.1016/j.jtemb.2021.126754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/03/2021] [Accepted: 03/29/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Periodontitis (PD) is a multifaceted inflammatory disease connected to bacterial infection that results in the destruction of tooth supporting structures and eventually tooth loss. Given their involvement in infection and inflammation, both metallothionein (MT) and zinc (Zn) might play vital roles in the development and progression of PD. More specifically, both MT and Zn are heavily involved in regulating immune functions, controlling bacterial infection, balancing inflammatory responses, and reducing oxidative stress, all of which are associated with the pathogenesis of PD. OBJECTIVE This review paper will explore the physiological functions of MT and Zn and hypothesise how dysregulation could negatively affect periodontal health, leading to PD. FINDINGS Bacterial lipopolysaccharide (LPS) derived from periodontal pathogens, namely P. gingivalis initiates the acute phase response, thus upregulating the expression of MT which leads to the subsequent deficiency of Zn, a hallmark of periodontal disease. This deficiency leads to ineffective NETosis, increases the permeability of the gingival epithelium, and disrupts the humoral immune response, collectively contributing to PD. In addition, the presence of LPS in Zn deficient conditions favours M1 macrophage polarisation and maturation of dendritic cells, and also inhibits the anti-inflammatory activity of regulatory T cells. Collectively, these observations could theoretically give rise to the chronic inflammation seen in PD. CONCLUSION A disrupted MT and Zn homeostasis is expected to exert an adverse impact on periodontal health and contribute to the development and progression of PD.
Collapse
Affiliation(s)
- Jazli Aziz
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia; Dept. of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | | | - Rathna Devi Vaithilingam
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia
| |
Collapse
|
9
|
Tsuzuno T, Takahashi N, Yamada-Hara M, Yokoji-Takeuchi M, Sulijaya B, Aoki-Nonaka Y, Matsugishi A, Katakura K, Tabeta K, Yamazaki K. Ingestion of Porphyromonas gingivalis exacerbates colitis via intestinal epithelial barrier disruption in mice. J Periodontal Res 2021; 56:275-288. [PMID: 33512709 DOI: 10.1111/jre.12816] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/07/2020] [Accepted: 10/18/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This study aimed to evaluate the effects of ingested periodontal pathogens on experimental colitis in mice and to elucidate its underlying mechanisms. BACKGROUND Inflammatory bowel disease (IBD) is defined as a chronic intestinal inflammation that results in damage to the gastrointestinal tract. Epidemiological studies have shown an association between IBD and periodontitis. Although a large number of ingested oral bacteria reach gastrointestinal tract constantly, the effect of ingested periodontal pathogens on intestinal inflammation is still unknown. METHODS Experimental colitis was induced by inclusion of dextran sodium sulfate solution in drinking water of the mice. Major periodontal pathogens (Porphyromonas gingivalis, Prevotella intermedia, and Fusobacterium nucleatum) were administered orally every day during the experiment. The severity of colitis between the groups was compared. In vitro studies of the intestinal epithelial cell line were conducted to explore the molecular mechanisms by which periodontal pathogens affect the development of colitis. RESULTS The oral administration of P. gingivalis significantly increased the severity of colitis when compared to other pathogens in the DSS-induced colitis model. The ingested P. gingivalis disrupted the colonic epithelial barrier by decreasing the expression of tight junction proteins in vivo. In vitro permeability assays using the intestinal epithelial cell line suggested the P. gingivalis-specific epithelial barrier disruption. The possible involvement of gingipains in the exacerbation of colitis was implied by using P. gingivalis lacking gingipains. CONCLUSION Porphyromonas gingivalis exacerbates gastrointestinal inflammation by directly interacting with the intestinal epithelial barrier in a susceptible host.
Collapse
Affiliation(s)
- Takahiro Tsuzuno
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Takahashi
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Miki Yamada-Hara
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Mai Yokoji-Takeuchi
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Benso Sulijaya
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Yukari Aoki-Nonaka
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Aoi Matsugishi
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kyoko Katakura
- Department of Gastroenterology, Iwase general hospital, Fukushima, Japan
| | - Koichi Tabeta
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuhisa Yamazaki
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
10
|
Yang Y, Huang Y, Li W. Autophagy and its significance in periodontal disease. J Periodontal Res 2020; 56:18-26. [PMID: 33247437 DOI: 10.1111/jre.12810] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/24/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022]
Abstract
Autophagy is an evolutionarily conserved process essential for cellular homeostasis and human health. As a lysosome-dependent degradation pathway, autophagy acts as a modulator of the pathogenesis of diverse diseases. The relationship between autophagy and oral diseases has been explored in recent years, and there is increasing interest in the role of autophagy in periodontal disease. Periodontal disease is a prevalent chronic inflammatory disorder characterized by the destruction of periodontal tissues. It is initiated through pathogenic bacterial infection and interacts with the host immune defense, leading to inflammation and alveolar bone resorption. In this review, we outline the machinery of autophagy and present an overview of work on the significance of autophagy in regulating pathogen invasion, the immune response, inflammation, and alveolar bone homeostasis of periodontal disease. Existing data provide support for the importance of autophagy as a multi-dimensional regulator in the pathogenesis of periodontal disease and demonstrate the importance of future research on the potential roles of autophagy in periodontal disease.
Collapse
Affiliation(s)
- Yuhui Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yiping Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
11
|
Meghil MM, Tawfik OK, Elashiry M, Rajendran M, Arce RM, Fulton DJ, Schoenlein PV, Cutler CW. Disruption of Immune Homeostasis in Human Dendritic Cells via Regulation of Autophagy and Apoptosis by Porphyromonas gingivalis. Front Immunol 2019; 10:2286. [PMID: 31608069 PMCID: PMC6769118 DOI: 10.3389/fimmu.2019.02286] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
As fundamental processes of immune homeostasis, autophagy, and apoptosis must be maintained to mitigate risk of chronic inflammation and autoimmune diseases. Periodontitis is a chronic inflammatory disease characterized by oral microbial dysbiosis, and dysregulation of dendritic cell (DC) and T cell responses. The aim of this study was to elucidate the underlying mechanisms by which the oral microbe Porphyromonas gingivalis (P. gingivalis) manipulates dendritic cell signaling to perturb both autophagy and apoptosis. Using a combination of Western blotting, flow cytometry, qRT-PCR and immunofluorescence analysis, we show a pivotal role for the minor (Mfa1) fimbriae of P. gingivalis in nuclear/cytoplasmic shuttling of Akt and FOXO1 in human monocyte-derived DCs. Mfa1-induced Akt nuclear localization and activation ultimately induced mTOR. Activation of the Akt/mTOR axis downregulated intracellular LC3II, also known as Atg8, required for autophagosome formation and maturation. Use of allosteric panAkt inhibitor MK2206 and mTOR inhibitor rapamycin confirmed the role of Akt/mTOR signaling in autophagy inhibition by P. gingivalis in DCs. Interestingly, this pathway was also linked to induction of the anti-apoptotic protein Bcl2, decreased caspase-3 cleavage and decreased expression of pro-apoptotic proteins Bax and Bim, thus promoting longevity of host DCs. Addition of ABT-199 peptide to disrupt the interaction of antiapoptotic Bcl2 and its proapoptotic partners BAK/BAX restored apoptotic death to P. gingivalis-infected DC cells. In summary, we have identified the underlying mechanism by which P. gingivalis promotes its own survival and that of its host DCs.
Collapse
Affiliation(s)
- Mohamed M Meghil
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Omnia K Tawfik
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Mahmoud Elashiry
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Mythilypriya Rajendran
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Roger M Arce
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - David J Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Patricia V Schoenlein
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
12
|
Song L, Dong G, Guo L, Graves DT. The function of dendritic cells in modulating the host response. Mol Oral Microbiol 2017; 33:13-21. [PMID: 28845602 DOI: 10.1111/omi.12195] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2017] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are antigen-presenting cells that capture, process, and present antigens to lymphocytes to initiate and regulate the adaptive immune response. DCs detect bacteria in skin and mucosa and migrate into regional lymph nodes, where they stimulate antigen-specific T and B lymphocyte activation and proliferation. DCs direct CD4 T cells to differentiate to T-cell subsets such as T helper cells types 1, 2, and 17, and regulatory T cells. The periodontium is chronically exposed to oral bacteria that stimulate an inflammatory response to induce gingivitis or periodontitis. DCs play both protective and destructive roles through activation of the acquired immune response and are also reported to be a source of osteoclast precursors that promote bone resorption. FOXO1, a member of the forkhead box O family of transcription factors, plays a significant role in the activation of DCs. The function of DCs in periodontal inflammation has been investigated in a mouse model by lineage-specific deletion of FOXO1 in these cells. Deletion of FOXO1 reduces DC protective function and enhances susceptibility to periodontitis. The kinase Akt, phosphorylates FOXO1 to inhibit FOXO activity. Hence the Akt-FOXO1 axis may play a key role in regulating DCs to have a significant impact on periodontal disease.
Collapse
Affiliation(s)
- L Song
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - G Dong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - L Guo
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Molecular Laboratory for Gene Therapy and Tooth Regeneration and Department of Orthodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - D T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
13
|
Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 2017; 151:363-374. [PMID: 28542929 DOI: 10.1111/imm.12760] [Citation(s) in RCA: 843] [Impact Index Per Article: 105.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023] Open
Abstract
The microbiota plays a central role in human health and disease by shaping immune development, immune responses and metabolism, and by protecting from invading pathogens. Technical advances that allow comprehensive characterization of microbial communities by genetic sequencing have sparked the hunt for disease-modulating bacteria. Emerging studies in humans have linked the increased abundance of Prevotella species at mucosal sites to localized and systemic disease, including periodontitis, bacterial vaginosis, rheumatoid arthritis, metabolic disorders and low-grade systemic inflammation. Intriguingly, Prevotella abundance is reduced within the lung microbiota of patients with asthma and chronic obstructive pulmonary disease. Increased Prevotella abundance is associated with augmented T helper type 17 (Th17) -mediated mucosal inflammation, which is in line with the marked capacity of Prevotella in driving Th17 immune responses in vitro. Studies indicate that Prevotella predominantly activate Toll-like receptor 2, leading to production of Th17-polarizing cytokines by antigen-presenting cells, including interleukin-23 (IL-23) and IL-1. Furthermore, Prevotella stimulate epithelial cells to produce IL-8, IL-6 and CCL20, which can promote mucosal Th17 immune responses and neutrophil recruitment. Prevotella-mediated mucosal inflammation leads to systemic dissemination of inflammatory mediators, bacteria and bacterial products, which in turn may affect systemic disease outcomes. Studies in mice support a causal role of Prevotella as colonization experiments promote clinical and inflammatory features of human disease. When compared with strict commensal bacteria, Prevotella exhibit increased inflammatory properties, as demonstrated by augmented release of inflammatory mediators from immune cells and various stromal cells. These findings indicate that some Prevotella strains may be clinically important pathobionts that can participate in human disease by promoting chronic inflammation.
Collapse
Affiliation(s)
- Jeppe Madura Larsen
- Department of Technology, Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark.,National Food Institute, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|