1
|
Atagi Y, Homma Y, Yamashi S, Kikuchi K, Nagashima Y. Fatal Renal Abscess Caused by Porphyromonas gingivalis and Subcapsular Hemorrhage, Japan. Emerg Infect Dis 2024; 30:2214-2217. [PMID: 39320242 PMCID: PMC11431907 DOI: 10.3201/eid3010.240078] [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: 09/26/2024] Open
Abstract
A 61-year-old man in Japan with abdominal pain was suspected of having a renal tumor. Despite initial treatment, his condition rapidly deteriorated, leading to death. Postmortem examination revealed a renal abscess and sepsis caused by Porphyromonas gingivalis. This case underscores the need to consider atypical pathogens in renal masses.
Collapse
|
2
|
Blancas-Luciano BE, Becker-Fauser I, Zamora-Chimal J, Jiménez-García L, Lara-Martínez R, Pérez-Torres A, González del Pliego M, Aguirre-Benítez EL, Fernández-Presas AM. Cystatin C: immunoregulation role in macrophages infected with Porphyromonas gingivalis. PeerJ 2024; 12:e17252. [PMID: 38708345 PMCID: PMC11067906 DOI: 10.7717/peerj.17252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/26/2024] [Indexed: 05/07/2024] Open
Abstract
Background Periodontitis is a chronic infectious disease, characterized by an exacerbated inflammatory response and a progressive loss of the supporting tissues of the teeth. Porphyromonas gingivalis is a key etiologic agent in periodontitis. Cystatin C is an antimicrobial salivary peptide that inhibits the growth of P. gingivalis. This study aimed to evaluate the antimicrobial activity of this peptide and its effect on cytokine production, nitric oxide (NO) release, reactive oxygen species (ROS) production, and programmed cell death in human macrophages infected with P. gingivalis. Methods Monocyte-derived macrophages generated from peripheral blood were infected with P. gingivalis (MOI 1:10) and stimulated with cystatin C (2.75 µg/ml) for 24 h. The intracellular localization of P. gingivalis and cystatin C was determined by immunofluorescence and transmission electron microscopy (TEM). The intracellular antimicrobial activity of cystatin C in macrophages was assessed by counting Colony Forming Units (CFU). ELISA assay was performed to assess inflammatory (TNFα, IL-1β) and anti-inflammatory (IL-10) cytokines. The production of nitrites and ROS was analyzed by Griess reaction and incubation with 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA), respectively. Programmed cell death was assessed with the TUNEL assay, Annexin-V, and caspase activity was also determined. Results Our results showed that cystatin C inhibits the extracellular growth of P. gingivalis. In addition, this peptide is internalized in the infected macrophage, decreases the intracellular bacterial load, and reduces the production of inflammatory cytokines and NO. Interestingly, peptide treatment increased ROS production and substantially decreased bacterial-induced macrophage apoptosis. Conclusions Cystatin C has antimicrobial and immuno-regulatory activity in macrophages infected with P. gingivalis. These findings highlight the importance of understanding the properties of cystatin C for its possible therapeutic use against oral infections such as periodontitis.
Collapse
Affiliation(s)
- Blanca Esther Blancas-Luciano
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Microbiología y Parasitologia, Facultad de Medicina, Ciudad Universitaria, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ingeborg Becker-Fauser
- Unidad de Investigación en Medicina Experimental, Hospital General de México, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Hospital General de México, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis Jiménez-García
- Departamento de Biología Celular. Facultad de Ciencias, Ciudad Universitaria, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Reyna Lara-Martínez
- Departamento de Biología Celular. Facultad de Ciencias, Ciudad Universitaria, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Armando Pérez-Torres
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Margarita González del Pliego
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Elsa Liliana Aguirre-Benítez
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ana María Fernández-Presas
- Departamento de Microbiología y Parasitologia, Facultad de Medicina, Ciudad Universitaria, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Centro de Investigación en Ciencias de la Salud, Huixquilucan, Universidad Anáhuac, Estado de México, México
| |
Collapse
|
3
|
Blancas-Luciano BE, Zamora-Chimal J, da Silva-de Rosenzweig PG, Ramos-Mares M, Fernández-Presas AM. Macrophages immunomodulation induced by Porphyromonas gingivalis and oral antimicrobial peptides. Odontology 2023; 111:778-792. [PMID: 36897441 PMCID: PMC10492884 DOI: 10.1007/s10266-023-00798-w] [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: 08/25/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023]
Abstract
Porphyromonas gingivalis is a keystone pathogen associated with periodontitis development, a chronic inflammatory pathology characterized by the destruction of the supporting teeth structure. Macrophages are recruited cells in the inflammatory infiltrate from patients with periodontitis. They are activated by the P. gingivalis virulence factors arsenal, promoting an inflammatory microenvironment characterized by cytokine production (TNF-α, IL-1β, IL-6), prostaglandins, and metalloproteinases (MMPs) that foster the tissular destruction characteristic of periodontitis. Furthermore, P. gingivalis suppresses the generation of nitric oxide, a potent antimicrobial molecule, through its degradation, and incorporating its byproducts as a source of energy. Oral antimicrobial peptides can contribute to controlling the disease due to their antimicrobial and immunoregulatory activity, which allows them to maintain homeostasis in the oral cavity. This study aimed to analyze the immunopathological role of macrophages activated by P. gingivalis in periodontitis and suggested using antimicrobial peptides as therapeutic agents to treat the disease.
Collapse
Affiliation(s)
- Blanca Esther Blancas-Luciano
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Col. Universidad Nacional Autónoma de México, Av. Universidad 3000, CP 04510, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Ciudad Universitaria, Edificio D, 1° Piso, Mexico City, Mexico
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis, 148 Col. Doctores, Del. Cuauhtémoc, C.P. 06726, Mexico City, Mexico
| | - Pablo Gomes da Silva-de Rosenzweig
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, State of Mexico, Mexico
| | - Mariana Ramos-Mares
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, State of Mexico, Mexico
| | - Ana María Fernández-Presas
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Col. Universidad Nacional Autónoma de México, Av. Universidad 3000, CP 04510, Mexico City, Mexico.
| |
Collapse
|
4
|
Blancas-Luciano BE, Becker-Fauser I, Zamora-Chimal J, Delgado-Domínguez J, Ruíz-Remigio A, Leyva-Huerta ER, Portilla-Robertson J, Fernández-Presas AM. Antimicrobial and anti-inflammatory activity of Cystatin C on human gingival fibroblast incubated with Porphyromonas gingivalis. PeerJ 2022; 10:e14232. [PMID: 36312752 PMCID: PMC9615962 DOI: 10.7717/peerj.14232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/22/2022] [Indexed: 01/24/2023] Open
Abstract
Background Periodontal disease is considered one of the most prevalent chronic infectious diseases, often leading to the disruption of tooth-supporting tissues, including alveolar bone, causing tooth mobility and loss. Porphyromonas gingivalis is considered the major etiological agent of this disease, having a plethora of virulence factors, including, lipopolysaccharides (LPS), hemolysins, and proteinases. Antimicrobial peptides are one of the main components of the innate immune response that inhibit the growth of P. gingivalis. The aim of this study was to analyze the antimicrobial activity of cystatin C and to assess the effect on the inflammatory and anti-inflammatory cytokines, the production of reactive oxygen species, and in the release of nitric oxide by human gingival fibroblasts incubated with P. gingivalis in the presence and absence of cystatin C. Methods P. gingivalis ATCC 33277 was exposed to cystatin C for 24h and co-cultured with human gingival fibroblasts (HGFs) ATCC CRL-2014. The effect of cystatin on growth of P. gingivalis and HGFs was evaluated. Pro-inflammatory (TNFα, IL-1β) and anti-inflammatory (IL-10) cytokines were determined by ELISA in the supernatants of HGFs incubated with P. gingivalis exposed to cystatin C. Additionally, nitrites and reactive oxygen species (ROS) production were evaluated. Results Cystatin Cinhibited the growth of P. gingivalis without affecting HGFs. Incubation of HGFs with P. gingivalis led to a significant increase of TNF-α and IL-1β. In contrast, HGFs incubated with P. gingivalis exposed to cystatin C showed a decreased production of both cytokines, whereas IL-10 was enhanced. Incubation of HGFs with P. gingivalis led to an increase of nitric oxide (NO) and ROS production, which was reduced in the presence of the peptide. Conclusions Cystatin C inhibits the growth of P. gingivalis and decreases the inflammatory cytokines, ROS, and NO production during infection of HGFs with P. gingivalis. Knowledge on the antimicrobial and immunomodulatory properties of cystatin C could aid in the design of new therapeutic approaches to facilitate the elimination of this bacterium to improve the treatment of periodontal disease.
Collapse
Affiliation(s)
| | - Ingeborg Becker-Fauser
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - José Delgado-Domínguez
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - Adriana Ruíz-Remigio
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - Elba Rosa Leyva-Huerta
- Departmento de Medicina Oral y Patología, División de Posgrado, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, México
| | - Javier Portilla-Robertson
- Departmento de Medicina Oral y Patología, División de Posgrado, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, México
| | - Ana María Fernández-Presas
- Departamento de Microbiología y Parasitología, Universidad Nacional Autónoma de México, Mexico City, México,Centro de investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Mexico City, México
| |
Collapse
|
5
|
Gasmi Benahmed A, Kumar Mujawdiya P, Noor S, Gasmi A. Porphyromonas Gingivalis in the Development of Periodontitis: Impact on Dysbiosis and Inflammation. ARCHIVES OF RAZI INSTITUTE 2022; 77:1539-1551. [PMID: 37123122 PMCID: PMC10133641 DOI: 10.22092/ari.2021.356596.1875] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Revised: 11/20/2021] [Indexed: 05/02/2023]
Abstract
Chronic periodontitis is an inflammatory disease of the dental plaque and affects the soft tissues supporting the tooth. It is one of the most practical oral health issues across the globe and adversely affects the quality of life. In a neutrophil-mediated action, the inflammatory response to periodontitis destroys the periodontal ligaments, gums, the alveolar bone, and the cementum. Some of the most associated invasive pathogens with periodontitis are Porphyromonas Gingivalis, Aggregatibacter actinomycetecomitans, and Fusobacterium nucleatum. Google Scholar and PubMed were used to search the evidence using key terms like 'periodontitis,' 'Porphyromonas Gingivalis,' 'Oral Dysbiosis and Periodontitis,' 'Porphyromonas Gingivalis and Periodontitis,' etc. Only studies were included reviewing the Porphyromonas Gingivalis and its role in periodontitis. It has been observed from several oral pathogens that P. gingivalis has received immense attention due to a strong association between Porphyromonas Gingivalis and periodontal disease. Porphyromonas Gingivalis also disrupts the delicate balance between various members of the oral microbial communities and promotes oral dysbiosis. The dysbiotic state of the oral microbiome is distinct in functional capabilities and shows a higher expression of genes involved in lipopolysaccharide synthesis, energy regulation, and bacterial motility. Certain virulence factors such as gingipains, LPS, and fimbriae also increase the invasion and pathogenicity of Porphyromonas Gingivalis. Its presence in the periodontal tissues increases the secretion of numerous pro-inflammatory mediators such as TNF-α, IL-8, and IL-1β, leading to the destruction of soft gingival tissues and ligaments. Early detection of periodontitis and immediate treatment can prevent soft tissue destruction and dentition loss. In conclusion, details about the oral microbiome, oral dysbiosis, and inflammation may offer new therapeutic options in the future, including a personalized approach and the use of combination therapy.
Collapse
Affiliation(s)
- A Gasmi Benahmed
- Académie Internationale de Médecine Dentaire Intégrative, Paris, France
| | | | - S Noor
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Pakistan
| | - A Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
| |
Collapse
|
6
|
Schulz S, Stein JM, Schumacher A, Kupietz D, Yekta-Michael SS, Schittenhelm F, Conrads G, Schaller HG, Reichert S. Nonsurgical Periodontal Treatment Options and Their Impact on Subgingival Microbiota. J Clin Med 2022; 11:1187. [PMID: 35268280 PMCID: PMC8911148 DOI: 10.3390/jcm11051187] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Different periodontal treatment methods (quadrant-wise debridement, scaling and root planing (Q-SRP), full-mouth scaling (FMS), full-mouth disinfection (FMD), and FMD with adjuvant erythritol air-polishing (FMDAP)) were applied in periodontitis patients (stage III/IV). The study objective (substudy of ClinicalTrials.gov Identifier: NCT03509233) was to compare the impact of treatments on subgingival colonization. METHODS Forty patients were randomized to the treatment groups. Periodontal parameters and subgingival colonization were evaluated at baseline and 3 and 6 months after treatment. RESULTS Positive changes in clinical parameters were recorded in every treatment group during the 3-month follow-up period, but did not always continue. In three groups, specific bacteria decreased after 3 months; however, this was associated with a renewed increase after 6 months (FMS: Porphyromonas gingivalis; FMD: Eubacterium nodatum, Prevotella dentalis; and FMDAP: uncultured Prevotella sp.). CONCLUSIONS The benefit of all clinical treatments measured after 3 months was associated with a decrease in pathogenic bacteria in the FMS, FMD, and FMDAP groups. However, after 6 months, we observed further improvement or some stagnation in clinical outcomes accompanied by deterioration of the microbiological profile. Investigating the subgingival microbiota might help appraise successful periodontal treatment and implement individualized therapy.
Collapse
Affiliation(s)
- Susanne Schulz
- Department of Operative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany; (A.S.); (D.K.); (H.-G.S.); (S.R.)
| | - Jamal M. Stein
- Department of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital (RWTH) Aachen, 52074 Aachen, Germany; (J.M.S.); (S.S.Y.-M.); (F.S.); (G.C.)
- Private Practice, 52062 Aachen, Germany
| | - Anne Schumacher
- Department of Operative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany; (A.S.); (D.K.); (H.-G.S.); (S.R.)
| | - David Kupietz
- Department of Operative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany; (A.S.); (D.K.); (H.-G.S.); (S.R.)
| | - Sareh S. Yekta-Michael
- Department of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital (RWTH) Aachen, 52074 Aachen, Germany; (J.M.S.); (S.S.Y.-M.); (F.S.); (G.C.)
| | - Florian Schittenhelm
- Department of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital (RWTH) Aachen, 52074 Aachen, Germany; (J.M.S.); (S.S.Y.-M.); (F.S.); (G.C.)
- Private Practice, 52062 Aachen, Germany
| | - Georg Conrads
- Department of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital (RWTH) Aachen, 52074 Aachen, Germany; (J.M.S.); (S.S.Y.-M.); (F.S.); (G.C.)
| | - Hans-Günter Schaller
- Department of Operative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany; (A.S.); (D.K.); (H.-G.S.); (S.R.)
| | - Stefan Reichert
- Department of Operative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany; (A.S.); (D.K.); (H.-G.S.); (S.R.)
| |
Collapse
|
7
|
Dhaliwal JS, Abd Rahman NA, Ming LC, Dhaliwal SKS, Knights J, Albuquerque Junior RF. Microbial Biofilm Decontamination on Dental Implant Surfaces: A Mini Review. Front Cell Infect Microbiol 2021; 11:736186. [PMID: 34692562 PMCID: PMC8531646 DOI: 10.3389/fcimb.2021.736186] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction After insertion into the bone, implants osseointegrate, which is required for their long-term success. However, inflammation and infection around the implants may lead to implant failure leading to peri-implantitis and loss of supporting bone, which may eventually lead to failure of implant. Surface chemistry of the implant and lack of cleanliness on the part of the patient are related to peri-implantitis. The only way to get rid of this infection is decontamination of dental implants. Objective This systematic review intended to study decontamination of microbial biofilm methods on titanium implant surfaces used in dentistry. Methods The electronic databases Springer Link, Science Direct, and PubMed were explored from their inception until December 2020 to identify relevant studies. Studies included had to evaluate the efficiency of new strategies either to prevent formation of biofilm or to treat matured biofilm on dental implant surfaces. Results and Discussion In this systematic review, 17 different groups of decontamination methods were summarized from 116 studies. The decontamination methods included coating materials, mechanical cleaning, laser treatment, photodynamic therapy, air polishing, anodizing treatment, radiation, sonication, thermal treatment, ultrasound treatment, chemical treatment, electrochemical treatment, antimicrobial drugs, argon treatment, and probiotics. Conclusion The findings suggest that most of the decontamination methods were effective in preventing the formation of biofilm and in decontaminating established biofilm on dental implants. This narrative review provides a summary of methods for future research in the development of new dental implants and decontamination techniques.
Collapse
Affiliation(s)
- Jagjit Singh Dhaliwal
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei, Darussalam, Gadong, Brunei
| | - Nurul Adhwa Abd Rahman
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei, Darussalam, Gadong, Brunei
| | - Long Chiau Ming
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei, Darussalam, Gadong, Brunei
| | - Sachinjeet Kaur Sodhi Dhaliwal
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei, Darussalam, Gadong, Brunei
| | - Joe Knights
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei, Darussalam, Gadong, Brunei
| | | |
Collapse
|
8
|
Abdul Rahman R, Lamarca A, Hubner RA, Valle JW, McNamara MG. The Microbiome as a Potential Target for Therapeutic Manipulation in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13153779. [PMID: 34359684 PMCID: PMC8345056 DOI: 10.3390/cancers13153779] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Pancreatic cancer is one of the most lethal cancers. It is a difficult cancer to treat, and the complexity surrounding the pancreatic tumour is one of the contributing factors. The microbiome is the collection of microorganisms within an environment and its genetic material. They reside on body surfaces and most abundantly within the human gut in symbiotic balance with their human host. Disturbance in the balance can lead to many diseases, including cancers. Significant advances have been made in cancer treatment since the introduction of immunotherapy, and the microbiome may play a part in the outcome and survival of patients with cancer, especially those treated with immunotherapy. Immunotherapy use in pancreatic cancer remains challenging. This review will focus on the potential interaction of the microbiome with pancreas cancer and how this could be manipulated. Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers and is projected to be the second most common cause of cancer-related death by 2030, with an overall 5-year survival rate between 7% and 9%. Despite recent advances in surgical, chemotherapy, and radiotherapy techniques, the outcome for patients with PDAC remains poor. Poor prognosis is multifactorial, including the likelihood of sub-clinical metastatic disease at presentation, late-stage at presentation, absence of early and reliable diagnostic biomarkers, and complex biology surrounding the extensive desmoplastic PDAC tumour micro-environment. Microbiota refers to all the microorganisms found in an environment, whereas microbiome is the collection of microbiota and their genome within an environment. These organisms reside on body surfaces and within mucosal layers, but are most abundantly found within the gut. The commensal microbiome resides in symbiosis in healthy individuals and contributes to nutritive, metabolic and immune-modulation to maintain normal health. Dysbiosis is the perturbation of the microbiome that can lead to a diseased state, including inflammatory bowel conditions and aetiology of cancer, such as colorectal and PDAC. Microbes have been linked to approximately 10% to 20% of human cancers, and they can induce carcinogenesis by affecting a number of the cancer hallmarks, such as promoting inflammation, avoiding immune destruction, and microbial metabolites can deregulate host genome stability preceding cancer development. Significant advances have been made in cancer treatment since the advent of immunotherapy. The microbiome signature has been linked to response to immunotherapy and survival in many solid tumours. However, progress with immunotherapy in PDAC has been challenging. Therefore, this review will focus on the available published evidence of the microbiome association with PDAC and explore its potential as a target for therapeutic manipulation.
Collapse
Affiliation(s)
- Rozana Abdul Rahman
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust/Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK; (A.L.); (R.A.H.)
| | - Richard A. Hubner
- Department of Medical Oncology, The Christie NHS Foundation Trust/Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK; (A.L.); (R.A.H.)
| | - Juan W. Valle
- Division of Cancer Sciences, University of Manchester/Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
| | - Mairéad G. McNamara
- Division of Cancer Sciences, University of Manchester/Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
- Correspondence:
| |
Collapse
|
9
|
Mekhemar M, Geib M, Kumar M, Radha, Hassan Y, Dörfer C. Salvadora persica: Nature's Gift for Periodontal Health. Antioxidants (Basel) 2021; 10:712. [PMID: 33946353 PMCID: PMC8146554 DOI: 10.3390/antiox10050712] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 12/11/2022] Open
Abstract
Salvadora persica (SP) extract, displays very valuable biotherapeutic capacities such as antimicrobial, antioxidant, antiparasitic and anti-inflammatory effects. Numerous investigations have studied the pharmacologic actions of SP in oral disease therapies but its promising outcomes in periodontal health and treatment are not yet entirely described. The current study has been planned to analyze the reported effects of SP as a support to periodontal therapy to indorse regeneration and healing. In consort with clinical trials, in vitro investigations show the advantageous outcomes of SP adjunctive to periodontal treatment. Yet, comprehensive supplementary preclinical and clinical investigations at molecular and cellular levels are indispensable to reveal the exact therapeutic mechanisms of SP and its elements for periodontal health and therapy.
Collapse
Affiliation(s)
- Mohamed Mekhemar
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrecht’s University, 24105 Kiel, Germany; (Y.H.); (C.D.)
| | - Mathias Geib
- Dr. Geib Private Dental Clinic, Frankfurter Landstraße 79, 61352 Bad Homburg, Germany;
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR—Central Institute for Research on Cotton Technology, Mumbai 400019, India;
| | - Radha
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India;
| | - Yasmine Hassan
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrecht’s University, 24105 Kiel, Germany; (Y.H.); (C.D.)
| | - Christof Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrecht’s University, 24105 Kiel, Germany; (Y.H.); (C.D.)
| |
Collapse
|
10
|
The Emerging Role of Microbiota and Microbiome in Pancreatic Ductal Adenocarcinoma. Biomedicines 2020; 8:biomedicines8120565. [PMID: 33287196 PMCID: PMC7761686 DOI: 10.3390/biomedicines8120565] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignant tumors due to the absence of biomarkers for early-stage detection and poor response to therapy. Since mounting evidence supports the role of microbiota composition in tumorigenesis and cancer treatment, the link between microbiome and PDAC has been described. In this review, we summarize the current knowledge regarding the impact of the gut and oral microbiome on the risk of PDAC development. Microenvironment-driven therapy and immune system interactions are also discussed. More importantly, we provide an overview of the clinical trials evaluating the microbiota role in the risk, prognosis, and treatment of patients suffering from PDAC and solid tumors. According to the research findings, immune tolerance might result from the microbiota-derived remodeling of pancreatic tumor microenvironment. Thus, microbiome profiling and targeting represent the potential trend to enhance antitumor immunity and improve the efficacy of PDAC treatment.
Collapse
|
11
|
Johnson A, He JL, Kong F, Huang YC, Thomas S, Lin HTV, Kong ZL. Surfactin-Loaded ĸ-Carrageenan Oligosaccharides Entangled Cellulose Nanofibers as a Versatile Vehicle Against Periodontal Pathogens. Int J Nanomedicine 2020; 15:4021-4047. [PMID: 32606662 PMCID: PMC7293418 DOI: 10.2147/ijn.s238476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Periodontitis is a chronic inflammatory disease associated with microbial accumulation. The purpose of this study was to reuse the agricultural waste to produce cellulose nanofibers (CNF) and further modification of the CNF with κ-carrageenan oligosaccharides (CO) for drug delivery. In addition, this study is focused on the antimicrobial activity of surfactin-loaded CO-CNF towards periodontal pathogens. MATERIALS AND METHODS A chemo-mechanical method was used to extract the CNF and the modification was done by using CO. The studies were further proceeded by adding different quantities of surfactin [50 mg (50 SNPs), 100 mg (100 SNPs), 200 mg (200 SNPs)] into the carrier (CO-CNF). The obtained materials were characterized, and the antimicrobial activity of surfactin-loaded CO-CNF was evaluated. RESULTS The obtained average size of CNF and CO-CNF after ultrasonication was 263 nm and 330 nm, respectively. Microscopic studies suggested that the CNF has a short diameter with long length and CO became cross-linked to form as beads within the CNF network. The addition of CO improved the degradation temperature, crystallinity, and swelling property of CNF. The material has a controlled drug release, and the entrapment efficiency and loading capacity of the drug were 53.15 ± 2.36% and 36.72 ± 1.24%, respectively. It has antioxidant activity and inhibited the growth of periodontal pathogens such as Streptococcus mutans and Porphyromonas gingivalis by preventing the biofilm formation, reducing the metabolic activity, and promoting the oxidative stress. CONCLUSION The study showed the successful extraction of CNF and modification with CO improved the physical parameters of the CNF. In addition, surfactin-loaded CO-CNF has potential antimicrobial activity against periodontal pathogens. The obtained biomaterial is economically valuable and has great potential for biomedical applications.
Collapse
Affiliation(s)
- Athira Johnson
- Department of Food Science, National Taiwan Ocean University, Keelung20224, Taiwan
| | - Jia-Ling He
- Department of Food Science, National Taiwan Ocean University, Keelung20224, Taiwan
| | - Fanbin Kong
- Department of Food Science and Technology, University of Georgia, GA30602, U.S.A
| | - Yi-Cheng Huang
- Department of Food Science, National Taiwan Ocean University, Keelung20224, Taiwan
| | - Sabu Thomas
- School of Chemical Sciences, Mahatma Gandhi University, Kottayam, Kerala686560, India
| | - Hong-Ting Victor Lin
- Department of Food Science, National Taiwan Ocean University, Keelung20224, Taiwan
| | - Zwe-Ling Kong
- Department of Food Science, National Taiwan Ocean University, Keelung20224, Taiwan
| |
Collapse
|
12
|
Mahendra CK, Tan LTH, Lee WL, Yap WH, Pusparajah P, Low LE, Tang SY, Chan KG, Lee LH, Goh BH. Angelicin-A Furocoumarin Compound With Vast Biological Potential. Front Pharmacol 2020; 11:366. [PMID: 32372949 PMCID: PMC7176996 DOI: 10.3389/fphar.2020.00366] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Angelicin, a member of the furocoumarin group, is related to psoralen which is well known for its effectiveness in phototherapy. The furocoumarins as a group have been studied since the 1950s but only recently has angelicin begun to come into its own as the subject of several biological studies. Angelicin has demonstrated anti-cancer properties against multiple cell lines, exerting effects via both the intrinsic and extrinsic apoptotic pathways, and also demonstrated an ability to inhibit tubulin polymerization to a higher degree than psoralen. Besides that, angelicin too demonstrated anti-inflammatory activity in inflammatory-related respiratory and neurodegenerative ailments via the activation of NF-κB pathway. Angelicin also showed pro-osteogenesis and pro-chondrogenic effects on osteoblasts and pre-chondrocytes respectively. The elevated expression of pro-osteogenic and chondrogenic markers and activation of TGF-β/BMP, Wnt/β-catenin pathway confirms the positive effect of angelicin bone remodeling. Angelicin also increased the expression of estrogen receptor alpha (ERα) in osteogenesis. Other bioactivities, such as anti-viral and erythroid differentiating properties of angelicin, were also reported by several researchers with the latter even displaying an even greater aptitude as compared to the commonly prescribed drug, hydroxyurea, which is currently on the market. Apart from that, recently, a new application for angelicin against periodontitis had been studied, where reduction of bone loss was indirectly caused by its anti-microbial properties. All in all, angelicin appears to be a promising compound for further studies especially on its mechanism and application in therapies for a multitude of common and debilitating ailments such as sickle cell anaemia, osteoporosis, cancer, and neurodegeneration. Future research on the drug delivery of angelicin in cancer, inflammation and erythroid differentiation models would aid in improving the bioproperties of angelicin and efficacy of delivery to the targeted site. More in-depth studies of angelicin on bone remodeling, the pro-osteogenic effect of angelicin in various bone disease models and the anti-viral implications of angelicin in periodontitis should be researched. Finally, studies on the binding of angelicin toward regulatory genes, transcription factors, and receptors can be done through experimental research supplemented with molecular docking and molecular dynamics simulation.
Collapse
Affiliation(s)
- Camille Keisha Mahendra
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Loh Teng Hern Tan
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Wai Leng Lee
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Priyia Pusparajah
- Medical Health and Translational Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Liang Ee Low
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Siah Ying Tang
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Subang Jaya, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Subang Jaya, Malaysia
| | - Kok Gan Chan
- International Genome Centre, Jiangsu University, Zhenjiang, China
- Division of Genetics and Molecular Biology, Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Learn Han Lee
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Subang Jaya, Malaysia
| |
Collapse
|
13
|
Liu XB, Gao ZY, Sun CT, Wen H, Gao B, Li SB, Tong Q. The potential role of P.gingivalis in gastrointestinal cancer: a mini review. Infect Agent Cancer 2019; 14:23. [PMID: 31516546 PMCID: PMC6734237 DOI: 10.1186/s13027-019-0239-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/21/2019] [Indexed: 02/20/2023] Open
Abstract
Bacterial infection may be involved in the entire process of tissue carcinogenesis by directly or indirectly affecting the occurrence and development of tumors. Porphyromonas gingivalis (P.gingivalis) is an important pathogen causing periodontitis. Periodontitis may promote the occurrence of various tumors. Gastrointestinal tumors are common malignant tumors with high morbidity, high mortality, and low early diagnosis rate. With the rapid development of molecularbiotechnology, the role of P.gingivalis in digestive tract tumors has been increasingly explored. This article reviews the correlation between P.gingivalis and gastrointestinal cancer and the pathogenesis of the latter. The relationship among P.gingivalis, periodontal disease, and digestive tract tumors must be clarifiedthrough a multi-center, prospective, large-scale study.
Collapse
Affiliation(s)
- Xiao-Bo Liu
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, 32 south renmin road, Shiyan, Hubei 442000 People's Republic of China
| | - Zi-Ye Gao
- Department of Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 Hubei China
| | - Chuan-Tao Sun
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, 32 south renmin road, Shiyan, Hubei 442000 People's Republic of China
| | - Hui Wen
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, 32 south renmin road, Shiyan, Hubei 442000 People's Republic of China
| | - Bo Gao
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 Hubei China
| | - Sheng-Bao Li
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, 32 south renmin road, Shiyan, Hubei 442000 People's Republic of China
| | - Qiang Tong
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, 32 south renmin road, Shiyan, Hubei 442000 People's Republic of China
| |
Collapse
|
14
|
Shao J, Zhang M, Wu L, Jia XW, Jin YH, Zeng XT. DEFB1 rs11362 Polymorphism and Risk of Chronic Periodontitis: A Meta-Analysis of Unadjusted and Adjusted Data. Front Genet 2019; 10:179. [PMID: 30915104 PMCID: PMC6422869 DOI: 10.3389/fgene.2019.00179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 02/19/2019] [Indexed: 01/11/2023] Open
Abstract
Objective: Chronic periodontitis (CP) is a growing problem that affects the worldwide population, having significant impacts on people's daily lives and economic development. Genetics is an important component in the determination of individual susceptibility to periodontal diseases. Numerous studies have been performed to investigate the association between beta defensin 1 (DEFB1) rs11362 polymorphism and risk of CP, but the results are still inconclusive. Therefore, we conducted this meta-analysis to ascertain whether this variation in DEFB1 is associated with CP susceptibility. Methods: The relevant studies were searched in PubMed and Chinese National Knowledge Infrastructure (CNKI) databases up to January 9, 2018. Two independent authors selected citations and extracted the data from eligible studies. Odds ratios (ORs) with their 95% confidence intervals (95% CIs) were used to assess the strength of the association. Results: Seven case-control studies were included in this meta-analysis. Based on unadjusted data, there was no obvious association between DEFB1 rs11362 polymorphism and CP risk in all genetic models (A vs. G: OR = 0.86, 95%CI = 0.61–1.20; AA vs. GG: OR = 0.83, 95% CI = 00.50–1.39; AG vs. GG: OR = 1.01, 95%CI = 0.73–1.39; AG+AA vs. GG: OR = 0.91, 95% CI = 00.74–1.11; and AA vs. AG+GG: OR = 0.83, 95% CI = 00.57–1.21); the results of adjusted data also showed no significant relationship. Subgroup analyses based on ethnicity, participants' smoking status, HWE in controls and severity of CP all revealed similar results to that of the overall analysis. Sensitivity analysis indicated the results were robust and no evidence of publication bias was found. Conclusions: Our meta-analysis suggests that DEFB1 rs11362 polymorphism may not have an important effect on the risk of CP. Further large-scale and well-designed studies are necessary to validate our conclusion in the future.
Collapse
Affiliation(s)
- Jun Shao
- Department of Stomatology, Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou, China
| | - Miao Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Center for Evidence-Based and Translational Medicine, Wuhan University, Wuhan, China.,Department of Evidence-Based Medicine and Clinical Epidemiology, The Second Clinical College, Wuhan University, Wuhan, China.,Department of Nursing, HOPE School of Nursing, School of Health Sciences, Wuhan University, Wuhan, China
| | - Lan Wu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Center for Evidence-Based and Translational Medicine, Wuhan University, Wuhan, China.,Department of Evidence-Based Medicine and Clinical Epidemiology, The Second Clinical College, Wuhan University, Wuhan, China.,Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiao-Wei Jia
- Department of Stomatology, Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou, China
| | - Ying-Hui Jin
- Department of Stomatology, Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou, China.,Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Center for Evidence-Based and Translational Medicine, Wuhan University, Wuhan, China.,Department of Evidence-Based Medicine and Clinical Epidemiology, The Second Clinical College, Wuhan University, Wuhan, China
| | - Xian-Tao Zeng
- Department of Stomatology, Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou, China.,Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Center for Evidence-Based and Translational Medicine, Wuhan University, Wuhan, China.,Department of Evidence-Based Medicine and Clinical Epidemiology, The Second Clinical College, Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Shang L, Wang T, Tong D, Kang W, Liang Q, Ge S. Prolyl hydroxylases positively regulated LPS-induced inflammation in human gingival fibroblasts via TLR4/MyD88-mediated AKT/NF-κB and MAPK pathways. Cell Prolif 2018; 51:e12516. [PMID: 30091492 PMCID: PMC6528886 DOI: 10.1111/cpr.12516] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Prolyl hydroxylases (PHDs) play essential roles in oxygen-sensing system, whereas the effects of PHDs on inflammation have not been totally uncovered. Our study aimed to investigate the role of PHDs in lipopolysaccharide (LPS)-induced inflammation of human gingival fibroblasts (HGFs) and clarify the potential mechanisms. MATERIALS AND METHODS A pan hydroxylase inhibitor, dimethyloxallyl glycine (DMOG), and RNA interference were used to explore the role of PHDs in inflammation. Cytotoxic effect of DMOG was determined by cell-counting kit-8 and flow cytometry respectively. The secretion levels of IL-6 and IL-8 were assessed by ELISA. The mRNA levels of inflammatory cytokines, Toll-like receptor (TLR) 4 and MyD88 were evaluated by quantitative real-time PCR. The activation of NF-κB, mitogen-activated protein kinase (MAPK) and PI3K/AKT pathways were detected by western blot and the nuclear translocation of NF-κB p65 was examined by immunofluorescence. Downregulation of PHD1 and PHD2 was performed with siRNA transfection. RESULTS Dimethyloxallyl glycine inhibited LPS-induced inflammatory cytokine, TLR4 and MyD88 expression in gene level and the elevated secretion of IL-6 and IL-8 was also downregulated. Additionally, LPS-induced activation of NF-κB, MAPK and AKT pathways was abolished by DMOG treatment. Importantly, LPS-induced inflammatory cytokine expression was merely suppressed by PHD2 knockdown. CONCLUSIONS Prolyl hydroxylases acted as a positive regulator in LPS-induced inflammation of HGFs via TLR4/MyD88-mediated NF-κB, MAPK and AKT signalling pathways and PHD2 among three isoforms was principally responsible for the effects.
Collapse
Affiliation(s)
- Lingling Shang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Ting Wang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Dongdong Tong
- Department of Oral maxillofacial SurgerySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Wenyan Kang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Qianyu Liang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Shaohua Ge
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| |
Collapse
|
16
|
Lu W, Gu JY, Zhang YY, Gong DJ, Zhu YM, Sun Y. Tolerance induced by Porphyromonas gingivalis may occur independently of TLR2 and TLR4. PLoS One 2018; 13:e0200946. [PMID: 30040860 PMCID: PMC6057631 DOI: 10.1371/journal.pone.0200946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/05/2018] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Periodontitis is a microbe-induced chronic inflammatory disease. Previous exposure of the host to bacteria or their virulence factors leads to refractory responses to further stimuli, which is called tolerance. Porphyromonas gingivalis (P. gingivalis) is one of the most important pathogenic microorganisms associated with periodontitis, and is a potent inducer of pro- and anti-inflammatory cytokines. The aim of this study was to explore the roles and possible mechanisms of tolerance induced by P. gingivalis. METHODS THP-1-derived macrophages were pretreated with 1x108 colony-forming units/ml P. gingivalis ATCC 33277 or 21 clinical isolates from moderate to severe chronic periodontitis patients (24 h), washed (2 h) and treated with P. gingivalis ATCC 33277 or the same clinical isolates again (24 h). Levels of pro-inflammatory cytokines TNF-α and IL-1β and anti-inflammatory cytokine IL-10 in supernatants were detected by ELISA. Moreover, to identify the possible mechanisms for the changes in cytokine secretion, Toll-like receptor 2 (TLR2) and TLR4 protein expressions were explored in these cells by flow cytometry. RESULTS After repeated challenge with P. gingivalis ATCC 33277 or clinical isolates, production of TNF-α and IL-1β in macrophages was decreased significantly compared with that following a single stimulation (p<0.05), while only comparable levels of IL-10 were detected in P. gingivalis ATCC 33277 or clinical isolate-tolerized cells (p>0.05). In addition, there was interstrain variability in the ability to induce IL-1β and IL-10 production after repeated P. gingivalis stimulation. However, no significant changes in TLR2 or TLR4 were detected in macrophages that were repeatedly treated with P. gingivalis ATCC 33277 or clinical isolates compared with those stimulated with P. gingivalis only once (p>0.05). CONCLUSIONS Repeated P. gingivalis stimulation triggered tolerance, which might contribute to limiting periodontal inflammation. However, tolerance induced by P. gingivalis might develop independently of TLR2 and TLR4 and be related to molecules in signaling pathways downstream of TLR2 and TLR4.
Collapse
Affiliation(s)
- Wei Lu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jian-yu Gu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yao-yao Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Dan-Jun Gong
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Department of Stomatology, Suzhou Hospital, Suzhou, China
| | - Yi-ming Zhu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Ying Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- * E-mail:
| |
Collapse
|
17
|
Phenotypic Variation during Biofilm Formation: Implications for Anti-Biofilm Therapeutic Design. MATERIALS 2018; 11:ma11071086. [PMID: 29949876 PMCID: PMC6073711 DOI: 10.3390/ma11071086] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022]
Abstract
Various bacterial species cycle between growth phases and biofilm formation, of which the latter facilitates persistence in inhospitable environments. These phases can be generally characterized by one or more cellular phenotype(s), each with distinct virulence factor functionality. In addition, a variety of phenotypes can often be observed within the phases themselves, which can be dependent on host conditions or the presence of nutrient and oxygen gradients within the biofilm itself (i.e., microenvironments). Currently, most anti-biofilm strategies have targeted a single phenotype; this approach has driven effective, yet incomplete, protection due to the lack of consideration of gene expression dynamics throughout the bacteria’s pathogenesis. As such, this article provides an overview of the distinct phenotypes found within each biofilm development phase and demonstrates the unique anti-biofilm solutions each phase offers. However, we conclude that a combinatorial approach must be taken to provide complete protection against biofilm forming bacterial and their resulting diseases.
Collapse
|