1
|
de Carvalho KLK, Porto AN, Aranha AMF, Freitas GP, Volpato LER. Evaluation of A. actinomycetemcomitans and P. gingivalis from the mouth of patients irradiated in the head and neck region: a cross-sectional study. Support Care Cancer 2024; 32:770. [PMID: 39496978 DOI: 10.1007/s00520-024-08970-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: 07/18/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024]
Abstract
PURPOSE This study aimed to quantify Aggregatibacter actinomycetemcomitans (A.a) and Porphyromonas gingivalis (P.g) from the mouth of head and neck irradiated and cancer-free patients. METHODS Information such as age, presence of tongue coating, salivary flow, and biofilm were collected from head and neck irradiated patients (Group 1) and compared the results with a group of cancer-free individuals (Group 2). The presence of tongue coating was clinically examined. Sialometry was performed through a stimulating technique by chewing paraffin. Microbiological samples were collected from buccal and labial mucosa and tongue dorsum. Subsequently, the samples were processed and analyzed by qPCR to detect the presence and quantify the bacteria. RESULTS There was a statistical difference in the quantity of bacteria among the 24 individuals in Group 1 (A.a, 2817 ± 8718; P.g, 3145 ± 11297) and 26 individuals in Group 2 (A.a, 133996 ± 398545; P.g, 60 ± 195) regarding tongue coating (Group 1, A.a 2194.6 ± 4641.5; Group 2, A.a 92767.8 ± 333385.7) and salivary volume (Group 1, 0.69 mL; Group 2, 3.09 mL). The linear regression analysis found that the variable group was the main responsible for the difference in the quantity of periodontal pathogens (p-value < 0.001). There was no statistical difference in the amount of Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis between totally edentulous and partially edentulous (with 12 or fewer teeth) patients. CONCLUSION Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis were present in significant amounts in patients of both groups, with a greater quantity in cancer-free individuals.
Collapse
Affiliation(s)
- Karine Lima Kido de Carvalho
- Research Program On Integrated Dental Sciences, School of Dentistry, University of Cuiabá, Avenida Manoel José de Arruda 3100, Jardim Europa, Cuiabá, MT, 78065-900, Brazil
| | - Alessandra Nogueira Porto
- Research Program On Integrated Dental Sciences, School of Dentistry, University of Cuiabá, Avenida Manoel José de Arruda 3100, Jardim Europa, Cuiabá, MT, 78065-900, Brazil
| | - Andreza Maria Fabio Aranha
- Research Program On Integrated Dental Sciences, School of Dentistry, University of Cuiabá, Avenida Manoel José de Arruda 3100, Jardim Europa, Cuiabá, MT, 78065-900, Brazil
| | - Gileade Pereira Freitas
- Research Program On Integrated Dental Sciences, School of Dentistry, University of Cuiabá, Avenida Manoel José de Arruda 3100, Jardim Europa, Cuiabá, MT, 78065-900, Brazil.
| | - Luiz Evaristo Ricci Volpato
- Research Program On Integrated Dental Sciences, School of Dentistry, University of Cuiabá, Avenida Manoel José de Arruda 3100, Jardim Europa, Cuiabá, MT, 78065-900, Brazil
- Department of Dentistry, Hospital de Câncer de Mato Grosso, Cuiabá, MT, Brazil
| |
Collapse
|
2
|
Catalan EA, Seguel-Fuentes E, Fuentes B, Aranguiz-Varela F, Castillo-Godoy DP, Rivera-Asin E, Bocaz E, Fuentes JA, Bravo D, Schinnerling K, Melo-Gonzalez F. Oral Pathobiont-Derived Outer Membrane Vesicles in the Oral-Gut Axis. Int J Mol Sci 2024; 25:11141. [PMID: 39456922 PMCID: PMC11508520 DOI: 10.3390/ijms252011141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Oral pathobionts are essential in instigating local inflammation within the oral cavity and contribute to the pathogenesis of diseases in the gastrointestinal tract and other distant organs. Among the Gram-negative pathobionts, Porphyromonas gingivalis and Fusobacterium nucleatum emerge as critical drivers of periodontitis, exerting their influence not only locally but also as inducers of gut dysbiosis, intestinal disturbances, and systemic ailments. This dual impact is facilitated by their ectopic colonization of the intestinal mucosa and the subsequent mediation of distal systemic effects by releasing outer membrane vesicles (OMVs) into circulation. This review elucidates the principal components of oral pathobiont-derived OMVs implicated in disease pathogenesis within the oral-gut axis, detailing virulence factors that OMVs carry and their interactions with host epithelial and immune cells, both in vitro and in vivo. Additionally, we shed light on the less acknowledged interplay between oral pathobionts and the gut commensal Akkermansia muciniphila, which can directly impede oral pathobionts' growth and modulate bacterial gene expression. Notably, OMVs derived from A. muciniphila emerge as promoters of anti-inflammatory effects within the gastrointestinal and distant tissues. Consequently, we explore the potential of A. muciniphila-derived OMVs to interact with oral pathobionts and prevent disease in the oral-gut axis.
Collapse
Affiliation(s)
- Eduardo A. Catalan
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Emilio Seguel-Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Brandon Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Aranguiz-Varela
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Daniela P. Castillo-Godoy
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elizabeth Rivera-Asin
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elisa Bocaz
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile;
| | - Denisse Bravo
- Cellular Interactions Laboratory, Faculty of Dentistry, Universidad Andrés Bello, Santiago 8370133, Chile;
| | - Katina Schinnerling
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Melo-Gonzalez
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| |
Collapse
|
3
|
Stocke K, Lamont G, Tan J, Scott DA. Delineation of global, absolutely essential and conditionally essential pangenomes of Porphyromonas gingivalis. Sci Rep 2024; 14:22247. [PMID: 39333542 PMCID: PMC11436796 DOI: 10.1038/s41598-024-72451-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/06/2024] [Indexed: 09/29/2024] Open
Abstract
Porphyromonas gingivalis is a Gram-negative, anaerobic oral pathobiont, an etiological agent of periodontitis and the most commonly studied periodontal bacterium. Multiple low passage clinical isolates were sequenced, and their genomes compared to several laboratory strains. Phylogenetic distances were mapped, a gene absence-presence matrix generated, and core (present in all genomes) and accessory (absent in one or more genomes) genes delineated. Subsequently, a second pangenome delineating the prevalence of inherently essential genes was generated. The prevalence of genes conditionally essential for surviving tobacco exposure, abscess formation and epithelial invasion was also determined, in addition to genes encoding key proteolytic enzymes containing putative signal peptides. While the absolutely essential pangenome was highly conserved, significant differences in the complete and conditionally essential pangenomes were apparent. Thus, genetic plasticity appears to lie primarily in gene sets facilitating adaptation to variant disease-related environments. Those genes that are highly pervasive in the P. gingivalis absolutely essential pangenome or are highly prevalent and essential for fitness in disease-relevant models, may represent particularly attractive therapeutic targets worthy of further investigation. As mutations in absolutely essential genes are expected to be lethal, the data provided herein should also facilitate improved planning for P. gingivalis gene mutation strategies.
Collapse
Affiliation(s)
- Kendall Stocke
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, 501 S. Preston St., Louisville, KY, 40292, USA
| | - Gwyneth Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, 501 S. Preston St., Louisville, KY, 40292, USA
| | - Jinlian Tan
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, 501 S. Preston St., Louisville, KY, 40292, USA
| | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, 501 S. Preston St., Louisville, KY, 40292, USA.
- Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville, Louisville, KY, 40292, USA.
| |
Collapse
|
4
|
Lithgow KV, Buchholz VCH, Ku E, Konschuh S, D'Aubeterre A, Sycuro LK. Protease activities of vaginal Porphyromonas species disrupt coagulation and extracellular matrix in the cervicovaginal niche. NPJ Biofilms Microbiomes 2022; 8:8. [PMID: 35190575 PMCID: PMC8861167 DOI: 10.1038/s41522-022-00270-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
Porphyromonas asaccharolytica and Porphyromonas uenonis are common inhabitants of the vaginal microbiome, but their presence has been linked to adverse health outcomes for women, including bacterial vaginosis and preterm birth. However, little is known about the pathogenesis mechanisms of these bacteria. The related oral opportunistic pathogen, Porphyromonas gingivalis, is comparatively well-studied and known to secrete numerous extracellular matrix-targeting proteases. Among these are the gingipain family of cysteine proteases that drive periodontal disease progression and hematogenic transmission to the placenta. In this study, we demonstrate that vaginal Porphyromonas species secrete broad-acting proteases capable of freely diffusing within the cervicovaginal niche. These proteases degrade collagens that are enriched within the cervix (type I) and chorioamniotic membranes (type IV), as well as fibrinogen, which inhibits clot formation. Bioinformatic queries confirmed the absence of gingipain orthologs and identified five serine, cysteine, and metalloprotease candidates in each species. Inhibition assays revealed that each species' proteolytic activity can be partially attributed to a secreted metalloprotease with broad substrate specificity that is distantly related to the P. gingivalis endopeptidase PepO. This characterization of virulence activities in vaginal Porphyromonas species highlights their potential to alter the homeostasis of reproductive tissues and harm human pregnancy through clotting disruption, fetal membrane weakening, and premature cervical remodeling.
Collapse
Affiliation(s)
- Karen V Lithgow
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Vienna C H Buchholz
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Emily Ku
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Shaelen Konschuh
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Ana D'Aubeterre
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Laura K Sycuro
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
- International Microbiome Centre, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
5
|
Crooks TA, Madison JD, Walsh DM, Herbert WG, Jeraldo PR, Chia N, Cliby WA, Kaufmann SH, Walther-Antonio MRS. Porphyromonas somerae Invasion of Endometrial Cancer Cells. Front Microbiol 2021; 12:674835. [PMID: 34367083 PMCID: PMC8343132 DOI: 10.3389/fmicb.2021.674835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022] Open
Abstract
Recent evidence suggests an association between endometrial cancer and the understudied bacterial species Porphyromonas somerae. This association was demonstrated in previous work that indicated a significantly enriched abundance of P. somerae in the uterine microbiome of endometrial cancer patients. Given the known associations of the Porphyromonas genus and oral cancer, we hypothesized that P. somerae may play a similar pathogenic role in endometrial cancer via intracellular activity. Before testing our hypothesis, we first characterized P. somerae biology, as current background data is limited. These novel characterizations include growth curves in liquid medium and susceptibility tests to antibiotics. We tested our hypothesis by examining growth changes in response to 17β-estradiol, a known risk factor for endometrial cancer, followed by metabolomic profiling in the presence and absence of 17β-estradiol. We found that P. somerae exhibits increased growth in the presence of 17β-estradiol of various concentrations. However, we did not find significant changes in metabolite levels in response to 17β-estradiol. To study direct host-microbe interactions, we used in vitro invasion assays under hypoxic conditions and found evidence for intracellular invasion of P. somerae in endometrial adenocarcinoma cells. We also examined these interactions in the presence of 17β-estradiol but did not observe changes in invasion frequency. Invasion was shown using three lines of evidence including visualization via differential staining and brightfield microscopy, increased frequency of bacterial recovery after co-culturing, and in silico methods to detail relevant genomic and transcriptomic components. These results underscore potential intracellular phenotypes of P. somerae within the uterine microbiome. Furthermore, these results raise new questions pertaining to the role of P. somerae in the progression of endometrial cancer.
Collapse
Affiliation(s)
- Taylor A Crooks
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Microbiology and Immunology, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Joseph D Madison
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Biology Department, University of Massachusetts Boston, Boston, MA, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Dana M Walsh
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
| | - William G Herbert
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Patricio R Jeraldo
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Nicholas Chia
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - William A Cliby
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| | - Scott H Kaufmann
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Marina R S Walther-Antonio
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Crooks TA, Madison JD, Walsh DM, Herbert WG, Jeraldo PR, Chia N, Cliby WA, Kaufmann SH, Walther-Antonio MRS. Porphyromonas somerae Invasion of Endometrial Cancer Cells. Front Microbiol 2021. [PMID: 34367083 DOI: 10.3389/fmicb.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
Recent evidence suggests an association between endometrial cancer and the understudied bacterial species Porphyromonas somerae. This association was demonstrated in previous work that indicated a significantly enriched abundance of P. somerae in the uterine microbiome of endometrial cancer patients. Given the known associations of the Porphyromonas genus and oral cancer, we hypothesized that P. somerae may play a similar pathogenic role in endometrial cancer via intracellular activity. Before testing our hypothesis, we first characterized P. somerae biology, as current background data is limited. These novel characterizations include growth curves in liquid medium and susceptibility tests to antibiotics. We tested our hypothesis by examining growth changes in response to 17β-estradiol, a known risk factor for endometrial cancer, followed by metabolomic profiling in the presence and absence of 17β-estradiol. We found that P. somerae exhibits increased growth in the presence of 17β-estradiol of various concentrations. However, we did not find significant changes in metabolite levels in response to 17β-estradiol. To study direct host-microbe interactions, we used in vitro invasion assays under hypoxic conditions and found evidence for intracellular invasion of P. somerae in endometrial adenocarcinoma cells. We also examined these interactions in the presence of 17β-estradiol but did not observe changes in invasion frequency. Invasion was shown using three lines of evidence including visualization via differential staining and brightfield microscopy, increased frequency of bacterial recovery after co-culturing, and in silico methods to detail relevant genomic and transcriptomic components. These results underscore potential intracellular phenotypes of P. somerae within the uterine microbiome. Furthermore, these results raise new questions pertaining to the role of P. somerae in the progression of endometrial cancer.
Collapse
Affiliation(s)
- Taylor A Crooks
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Microbiology and Immunology, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Joseph D Madison
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Biology Department, University of Massachusetts Boston, Boston, MA, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Dana M Walsh
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
| | - William G Herbert
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Patricio R Jeraldo
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Nicholas Chia
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - William A Cliby
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| | - Scott H Kaufmann
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Marina R S Walther-Antonio
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
7
|
Miller DP, Scott DA. Inherently and Conditionally Essential Protein Catabolism Genes of Porphyromonas gingivalis. Trends Microbiol 2020; 29:54-64. [PMID: 33071035 DOI: 10.1016/j.tim.2020.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/01/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Proteases are critical virulence determinants of Porphyromonas gingivalis, an emerging Alzheimer's disease, cancer, and arthritis pathogen and established agent of periodontitis. Transposon sequencing has been employed to define the core essential genome of this bacterium and genes conditionally essential in multiple environments - abscess formation; epithelial colonization; and cigarette smoke toxin exposure; as well as to elucidate genes required for iron acquisition and a functional type 9 secretion system. Validated and predicted protein catabolism genes identified include a combination of established virulence factors and a larger set of seemingly more mundane proteolytic genes. The functions and relevance of genes that share essentiality in multiple disease-relevant conditions are examined. These common stress-related genes may represent particularly attractive therapeutic targets for the control of P. gingivalis infections.
Collapse
Affiliation(s)
- Daniel P Miller
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
| | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
8
|
Gao L, Ma Y, Li X, Zhang L, Zhang C, Chen Q, Zhao C. Research on the roles of genes coding ATP‐binding cassette transporters in
Porphyromonas gingivalis
pathogenicity. J Cell Biochem 2019; 121:93-102. [PMID: 31081181 DOI: 10.1002/jcb.28887] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Li Gao
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Yuanyuan Ma
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
- Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
| | - Xiting Li
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Liping Zhang
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Chi Zhang
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Qianying Chen
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Chuanjiang Zhao
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| |
Collapse
|
9
|
Menzel LP, Ruddick W, Chowdhury MH, Brice DC, Clance R, Porcelli E, Ryan LK, Lee J, Yilmaz Ö, Kirkwood KL, McMahon L, Tran A, Diamond G. Activation of vitamin D in the gingival epithelium and its role in gingival inflammation and alveolar bone loss. J Periodontal Res 2019; 54:444-452. [PMID: 30802957 DOI: 10.1111/jre.12646] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/15/2019] [Accepted: 02/06/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND OBJECTIVE Both chronic and aggressive periodontal disease are associated with vitamin D deficiency. The active form of vitamin D, 1,25(OH)2 D3 , induces the expression of the antimicrobial peptide LL-37 and innate immune mediators in cultured human gingival epithelial cells (GECs). The aim of this study was to further delineate the mechanism by which vitamin D enhances the innate defense against the development of periodontal disease (PD). MATERIALS AND METHODS Wild-type C57Bl/6 mice were made deficient in vitamin D by dietary restriction. Cultured primary and immortalized GEC were stimulated with 1,25(OH)2 D3 , followed by infection with Porphyromonas gingivalis, and viable intracellular bacteria were quantified. Conversion of vitamin D3 to 25(OH)D3 and 1,25(OH)2 D3 was quantified by ELISA. Effect of vitamin D on basal IL-1α expression in mice was determined by topical administration to the gingiva of wild-type mice, followed by qRT-PCR. RESULTS Dietary restriction of vitamin D led to alveolar bone loss and increased inflammation in the gingiva in the mouse model. In primary human GEC and established human cell lines, treatment of GEC with 1,25(OH)2 D3 inhibited the intracellular growth of P. gingivalis. Cultured GEC expressed two 25-hydroxylases (CYP27A1 and CYP2R1), as well as 1-α hydroxylase, enabling conversion of vitamin D to both 25(OH)D3 and 1,25(OH)2 D3 . Topical application of both vitamin D3 and 1,25(OH)2 D3 to the gingiva of mice led to rapid inhibition of IL-1α expression, a prominent pro-inflammatory cytokine associated with inflammation, which also exhibited more than a 2-fold decrease from basal levels in OKF6/TERT1 cells upon 1,25(OH)2 D3 treatment, as determined by RNA-seq. CONCLUSION Vitamin D deficiency in mice contributes to PD, recapitulating the association seen in humans, and provides a unique model to study the development of PD. Vitamin D increases the activity of GEC against the invasion of periodontal pathogens and inhibits the inflammatory response, both in vitro and in vivo. GEC can convert inactive vitamin D to the active form in situ, supporting the hypothesis that vitamin D can be applied directly to the gingiva to prevent or treat periodontal disease.
Collapse
Affiliation(s)
- Lorenzo P Menzel
- Department of Oral Biology, University of Florida, Gainesville, Florida
| | - Willam Ruddick
- Department of Oral Biology, University of Florida, Gainesville, Florida
| | | | - David C Brice
- Department of Oral Biology, University of Florida, Gainesville, Florida
| | - Ryan Clance
- Department of Oral Biology, University of Florida, Gainesville, Florida
| | - Emily Porcelli
- Department of Oral Biology, University of Florida, Gainesville, Florida
| | - Lisa K Ryan
- Division of Infectious Diseases and Global Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Jungnam Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida
| | - Özlem Yilmaz
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, South Carolina.,Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Keith L Kirkwood
- Department of Oral Biology, State University of New York at Buffalo, Buffalo, New York
| | - Laura McMahon
- Department of Oral Biology, Rutgers New Jersey Dental School, Newark, New Jersey
| | - Amy Tran
- Department of Oral Biology, Rutgers New Jersey Dental School, Newark, New Jersey
| | - Gill Diamond
- Department of Oral Biology, University of Florida, Gainesville, Florida
| |
Collapse
|
10
|
Ebersole JL, Kirakodu S, Novak MJ, Orraca L, Stormberg AJ, Gonzalez-Martinez J, Burgos A, Gonzalez OA. Comparative analysis of expression of microbial sensing molecules in mucosal tissues with periodontal disease. Immunobiology 2018; 224:196-206. [PMID: 30470434 DOI: 10.1016/j.imbio.2018.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/13/2018] [Indexed: 12/30/2022]
Abstract
Host-derived pattern recognition receptors (PRRs) are necessary for effective innate immune engagement of pathogens that express microbial-associated molecular patterns (MAMP) ligands for these PRRs. This study used a nonhuman primate model to evaluate the expression of these sensing molecules in gingival tissues. Macaca mulatta aged 12-24 with a healthy periodontium (n = 13) or periodontitis (n = 11) provided gingival tissues for assessment of naturally-occurring periodontitis. An additional group of animals (12-23 years; n = 18) was subjected to a 5 month longitudinal study examining the initiation and progression of periodontitis, RNA was isolated and microarray analysis conducted for gene expression of the sensing PRRs. The results demonstrated increased expression of various PRRs in naturally-occurring established periodontitis. Selected PRRs also correlated with both bleeding on probing (BOP) and pocket depth (PD) in the animals. The longitudinal model demonstrated multiple TLRs, as well as selected other PRRs that were significantly increased by 2 weeks during initiation of the lesion. While gene expression levels of various PRRs correlated with BOP and PD at baseline and resolution of disease, few correlated with these clinical parameters during initiation and progression of the lesion. These findings suggest that the levels of various PRRs are affected in established periodontitis lesions, and that PRR expression increased most dramatically during the initiation of the disease process, presumably in response to the juxtaposed microbial challenge to the tissues and goal of reestablishing homeostasis.
Collapse
Affiliation(s)
- J L Ebersole
- Center for Oral Health Research, University of Kentucky, Lexington, KY, United States.
| | - S Kirakodu
- Center for Oral Health Research, University of Kentucky, Lexington, KY, United States
| | - M J Novak
- Center for Oral Health Research, University of Kentucky, Lexington, KY, United States
| | - L Orraca
- School of Dental Medicine, University of Puerto Rico, San Juan, PR, United States
| | - A J Stormberg
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - J Gonzalez-Martinez
- Caribbean Primate Research Center, University of Puerto Rico, Toa Baja, PR, United States
| | - A Burgos
- Caribbean Primate Research Center, University of Puerto Rico, Toa Baja, PR, United States
| | - O A Gonzalez
- Center for Oral Health Research, University of Kentucky, Lexington, KY, United States; Division of Periodontics, College of Dentistry, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
11
|
Miller DP, Hutcherson JA, Wang Y, Nowakowska ZM, Potempa J, Yoder-Himes DR, Scott DA, Whiteley M, Lamont RJ. Genes Contributing to Porphyromonas gingivalis Fitness in Abscess and Epithelial Cell Colonization Environments. Front Cell Infect Microbiol 2017; 7:378. [PMID: 28900609 PMCID: PMC5581868 DOI: 10.3389/fcimb.2017.00378] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
Porphyromonas gingivalis is an important cause of serious periodontal diseases, and is emerging as a pathogen in several systemic conditions including some forms of cancer. Initial colonization by P. gingivalis involves interaction with gingival epithelial cells, and the organism can also access host tissues and spread haematogenously. To better understand the mechanisms underlying these properties, we utilized a highly saturated transposon insertion library of P. gingivalis, and assessed the fitness of mutants during epithelial cell colonization and survival in a murine abscess model by high-throughput sequencing (Tn-Seq). Transposon insertions in many genes previously suspected as contributing to virulence showed significant fitness defects in both screening assays. In addition, a number of genes not previously associated with P. gingivalis virulence were identified as important for fitness. We further examined fitness defects of four such genes by generating defined mutations. Genes encoding a carbamoyl phosphate synthetase, a replication-associated recombination protein, a nitrosative stress responsive HcpR transcription regulator, and RNase Z, a zinc phosphodiesterase, showed a fitness phenotype in epithelial cell colonization and in a competitive abscess infection. This study verifies the importance of several well-characterized putative virulence factors of P. gingivalis and identifies novel fitness determinants of the organism.
Collapse
Affiliation(s)
- Daniel P Miller
- Department of Oral Immunology and Infectious Diseases, University of LouisvilleLouisville, KY, United States
| | - Justin A Hutcherson
- Department of Oral Immunology and Infectious Diseases, University of LouisvilleLouisville, KY, United States
| | - Yan Wang
- Department of Oral Immunology and Infectious Diseases, University of LouisvilleLouisville, KY, United States
| | - Zuzanna M Nowakowska
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases, University of LouisvilleLouisville, KY, United States.,Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland.,Malopolska Centre of Biotechnology, Jagiellonian UniversityKrakow, Poland
| | | | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of LouisvilleLouisville, KY, United States
| | - Marvin Whiteley
- Department of Molecular Biosciences, University of Texas at AustinAustin, TX, United States
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of LouisvilleLouisville, KY, United States
| |
Collapse
|
12
|
Lasica AM, Ksiazek M, Madej M, Potempa J. The Type IX Secretion System (T9SS): Highlights and Recent Insights into Its Structure and Function. Front Cell Infect Microbiol 2017. [PMID: 28603700 DOI: 10.3389/fcimb.2017.00215.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Protein secretion systems are vital for prokaryotic life, as they enable bacteria to acquire nutrients, communicate with other species, defend against biological and chemical agents, and facilitate disease through the delivery of virulence factors. In this review, we will focus on the recently discovered type IX secretion system (T9SS), a complex translocon found only in some species of the Bacteroidetes phylum. T9SS plays two roles, depending on the lifestyle of the bacteria. It provides either a means of movement (called gliding motility) for peace-loving environmental bacteria or a weapon for pathogens. The best-studied members of these two groups are Flavobacterium johnsoniae, a commensal microorganism often found in water and soil, and Porphyromonas gingivalis, a human oral pathogen that is a major causative agent of periodontitis. In P. gingivalis and some other periodontopathogens, T9SS translocates proteins, especially virulence factors, across the outer membrane (OM). Proteins destined for secretion bear a conserved C-terminal domain (CTD) that directs the cargo to the OM translocon. At least 18 proteins are involved in this still enigmatic process, with some engaged in the post-translational modification of T9SS cargo proteins. Upon translocation across the OM, the CTD is removed by a protease with sortase-like activity and an anionic LPS is attached to the newly formed C-terminus. As a result, a cargo protein could be secreted into the extracellular milieu or covalently attached to the bacterial surface. T9SS is regulated by a two-component system; however, the precise environmental signal that triggers it has not been identified. Exploring unknown systems contributing to bacterial virulence is exciting, as it may eventually lead to new therapeutic strategies. During the past decade, the major components of T9SS were identified, as well as hints suggesting the possible mechanism of action. In addition, the list of characterized cargo proteins is constantly growing. The actual structure of the translocon, situated in the OM of bacteria, remains the least explored area; however, new technical approaches and increasing scientific attention have resulted in a growing body of data. Therefore, we present a compact up-to-date review of this topic.
Collapse
Affiliation(s)
- Anna M Lasica
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of DentistryLouisville, KY, United States.,Department of Bacterial Genetics, Faculty of Biology, Institute of Microbiology, University of WarsawWarsaw, Poland
| | - Miroslaw Ksiazek
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of DentistryLouisville, KY, United States.,Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Mariusz Madej
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of DentistryLouisville, KY, United States.,Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian UniversityKrakow, Poland
| |
Collapse
|
13
|
Lasica AM, Ksiazek M, Madej M, Potempa J. The Type IX Secretion System (T9SS): Highlights and Recent Insights into Its Structure and Function. Front Cell Infect Microbiol 2017; 7:215. [PMID: 28603700 PMCID: PMC5445135 DOI: 10.3389/fcimb.2017.00215] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/11/2017] [Indexed: 12/11/2022] Open
Abstract
Protein secretion systems are vital for prokaryotic life, as they enable bacteria to acquire nutrients, communicate with other species, defend against biological and chemical agents, and facilitate disease through the delivery of virulence factors. In this review, we will focus on the recently discovered type IX secretion system (T9SS), a complex translocon found only in some species of the Bacteroidetes phylum. T9SS plays two roles, depending on the lifestyle of the bacteria. It provides either a means of movement (called gliding motility) for peace-loving environmental bacteria or a weapon for pathogens. The best-studied members of these two groups are Flavobacterium johnsoniae, a commensal microorganism often found in water and soil, and Porphyromonas gingivalis, a human oral pathogen that is a major causative agent of periodontitis. In P. gingivalis and some other periodontopathogens, T9SS translocates proteins, especially virulence factors, across the outer membrane (OM). Proteins destined for secretion bear a conserved C-terminal domain (CTD) that directs the cargo to the OM translocon. At least 18 proteins are involved in this still enigmatic process, with some engaged in the post-translational modification of T9SS cargo proteins. Upon translocation across the OM, the CTD is removed by a protease with sortase-like activity and an anionic LPS is attached to the newly formed C-terminus. As a result, a cargo protein could be secreted into the extracellular milieu or covalently attached to the bacterial surface. T9SS is regulated by a two-component system; however, the precise environmental signal that triggers it has not been identified. Exploring unknown systems contributing to bacterial virulence is exciting, as it may eventually lead to new therapeutic strategies. During the past decade, the major components of T9SS were identified, as well as hints suggesting the possible mechanism of action. In addition, the list of characterized cargo proteins is constantly growing. The actual structure of the translocon, situated in the OM of bacteria, remains the least explored area; however, new technical approaches and increasing scientific attention have resulted in a growing body of data. Therefore, we present a compact up-to-date review of this topic.
Collapse
Affiliation(s)
- Anna M Lasica
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of DentistryLouisville, KY, United States.,Department of Bacterial Genetics, Faculty of Biology, Institute of Microbiology, University of WarsawWarsaw, Poland
| | - Miroslaw Ksiazek
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of DentistryLouisville, KY, United States.,Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Mariusz Madej
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of DentistryLouisville, KY, United States.,Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian UniversityKrakow, Poland
| |
Collapse
|
14
|
Krueger J, Pohl S, Preusse M, Kordes A, Rugen N, Schniederjans M, Pich A, Häussler S. Unravelling post-transcriptional PrmC-dependent regulatory mechanisms in Pseudomonas aeruginosa. Environ Microbiol 2016; 18:3583-3592. [PMID: 27376486 DOI: 10.1111/1462-2920.13435] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/28/2016] [Indexed: 02/04/2023]
Abstract
Transcriptional regulation has a central role in cellular adaptation processes and is well investigated. In contrast, the importance of the post-transcriptional regulation on these processes is less well defined. The technological advancements have been critical to precisely quantify protein and mRNA level changes and hold promise to provide more insights into how post-transcriptional regulation determines phenotypes. In Pseudomonas aeruginosa the methyltransferase PrmC methylates peptide chain release factors to facilitate translation termination. Loss of PrmC activity abolishes anaerobic growth and leads to reduced production of quorum sensing-associated virulence factors. Here, by applying SILAC technology in combination with mRNA-sequencing, they provide evidence that the P. aeruginosa phenotype can be attributed to a change in protein to mRNA ratios of selected protein groups. The UAG-dependent translation termination was more dependent on PrmC activity than the UAA- and UGA-dependent translation termination. Additionally, a bias toward UAG stop codons in global transcriptional regulators was found. The finding that this bias in stop codon usage determines the P. aeruginosa phenotype is unexpected and adds complexity to regulatory circuits. Via modulation of PrmC activity the bacterial cell can cross-regulate targets independently of transcriptional signals, a process with an underestimated impact on the bacterial phenotype.
Collapse
Affiliation(s)
- Jonas Krueger
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Research Core Unit Proteomics and Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Sarah Pohl
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany
| | - Matthias Preusse
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany
| | - Adrian Kordes
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Nils Rugen
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Monika Schniederjans
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany
| | - Andreas Pich
- Research Core Unit Proteomics and Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Susanne Häussler
- Institute for Molecular Bacteriology, TWINCORE GmbH, Centre for Experimental and Clinical Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany. .,Department of Molecular Bacteriology, Helmholtz for Centre Infection Research, Braunschweig, Germany.
| |
Collapse
|
15
|
Zhao J, Li Q, Pan CL, Liu JC, Wang HY, Tan LS, Pan YP. Gene expression changes in Porphyromonas gingivalis W83 after inoculation in rat oral cavity. BMC Microbiol 2015; 15:111. [PMID: 26001932 PMCID: PMC4493947 DOI: 10.1186/s12866-015-0438-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 05/05/2015] [Indexed: 12/02/2022] Open
Abstract
Background The development of chronic periodontitis was due to not only periodontal pathogens, but also the interaction between periodontal pathogens and host. The aim of this study is to investigate the alterations in gene expression in Porphyromonas gingivalis (P.gingivalis) W83 after inoculation in rat oral cavity. Results P.gingivalis W83 inoculation in rat oral cavity caused inflammatory responses in gingival tissues and destroyed host alveolar bone. Microarray analysis revealed that 42 genes were upregulated, and 22 genes were downregulated in the detected 1786 genes in the inoculated P.gingivalis W83. Real-time quantitative PCR detection confirmed the expression alterations in some selected genes. Products of these upregulated and downregulated genes are mainly related to transposon functions, cell transmembrane transportation, protein and nucleic acid metabolism, energy metabolism, cell division and bacterial pathogenicity. Conclusions P.gingivalis W83 has a pathogenic effect on host oral cavity. Meanwhile, inflammatory oral environment alters P.gingivalis W83 gene expression profile. These changes in gene expression may limit the proliferation and weaken the pathogenicity of P.gingivalis W83, and favor themselves to adapt local environment for survival.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Periodontology, School of Stomatology, China Medical University, Shenyang , Liaoning, China.
| | - Qian Li
- Department of Periodontology, School of Stomatology, China Medical University, Shenyang , Liaoning, China.
| | - Chun-Ling Pan
- Department of Periodontology, School of Stomatology, China Medical University, Shenyang , Liaoning, China.
| | - Jun-Chao Liu
- Department of Periodontology, School of Stomatology, China Medical University, Shenyang , Liaoning, China.
| | - Hong-Yan Wang
- Department of Periodontology, School of Stomatology, China Medical University, Shenyang , Liaoning, China.
| | - Li-Si Tan
- Department of Periodontology, School of Stomatology, China Medical University, Shenyang , Liaoning, China.
| | - Ya-Ping Pan
- Department of Periodontology, School of Stomatology, China Medical University, Shenyang , Liaoning, China.
| |
Collapse
|
16
|
Pustelny C, Brouwer S, Müsken M, Bielecka A, Dötsch A, Nimtz M, Häussler S. The peptide chain release factor methyltransferase PrmC is essential for pathogenicity and environmental adaptation of Pseudomonas aeruginosa PA14. Environ Microbiol 2012; 15:597-609. [PMID: 23278968 DOI: 10.1111/1462-2920.12040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 11/02/2012] [Indexed: 01/30/2023]
Abstract
Pseudomonas aeruginosa pathogenicity and its capability to adapt to multiple environments are dependent on the production of diverse virulence factors, controlled by the sophisticated quorum sensing (QS) network of P. aeruginosa. To better understand the molecular mechanisms that underlie this adaptation we searched for novel key regulators of virulence factor production by screening a PA14 transposon mutant library for potential candidates acting downstream of the unique 2-alkyl-4-quinolone (AQ) QS system of P. aeruginosa. We focused the work on a protein named HemK with high homology to PrmC of Escherichia coli displaying a similar enzymatic activity (therefore also referred to as PrmC). In this study, we demonstrate that PrmC is an S-adenosyl-l-methionine (AdoMet)-dependent methyltransferase of peptide chain release factors (RFs) essential for the expression of several virulence factors, such as pyocyanin, rhamnolipids and the type III-secreted toxin ExoT. Furthermore, the PA14_prmC mutant strain is unable to grow under anoxic conditions and has a significantly reduced pathogenicity in the infection model Galleria mellonella. Along with transcriptomic and proteomic analyses, the presented data indicate that the methylation of RFs in P. aeruginosa seems to have a global effect on cellular processes related to the virulence of this nosocomial pathogen.
Collapse
Affiliation(s)
- Christian Pustelny
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.
| | | | | | | | | | | | | |
Collapse
|
17
|
Saiki K, Konishi K. Strategies for targeting the gingipain secretion system of Porphyromonas gingivalis. J Oral Biosci 2012. [DOI: 10.1016/j.job.2012.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
18
|
Gao L, Xu Y, Meng S, Wu Y, Huang H, Su R, Zhao L. Identification of the putative specific pathogenic genes of Porphyromonas gingivalis with type II fimbriae. DNA Cell Biol 2012; 31:1027-37. [PMID: 22257441 DOI: 10.1089/dna.2011.1487] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Porphyromonas gingivalis, the key etiologic agent of periodontitis, can be classified into six types (I to V and Ib) based on the fimA genes that encode FimA (a subunit of fimbriae). Accumulated evidence indicates that P. gingivalis expressing Type II fimbriae (Pg-II) is the most frequent isolate from severe periodontitis cases and is more virulent than other types of P. gingivalis. However, during the Pg-II infection process, which specific virulence factors play the key role is still unclear. In this study, we examined the capabilities of three Pg-II strains to invade and modulate the inflammatory cytokine expression of human gingival epithelial cells (GECs) compared to two Pg-I strains. P. gingivalis oligo microarrays were used to compare gene expression profiles of Pg-II strains that invade GECs with Pg-I strains. The differential gene expression of Pg-II was confirmed by quantitative reverse transcription-polymerase chain reaction. Our results showed that all of the Pg-II strains could induce interleukin (IL)-1β and IL-6 secretion significantly when compared to Pg-I strains. Thirty-seven genes that were specifically expressed during the pathogenic process of Pg-II were identified by a microarray assay. These findings provide a new insight at the molecular level to explain the specific pathogenic mechanism of Pg-II strains.
Collapse
Affiliation(s)
- Li Gao
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Chengdu, PR China
| | | | | | | | | | | | | |
Collapse
|
19
|
Evidence of the adaptive response in Pseudomonas aeruginosa to 14 years of incubation in seawater. ANN MICROBIOL 2011. [DOI: 10.1007/s13213-011-0387-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
20
|
Dolgilevich S, Rafferty B, Luchinskaya D, Kozarov E. Genomic comparison of invasive and rare non-invasive strains reveals Porphyromonas gingivalis genetic polymorphisms. J Oral Microbiol 2011; 3. [PMID: 21541093 PMCID: PMC3086587 DOI: 10.3402/jom.v3i0.5764] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 01/20/2011] [Accepted: 02/03/2011] [Indexed: 11/14/2022] Open
Abstract
Background Porphyromonas gingivalis strains are shown to invade human cells in vitro with different invasion efficiencies, varying by up to three orders of magnitude. Objective We tested the hypothesis that invasion-associated interstrain genomic polymorphisms are present in P. gingivalis and that putative invasion-associated genes can contribute to P. gingivalis invasion. Design Using an invasive (W83) and the only available non-invasive P. gingivalis strain (AJW4) and whole genome microarrays followed by two separate software tools, we carried out comparative genomic hybridization (CGH) analysis. Results We identified 68 annotated and 51 hypothetical open reading frames (ORFs) that are polymorphic between these strains. Among these are surface proteins, lipoproteins, capsular polysaccharide biosynthesis enzymes, regulatory and immunoreactive proteins, integrases, and transposases often with abnormal GC content and clustered on the chromosome. Amplification of selected ORFs was used to validate the approach and the selection. Eleven clinical strains were investigated for the presence of selected ORFs. The putative invasion-associated ORFs were present in 10 of the isolates. The invasion ability of three isogenic mutants, carrying deletions in PG0185, PG0186, and PG0982 was tested. The PG0185 (ragA) and PG0186 (ragB) mutants had 5.1×103-fold and 3.6×103-fold decreased in vitro invasion ability, respectively. Conclusion The annotation of divergent ORFs suggests deficiency in multiple genes as a basis for P. gingivalis non-invasive phenotype.
Collapse
Affiliation(s)
- Svetlana Dolgilevich
- Section Oral and Diagnostic Sciences, Columbia University College of Dental Medicine, New York
| | | | | | | |
Collapse
|
21
|
Zhang D, Zheng H, Zhao J, Lin L, Li C, Liu J, Pan Y. Porphorymonas gingivalis induces intracellular adhesion molecule-1 expression in endothelial cells through the nuclear factor-kappaB pathway, but not through the p38 MAPK pathway. J Periodontal Res 2010; 46:31-8. [DOI: 10.1111/j.1600-0765.2010.01305.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
22
|
Saiki K, Konishi K. Identification of a novel Porphyromonas gingivalis outer membrane protein, PG534, required for the production of active gingipains. FEMS Microbiol Lett 2010; 310:168-74. [DOI: 10.1111/j.1574-6968.2010.02059.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
23
|
Suwannakul S, Stafford GP, Whawell SA, Douglas CWI. Identification of bistable populations of Porphyromonas gingivalis that differ in epithelial cell invasion. MICROBIOLOGY-SGM 2010; 156:3052-3064. [PMID: 20576685 DOI: 10.1099/mic.0.038075-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Bistable populations of bacteria give rise to two or more subtypes that exhibit different phenotypes. We have explored whether the periodontal pathogen Porphyromonas gingivalis exhibits bistable invasive phenotypes. Using a modified cell invasion assay, we show for the first time that there are two distinct subtypes within a population of P. gingivalis strains NCTC 11834 and W50 that display differences in their ability to invade oral epithelial cells. The highly invasive subtype invades cells at 10-30-fold higher levels than the poorly invasive subtype and remains highly invasive for approximately 12-16 generations. Analysis of the gingipain activity of these subtypes revealed that the highly invasive type had reduced cell-associated arginine-specific protease activity. The role of Arg-gingipain activity in invasion was verified by enhancement of invasion by rgpAB mutations and by inclusion of an Arg-gingipain inhibitor in invasion assays using wild-type bacteria. In addition, a population of ΔrgpAB bacteria did not contain a hyperinvasive subtype. Screening of the protease activity of wild-type populations of both strains identified high and low protease subtypes which also showed a corresponding reduction or enhancement, respectively, of invasive capabilities. Microarray analysis of these bistable populations revealed a putative signature set of genes that includes oxidative stress resistance and iron transport genes, and which might be critical to invasion of or survival within epithelial cells.
Collapse
Affiliation(s)
- S Suwannakul
- Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK
| | - G P Stafford
- Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK
| | - S A Whawell
- Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK
| | - C W I Douglas
- Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK
| |
Collapse
|
24
|
|
25
|
Tetratricopeptide repeat protein-associated proteins contribute to the virulence of Porphyromonas gingivalis. Infect Immun 2010; 78:2846-56. [PMID: 20351137 DOI: 10.1128/iai.01448-09] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gingivalis is one of the most etiologically important microorganisms in periodontal disease. We found in a previous study that PG1385 (TprA) protein, a tetratricopeptide repeat (TPR) protein, was upregulated in P. gingivalis wild-type cells placed in a mouse subcutaneous chamber and that a tprA mutant was clearly less virulent in the mouse subcutaneous abscess model (M. Yoshimura et al., Oral Microbiol. Immunol. 23:413-418, 2008). In the present study, we investigated the gene expression profile of tprA mutant cells placed in a mouse subcutaneous chamber and found that 9 genes, including PG2102 (tapA), PG2101 (tapB), and PG2100 (tapC) genes, were downregulated in the tprA mutant compared with those in the wild type. Expression of a cluster of tapA, tapB, and tapC genes of the mutant was also downregulated in an in vitro culture with enriched brain heart infusion medium. The TprA protein has three TPR motifs known as a protein-protein interaction module. Yeast two-hybrid system analysis and in vitro protein binding assays with immunoprecipitation and surface plasmon resonance detection revealed that the TprA protein could bind to TapA and TapB proteins. TprA and TapB proteins were located in the periplasmic space, whereas TapA, which appeared to be one of the C-terminal domain family proteins, was located at the outer membrane. We constructed tapA, tapB, and tapC single mutants and a tapA-tapB-tapC deletion mutant. In the mouse subcutaneous infection experiment, all of the mutants were less virulent than the wild type. These results suggest that TprA, TapA, TapB, and TapC are cooperatively involved in P. gingivalis virulence.
Collapse
|
26
|
Nguyen T, Zhang Z, Huang IH, Wu C, Merritt J, Shi W, Qi F. Genes involved in the repression of mutacin I production in Streptococcus mutans. MICROBIOLOGY-SGM 2009; 155:551-556. [PMID: 19202103 DOI: 10.1099/mic.0.021303-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Streptococcus mutans is considered a primary pathogen for human dental caries. Its ability to produce a variety of peptide antibiotics called mutacins may play an important role in its invasion and establishment in the dental biofilm. S. mutans strain UA140 produces two types of mutacins, the lantibiotic mutacin I and the non-lantibiotic mutacin IV. In a previous study, we constructed a random insertional-mutation library to screen for genes involved in regulating mutacin I production, and found 25 genes/operons that have a positive effect on mutacin I production. In this study, we continued our previous work to identify genes that are negatively involved in mutacin I production. By using a high-phosphate brain heart infusion agar medium that inhibited mutacin I production of the wild-type, we isolated 77 clones that consistently produced mutacin I under repressive conditions. From the 34 clones for which we were able to obtain a sequence, 17 unique genes were identified. These genes encompass a variety of functional groups, including central metabolism, surface binding and sugar transport, and unknown functions. Some of the 17 mutations were further characterized and shown to increase mutacin gene expression during growth when the gene is usually not expressed in the wild-type. These results further demonstrate an intimate and intricate connection between mutacin production and the overall cellular homeostasis.
Collapse
Affiliation(s)
- Trang Nguyen
- UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Zhijun Zhang
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73034, USA
| | - I-Hsiu Huang
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73034, USA
| | - Chenggang Wu
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73034, USA
| | - Justin Merritt
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73034, USA
| | - Wenyuan Shi
- UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Fengxia Qi
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73034, USA
| |
Collapse
|
27
|
Yoshimura M, Ohara N, Kondo Y, Shoji M, Okano S, Nakano Y, Abiko Y, Nakayama K. Proteome analysis of Porphyromonas gingivalis cells placed in a subcutaneous chamber of mice. ACTA ACUST UNITED AC 2008; 23:413-8. [PMID: 18793365 DOI: 10.1111/j.1399-302x.2008.00444.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
INTRODUCTION Porphyromonas gingivalis, an oral anaerobic bacterium, is considered a major pathogen for chronic periodontitis. Pathogenic bacteria usually upregulate or downregulate gene expression to combat the protective responses of their hosts. METHODS To determine what protein is regulated when P. gingivalis cells invade host tissues, we analyzed the proteome of P. gingivalis cells that were placed in a mouse subcutaneous chamber by two-dimensional gel electrophoresis and mass spectrometry. RESULTS Fourteen proteins were upregulated, while four proteins were downregulated. We focused on three upregulated proteins, PG1089 (DNA-binding response regulator RprY), PG1385 (TPR domain protein), and PG2102 (immunoreactive 61-kDa antigen), and constructed mutant strains that were defective in these proteins. Mouse abscess model experiments revealed that the mutant strain defective in PG1385 was clearly less virulent than the wild-type parent strain. CONCLUSION These results indicate that the PG1385 protein is involved in P. gingivalis virulence and that the method used here is useful when investigating the P. gingivalis proteins responsible for virulence.
Collapse
Affiliation(s)
- M Yoshimura
- Division of Microbiology and Oral Infection, Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Response of Porphyromonas gingivalis to heme limitation in continuous culture. J Bacteriol 2008; 191:1044-55. [PMID: 19028886 DOI: 10.1128/jb.01270-08] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Porphyromonas gingivalis is an anaerobic, asaccharolytic, gram-negative bacterium that has essential requirements for both iron and protoporphyrin IX, which it preferentially obtains as heme. A combination of large-scale quantitative proteomic analysis using stable isotope labeling strategies and mass spectrometry, together with transcriptomic analysis using custom-made DNA microarrays, was used to identify changes in P. gingivalis W50 protein and transcript abundances on changing from heme-excess to heme-limited continuous culture. This approach identified 160 genes and 70 proteins that were differentially regulated by heme availability, with broad agreement between the transcriptomic and proteomic data. A change in abundance of the enzymes of the aspartate and glutamate catabolic pathways was observed with heme limitation, which was reflected in organic acid end product levels of the culture fluid. These results demonstrate a shift from an energy-efficient anaerobic respiration to a less efficient process upon heme limitation. Heme limitation also resulted in an increase in abundance of a protein, PG1374, which we have demonstrated, by insertional inactivation, to have a role in epithelial cell invasion. The greater abundance of a number of transcripts/proteins linked to invasion of host cells, the oxidative stress response, iron/heme transport, and virulence of the bacterium indicates that there is a broad response of P. gingivalis to heme availability.
Collapse
|
29
|
Xia Q, Wang T, Taub F, Park Y, Capestany CA, Lamont RJ, Hackett M. Quantitative proteomics of intracellular Porphyromonas gingivalis. Proteomics 2008; 7:4323-37. [PMID: 17979175 DOI: 10.1002/pmic.200700543] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Whole-cell quantitative proteomic analyses were conducted to investigate the change from an extracellular to intracellular lifestyle for Porphyromonas gingivalis, a Gram-negative intracellular pathogen associated with periodontal disease. Global protein abundance data for P. gingivalis strain ATCC 33277 internalized for 18 h within human gingival epithelial cells and controls exposed to gingival cell culture medium were obtained at sufficient coverage to provide strong evidence that these changes are profound. A total of 385 proteins were overexpressed in internalized P. gingivalis relative to controls; 240 proteins were shown to be underexpressed. This represented in total about 28% of the protein encoding ORFs annotated for this organism, and slightly less than half of the proteins that were observed experimentally. Production of several proteases, including the classical virulence factors RgpA, RgpB, and Kgp, was decreased. A separate validation study was carried out in which a 16-fold dilution of the P. gingivalis proteome was compared to the undiluted sample in order to assess the quantitative false negative rate (all ratios truly alternative). Truly null (no change) abundance ratios from technical replicates were used to assess the rate of quantitative false positives over the entire proteome. A global comparison between the direction of abundance change observed and previously published bioinformatic gene pair predictions for P. gingivalis will assist with future studies of P. gingivalis gene regulation and operon prediction.
Collapse
Affiliation(s)
- Qiangwei Xia
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Role of the Clp system in stress tolerance, biofilm formation, and intracellular invasion in Porphyromonas gingivalis. J Bacteriol 2007; 190:1436-46. [PMID: 18065546 DOI: 10.1128/jb.01632-07] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Clp proteases and chaperones are ubiquitous among prokaryotes and eukaryotes, and in many pathogenic bacteria the Clp stress response system is also involved in regulation of virulence properties. In this study, the roles of ClpB, ClpC, and ClpXP in stress resistance, homotypic and heterotypic biofilm formation, and intracellular invasion in the oral opportunistic pathogen Porphyromonas gingivalis were investigated. Absence of ClpC and ClpXP, but not ClpB, resulted in diminished tolerance to high temperatures. Response to oxidative stress was not affected by the loss of any of the Clp proteins. The clpC and clpXP mutants demonstrated elevated monospecies biofilm formation, and the absence of ClpXP also enhanced heterotypic P. gingivalis-Streptococcus gordonii biofilm formation. All clp mutants adhered to gingival epithelial cells to the same level as the wild type; however, ClpC and ClpXP were found to be necessary for entry into host epithelial cells. ClpB did not play a role in entry but was required for intracellular replication and survival. ClpXP negatively regulated the surface exposure of the minor fimbrial (Mfa) protein subunit of P. gingivalis, which stimulates biofilm formation but interferes with epithelial cell entry. Collectively, these results show that the Clp protease complex and chaperones control several processes that are important for the colonization and survival of P. gingivalis in the oral cavity.
Collapse
|
31
|
Garbom S, Olofsson M, Björnfot AC, Srivastava MK, Robinson VL, Oyston PCF, Titball RW, Wolf-Watz H. Phenotypic characterization of a virulence-associated protein, VagH, of Yersinia pseudotuberculosis reveals a tight link between VagH and the type III secretion system. MICROBIOLOGY-SGM 2007; 153:1464-1473. [PMID: 17464060 DOI: 10.1099/mic.0.2006/000323-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recently, a number of attenuated mutants of Yersinia pseudotuberculosis have been identified using a bioinformatics approach. One of the target genes identified in that study was vagH, which the authors now characterized further. VagH shows homology to HemK of Escherichia coli, possessing methyltransferase activity similar to that of HemK, and targeting release factors 1 and 2. Microarray studies comparing the wild-type and the vagH mutant revealed that the mRNA levels of only a few genes were altered in the mutant. By proteome analysis, expression of the virulence determinant YopD was found to be increased, indicating a possible connection between VagH and the virulence plasmid-encoded type III secretion system (T3SS). Further analysis showed that Yop expression and secretion were repressed in a vagH mutant. This phenotype could be suppressed by trans-complementation with the wild-type vagH gene or by deletion of the negative regulator yopD. Also, in a similar manner to a T3SS-negative mutant, the avirulent vagH mutant was rapidly cleared from Peyer's patches and could not reach the spleen after oral infection of mice. In a manner analogous to that of T3SS mutants, the vagH mutant could not block phagocytosis by macrophages. However, a vagH mutant showed no defects in the T3SS-independent ability to proliferate intracellularly and replicated to levels similar to those of the wild-type in macrophages. In conclusion, the vagH mutant exhibits a virulence phenotype similar to that of a T3SS-negative mutant, indicating a tight link between VagH and type III secretion in Y. pseudotuberculosis.
Collapse
Affiliation(s)
- Sara Garbom
- Department of Molecular Biology, Umeå University, S-90187 Umeå, Sweden
| | - Martina Olofsson
- Department of Molecular Biology, Umeå University, S-90187 Umeå, Sweden
| | | | - Manoj Kumar Srivastava
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, S-90183 Umeå, Sweden
| | | | - Petra C F Oyston
- Biomedical Sciences, Dstl Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Richard W Titball
- Biomedical Sciences, Dstl Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Hans Wolf-Watz
- Department of Molecular Biology, Umeå University, S-90187 Umeå, Sweden
| |
Collapse
|
32
|
Xia Q, Wang T, Park Y, Lamont RJ, Hackett M. Differential quantitative proteomics of Porphyromonas gingivalis by linear ion trap mass spectrometry: non-label methods comparison, q-values and LOWESS curve fitting. INTERNATIONAL JOURNAL OF MASS SPECTROMETRY 2007; 259:105-116. [PMID: 19337574 PMCID: PMC2662607 DOI: 10.1016/j.ijms.2006.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Differential analysis of whole cell proteomes by mass spectrometry has largely been applied using various forms of stable isotope labeling. While metabolic stable isotope labeling has been the method of choice, it is often not possible to apply such an approach. Four different label free ways of calculating expression ratios in a classic "two-state" experiment are compared: signal intensity at the peptide level, signal intensity at the protein level, spectral counting at the peptide level, and spectral counting at the protein level. The quantitative data were mined from a dataset of 1245 qualitatively identified proteins, about 56% of the protein encoding open reading frames from Porphyromonas gingivalis, a Gram-negative intracellular pathogen being studied under extracellular and intracellular conditions. Two different control populations were compared against P. gingivalis internalized within a model human target cell line. The q-value statistic, a measure of false discovery rate previously applied to transcription microarrays, was applied to proteomics data. For spectral counting, the most logically consistent estimate of random error came from applying the locally weighted scatter plot smoothing procedure (LOWESS) to the most extreme ratios generated from a control technical replicate, thus setting upper and lower bounds for the region of experimentally observed random error.
Collapse
Affiliation(s)
- Qiangwei Xia
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Tiansong Wang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Yoonsuk Park
- Department of Oral Biology, University of Florida, Gainesville, FL 32610, USA
| | - Richard J. Lamont
- Department of Oral Biology, University of Florida, Gainesville, FL 32610, USA
| | - Murray Hackett
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
- Address correspondence to: Murray Hackett, Department of Chemical Engineering, Box 355014, University of Washington, Seattle, WA 98195, U.S.A., Telephone: 206 616-8071, Fax: 206 616-5721, E-mail:
| |
Collapse
|
33
|
He J, Miyazaki H, Anaya C, Yu F, Yeudall WA, Lewis JP. Role of Porphyromonas gingivalis FeoB2 in metal uptake and oxidative stress protection. Infect Immun 2006; 74:4214-23. [PMID: 16790796 PMCID: PMC1489711 DOI: 10.1128/iai.00014-06] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Porphyromonas gingivalis, a gram-negative anaerobic bacterium, is a recognized periodontopathogen. It exhibits a high degree of aerotolerance and is able to survive in host cells, indicating that efficient oxidative stress protection mechanisms must be present in this organism. Manganese homeostasis plays a major role in oxidative stress protection in a variety of organisms; however, the transport and role of this metal in P. gingivalis is not well understood. Analysis of the genome of P. gingivalis W83 revealed the presence of two genes encoding homologs of a ferrous iron transport protein, FeoB1 and FeoB2. FeoB2 has been implicated in manganese accumulation in P. gingivalis. We sought to determine the role of the FeoB2 protein in metal transport as well as its contribution to resistance to oxygen radicals. Quantitative reverse transcriptase PCR analyses demonstrated that expression of feoB2 is induced in the presence of oxygen. The role of FeoB2 was investigated using an isogenic mutant strain deficient in the putative transporter. We characterized the FeoB2-mediated metal transport using (55)Fe(2+) and (54)Mn(2+). The FeoB2-deficient mutant had dramatically reduced rates of manganese uptake (0.028 pmol/min/10(7) bacteria) compared with the parental strain (0.33 pmol/min/10(7) bacteria) (after 20 min of uptake using 50 nM of (54)Mn(2+)). The iron uptake rates, however, were higher in the mutant strain (0.75 pmol/min/10(7) bacteria) than in the wild type (0.39 pmol/min/10(7) bacteria). Interestingly, reduced survival rates were also noted for the mutant strain after exposure to H(2)O(2) and to atmospheric oxygen compared to the parental strain cultured under the same conditions. In addition, in vitro infection of host cells with the wild type, the FeoB2-deficient mutant, and the same-site revertant revealed that the mutant had a significantly decreased capability for intracellular survival in the host cells compared to the wild-type strain. Our results demonstrate that feoB2 encodes a major manganese transporter required for protection of the bacterium from oxidative stress generated by atmospheric oxygen and H(2)O(2). Furthermore, we show that FeoB2 and acquisition of manganese are required for intracellular survival of P. gingivalis in host cells.
Collapse
Affiliation(s)
- Jia He
- Philips Institute of Oral and Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, P.O. Box 980566, Richmond, VA 23298, USA
| | | | | | | | | | | |
Collapse
|
34
|
Andrian E, Grenier D, Rouabhia M. Porphyromonas gingivalis-epithelial cell interactions in periodontitis. J Dent Res 2006; 85:392-403. [PMID: 16632751 DOI: 10.1177/154405910608500502] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Emerging data on the consequences of the interactions between invasive oral bacteria and host cells have provided new insights into the pathogenesis of periodontal disease. Indeed, modulation of the mucosal epithelial barrier by pathogenic bacteria appears to be a critical step in the initiation and progression of periodontal disease. Periodontopathogens such as Porphyromonas gingivalis have developed different strategies to perturb the structural and functional integrity of the gingival epithelium. P. gingivalis adheres to, invades, and replicates within human epithelial cells. Adhesion of P. gingivalis to host cells is multimodal and involves the interaction of bacterial cell-surface adhesins with receptors expressed on the surfaces of epithelial cells. Internalization of P. gingivalis within host cells is rapid and requires both bacterial contact-dependent components and host-induced signaling pathways. P. gingivalis also subverts host responses to bacterial challenges by inactivating immune cells and molecules and by activating host processes leading to tissue destruction. The adaptive ability of these pathogens that allows them to survive within host cells and degrade periodontal tissue constituents may contribute to the initiation and progression of periodontitis. In this paper, we review current knowledge on the molecular cross-talk between P. gingivalis and gingival epithelial cells in the development of periodontitis.
Collapse
Affiliation(s)
- E Andrian
- Groupe de Recherche en Ecologie Buccale, Faculté de médecine dentaire, Université Laval, Quebec City, Canada
| | | | | |
Collapse
|
35
|
Capestany CA, Kuboniwa M, Jung IY, Park Y, Tribble GD, Lamont RJ. Role of the Porphyromonas gingivalis InlJ protein in homotypic and heterotypic biofilm development. Infect Immun 2006; 74:3002-5. [PMID: 16622239 PMCID: PMC1459709 DOI: 10.1128/iai.74.5.3002-3005.2006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The oral pathogen Porphyromonas gingivalis expresses a homolog of the internalin family protein InlJ. Inactivation of inlJ reduced monospecies biofilm formation by P. gingivalis. In contrast, heterotypic P. gingivalis-Streptococcus gordonii biofilm formation was enhanced in the InlJ-deficient mutant. The results indicate a nuanced role for InlJ in regulating biofilm accumulations of P. gingivalis.
Collapse
Affiliation(s)
- Cindy A Capestany
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA
| | | | | | | | | | | |
Collapse
|
36
|
Urnowey S, Ansai T, Bitko V, Nakayama K, Takehara T, Barik S. Temporal activation of anti- and pro-apoptotic factors in human gingival fibroblasts infected with the periodontal pathogen, Porphyromonas gingivalis: potential role of bacterial proteases in host signalling. BMC Microbiol 2006; 6:26. [PMID: 16524480 PMCID: PMC1431544 DOI: 10.1186/1471-2180-6-26] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Accepted: 03/08/2006] [Indexed: 01/07/2023] Open
Abstract
Background Porphyromonas gingivalis is the foremost oral pathogen of adult periodontitis in humans. However, the mechanisms of bacterial invasion and the resultant destruction of the gingival tissue remain largely undefined. Results We report host-P. gingivalis interactions in primary human gingival fibroblast (HGF) cells. Quantitative immunostaining revealed the need for a high multiplicity of infection for optimal infection. Early in infection (2–12 h), P. gingivalis activated the proinflammatory transcription factor NF-kappa B, partly via the PI3 kinase/AKT pathway. This was accompanied by the induction of cellular anti-apoptotic genes, including Bfl-1, Boo, Bcl-XL, Bcl2, Mcl-1, Bcl-w and Survivin. Late in infection (24–36 h) the anti-apoptotic genes largely shut down and the pro-apoptotic genes, including Nip3, Hrk, Bak, Bik, Bok, Bax, Bad, Bim and Moap-1, were activated. Apoptosis was characterized by nuclear DNA degradation and activation of caspases-3, -6, -7 and -9 via the intrinsic mitochondrial pathway. Use of inhibitors revealed an anti-apoptotic function of NF-kappa B and PI3 kinase in P. gingivalis-infected HGF cells. Use of a triple protease mutant P. gingivalis lacking three major gingipains (rgpA rgpB kgp) suggested a role of some or all these proteases in myriad aspects of bacteria-gingival interaction. Conclusion The pathology of the gingival fibroblast in P. gingivalis infection is affected by a temporal shift from cellular survival response to apoptosis, regulated by a number of anti- and pro-apoptotic molecules. The gingipain group of proteases affects bacteria-host interactions and may directly promote apoptosis by intracellular proteolytic activation of caspase-3.
Collapse
Affiliation(s)
- Sonya Urnowey
- Department of Biochemistry and Molecular Biology, University of South Alabama, College of Medicine, 307 University Blvd., Mobile, Alabama 36688-0002, USA
| | - Toshihiro Ansai
- Department of Preventive Dentistry, Kyushu Dental College, Kitakyushu 803-8580, Japan
| | - Vira Bitko
- Department of Biochemistry and Molecular Biology, University of South Alabama, College of Medicine, 307 University Blvd., Mobile, Alabama 36688-0002, USA
| | - Koji Nakayama
- Division of Microbiology and Oral Infection, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki 852-8588, Japan
| | - Tadamichi Takehara
- Department of Preventive Dentistry, Kyushu Dental College, Kitakyushu 803-8580, Japan
| | - Sailen Barik
- Department of Biochemistry and Molecular Biology, University of South Alabama, College of Medicine, 307 University Blvd., Mobile, Alabama 36688-0002, USA
| |
Collapse
|
37
|
Affiliation(s)
- Zhimin Feng
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | | |
Collapse
|
38
|
Zhou Q, Amar S. Identification of proteins differentially expressed in human monocytes exposed to Porphyromonas gingivalis and its purified components by high-throughput immunoblotting. Infect Immun 2006; 74:1204-14. [PMID: 16428770 PMCID: PMC1360359 DOI: 10.1128/iai.74.2.1204-1214.2006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To characterize the roles of Porphyromonas gingivalis and its components in disease processes, we investigated the cytokine profiles induced by live P. gingivalis, its lipopolysaccharide (LPS), and its major fimbrial protein, fimbrillin (FimA). A cytokine antibody array revealed that human monocyte-derived macrophages were induced to produce chemokines (e.g., monocyte chemoattractant protein 1, macrophage inflammatory protein 1beta [MIP-1beta], and MIP-3alpha) as early as 1 h after exposure to P. gingivalis, with production declining after 24 h of exposure. As expected, an extensive repertoire of inflammatory mediators increased subsequent to infection, most predominantly tumor necrosis factor alpha (TNF-alpha), interleukin 1beta (IL-1beta), IL-6, IL-10, and granulocyte-macrophage colony-stimulating factor. The induction of cytokines by P. gingivalis was not triggered simply by bacterial cell surface components, since purified P. gingivalis LPS and FimA induced similar patterns of cytokines, while the pattern of cytokines induced by live P. gingivalis was significantly different, indicating that the host defense system senses live bacteria differently than it does the cell surface components LPS and FimA. To further understand the mechanisms by which live P. gingivalis and its components exert their effects, we used a high-throughput immunoblot screening approach (Becton-Dickinson PowerBlot) to analyze intracellular proteins involved in P. gingivalis infection in human macrophages. Exposure of human macrophages to either live P. gingivalis, its LPS, or its FimA protein led to the up-regulation of 12, 8, and 10 proteins and the down-regulation of 15, 8, and 17 proteins, respectively. The expression of proteins involved in gene transcription (e.g., monocyte enhancer factor 2D [MEF2D], signal transducer and activator of transcription 1 [STAT1], STAT3, STAT6, and IL enhancer binding factors [ILF3]), of protein kinases (e.g., mitogen-activated protein kinase 3 [MAPK3], MAP3K8, double-stranded RNA-activated protein kinase [PRKR], and MAP2K4), and of proteins involved in immune responses (e.g., TNF super family member 6 [TNFSF6] and interferon-induced protein with tetratricopeptide repeat 4 [IFIT4]), apoptosis (e.g., genes associated with retinoid interferon-induced mortality 19 [GRIM19]), and other fundamental cellular processes (e.g., clathrin heavy-chain polypeptide, culreticulin, and Ras-associated protein RAB27A) was found to be modulated differentially by P. gingivalis, LPS, and FimA. These differential changes are interpreted as preferential signal pathway activation in host immune/inflammatory responses to P. gingivalis infection.
Collapse
Affiliation(s)
- Qingde Zhou
- Department of Periodontology and Oral Biology, School of Dental Medicine, Boston University Medical Center, 700 Albany Street W-201E, Boston, MA 02118, USA
| | | |
Collapse
|
39
|
Yilmaz O, Verbeke P, Lamont RJ, Ojcius DM. Intercellular spreading of Porphyromonas gingivalis infection in primary gingival epithelial cells. Infect Immun 2006; 74:703-10. [PMID: 16369027 PMCID: PMC1346639 DOI: 10.1128/iai.74.1.703-710.2006] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Porphyromonas gingivalis, an important periodontal pathogen, is an effective colonizer of oral tissues. The organism successfully invades, multiplies in, and survives for extended periods in primary gingival epithelial cells (GECs). It is unknown whether P. gingivalis resides in the cytoplasm of infected cells throughout the infection or can spread to adjacent cells over time. We developed a technique based on flow cytofluorometry and fluorescence microscopy to study propagation of the organism at different stages of infection of GECs. Results showed that P. gingivalis spreads cell to cell and that the amount of spreading increases gradually over time. There was a very low level of propagation of bacteria to uninfected cells early in the infection (3 h postinfection), but there were 20-fold and 45-fold increases in the propagation rate after 24 h and 48 h, respectively, of infection. Immunofluorescence microscopy of infected cells suggested that intercellular translocation of P. gingivalis may be mediated through actin-based membrane protrusions, bypassing the need for release of bacteria into extracellular medium. Consistent with these observations, cytochalasin D treatment of infected cells resulted in significant inhibition of bacterial spreading. This study shows for the first time that P. gingivalis disseminates from cell to cell without passing through the extracellular space. This mechanism of spreading may allow P. gingivalis to colonize oral tissues without exposure to the humoral immune response.
Collapse
Affiliation(s)
- Ozlem Yilmaz
- Department of Pathobiology, University of Washington, Box 357 238, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
40
|
Kucknoor AS, Mundodi V, Alderete JF. Adherence to human vaginal epithelial cells signals for increased expression of Trichomonas vaginalis genes. Infect Immun 2005; 73:6472-8. [PMID: 16177319 PMCID: PMC1230950 DOI: 10.1128/iai.73.10.6472-6478.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Host parasitism by Trichomonas vaginalis is complex, and the adhesion to vaginal epithelial cells (VECs) by trichomonads is preparatory to colonization of the vagina. Since we showed increased synthesis of adhesins after contact with VECs (A. F. Garcia, et al., Mol. Microbiol. 47:1207-1224, 2003) and more recently demonstrated up-regulated gene expression in VECs after parasite attachment (A. S. Kucknoor, et al., Cell. Microbiol. 7:887-897, 2005), we hypothesized that enhanced expression of adhesin and other genes would result from signaling of trichomonads following adherence. In order to identify the genes that are up-regulated, we constructed a subtraction cDNA library enriched for differentially expressed genes from the parasites that were in contact with the host cells. Thirty randomly selected cDNA clones representing the differentially regulated genes upon initial contact of parasites with host cells were sequenced. Several genes encoded functional proteins with specific functions known to be associated with colonization, such as adherence, change in morphology, and gene transcription and translation. Interestingly, genes unique to trichomonads with unknown functions were also up-regulated. Semiquantitative reverse transcription-PCR (RT-PCR) confirmed expression of select genes. An increased amount of protein was demonstrated by immunoblotting with monoclonal antibody. Finally, we showed the transcriptional regulation of some genes by iron by using RT-PCR. To our knowledge, this is the first report addressing the differential regulation of T. vaginalis genes immediately upon contact with VECs.
Collapse
Affiliation(s)
- Ashwini S Kucknoor
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, 78229, USA
| | | | | |
Collapse
|
41
|
Hosogi Y, Duncan MJ. Gene expression in Porphyromonas gingivalis after contact with human epithelial cells. Infect Immun 2005; 73:2327-35. [PMID: 15784578 PMCID: PMC1087432 DOI: 10.1128/iai.73.4.2327-2335.2005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Porphyromonas gingivalis, a gram-negative oral anaerobe, is strongly associated with adult periodontitis. The adherence of the organism to host epithelium signals changes in both cell types as bacteria initiate infection and colonization and epithelial cells rally their defenses. We hypothesized that the expression of a defined set of P. gingivalis genes would be consistently up-regulated during infection of HEp-2 human epithelial cells. P. gingivalis genome microarrays were used to compare the gene expression profiles of bacteria that adhered to HEp-2 cells and bacteria that were incubated alone. Genes whose expression was temporally up-regulated included those involved in the oxidative stress response and those encoding heat shock proteins that are essential to maintaining cell viability under adverse conditions. The results suggest that contact with epithelial cells induces in P. gingivalis stress-responsive pathways that promote the survival of the bacterium.
Collapse
Affiliation(s)
- Yumiko Hosogi
- Department of Molecular Genetics, The Forsyth Institute, 140 Fenway, Boston, MA 02115, USA
| | | |
Collapse
|