1
|
Kiarashi M, Yasamineh S. The role of cellular lipid metabolism and lipid-lowering drugs in periodontitis. Int Immunopharmacol 2025; 152:114434. [PMID: 40086058 DOI: 10.1016/j.intimp.2025.114434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Initiated by bacteria, periodontitis (PD) is a complex, chronic inflammatory disease of the supporting tissue of the gums and teeth. Also linked to PD include human papillomavirus (HPV), hepatitis B virus (HBV), Epstein-Barr virus (EBV), human cytomegalovirus (CMV), and Herpes Simplex Virus (HSV). PD also raises the risk of cardiovascular disease (CVD) because it triggers inflammatory reactions throughout the body. CVD and chronic PD were linked to significantly elevated levels of C-reactive protein and blood lipids. Furthermore, elevated lipid peroxidation (LPO) levels may influence PD-related inflammation and periodontium degradation. In addition, there was a correlation between a reduction in oxidized low-density lipoprotein (LDL) levels and a reduction in circulating oxidative stress (OS); this was shown to be achieved by improved dental hygiene and non-surgical periodontal treatment. Consequently, this research set out to examine the connections between lipid metabolism and PD, as well as the effects of PD on the efficacy of statins and other medications that decrease cholesterol, as well as inhibitors and other lipid-lowering agents. Additionally, it's worth mentioning that statins and other cholesterol-lowering drugs may affect gum and tooth health. We found that higher blood levels of bad cholesterol exacerbate PD. Furthermore, PD makes CVD worse. The involvement of proprotein convertase subtilisin/kexin type 9 (PCSK9) in bacterial infections and the development of PD is inversely proportional to the increase in LDL levels. The treatment of this disease could, therefore, benefit greatly by inhibiting this chemical. Medications that lower cholesterol levels may potentially help treat this problem. The possible side effects of this medication on PD patients need more investigation. We have reviewed the literature on PD and its relationship to lipid metabolism, LDL receptors, and lipid rafts. Afterward, we investigated the role of lipid metabolism in the local viral infection that causes PD. Lastly, we examined how statins and other lipid-lowering medications impact PD.
Collapse
Affiliation(s)
- Mohammad Kiarashi
- College of Dentistry, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| |
Collapse
|
2
|
DiGianivittorio P, Hinkel LA, Mackinder JR, Schutz K, Klein EA, Wargo MJ. The Pseudomonas aeruginosa sphBC genes are important for growth in the presence of sphingosine by promoting sphingosine metabolism. MICROBIOLOGY (READING, ENGLAND) 2025; 171:001520. [PMID: 39791474 PMCID: PMC11893366 DOI: 10.1099/mic.0.001520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025]
Abstract
Sphingoid bases, including sphingosine, are important components of the antimicrobial barrier at epithelial surfaces where they can cause growth inhibition and killing of susceptible bacteria. Pseudomonas aeruginosa is a common opportunistic pathogen that is less susceptible to sphingosine than many Gram-negative bacteria. Here, we determined that the deletion of the sphBCD operon reduced growth in the presence of sphingosine. Using deletion mutants, complementation and growth assays in P. aeruginosa PAO1, we determined that the sphC and sphB genes, encoding a periplasmic oxidase and periplasmic cytochrome c, respectively, were important for growth on sphingosine, while sphD was dispensable under these conditions. Deletion of sphBCD in P. aeruginosa PA14, Pseudomonas protegens Pf-5 and Pseudomonas fluorescens Pf01 also showed reduced growth in the presence of sphingosine. The P. aeruginosa sphBC genes were also important for growth in the presence of two other sphingoid bases, phytosphingosine and sphinganine. In WT P. aeruginosa, sphingosine is metabolized to an unknown non-inhibitory product, as sphingosine concentrations drop in the culture. However, in the absence of sphBC, sphingosine accumulates, pointing to SphC and SphB as having a role in sphingosine metabolism. Finally, the metabolism of sphingosine by WT P. aeruginosa protected susceptible cells from full growth inhibition by sphingosine, pointing to a role for sphingosine metabolism as a public good. This work shows that the metabolism of sphingosine by P. aeruginosa presents a novel pathway by which bacteria can alter host-derived sphingolipids, but it remains an open question whether SphB and SphC act directly on sphingosine.
Collapse
Affiliation(s)
- Pauline DiGianivittorio
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, USA
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, USA
| | - Lauren A. Hinkel
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, USA
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, USA
- Biology Department, Rutgers University-Camden, Camden, USA
| | - Jacob R. Mackinder
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, USA
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, USA
| | - Kristin Schutz
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, USA
| | - Eric A. Klein
- Biology Department, Rutgers University-Camden, Camden, USA
| | - Matthew J Wargo
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, USA
| |
Collapse
|
3
|
Pious A, Venkatasubramanian V, Singaravelu DK, Ramesh S, Ameen F, Veerappan A. Synthesis of cationic N-acylated thiazolidine for selective activity against Gram-positive bacteria and evaluation of N-acylation's role in membrane-disrupting activity. RSC Med Chem 2024:d4md00626g. [PMID: 39507614 PMCID: PMC11537284 DOI: 10.1039/d4md00626g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
The evolution of antimicrobial-resistant strains jeopardizes the existing clinical drugs and demands new therapeutic interventions. Herein, we report the synthesis of cationic thiazolidine bearing a quaternary pyridinium group, in which thiazolidine was N-acylated with fatty acid to establish a hydrophilic-lipophilic balance that disrupts bacterial membranes. The bacterial growth inhibition assays and hemolytic activity against human red blood cells indicate that the N-acylated cationic thiazolidine (QPyNATh) inhibits Gram-positive bacteria at lower minimum inhibitory concentrations (MIC) and is selective for bacteria over mammalian cells. N-Acylation modulates MIC, and it is found that the N-palmitoylated compound, QPyN16Th, had the lowest MIC (1.95 μM) against Gram-positive, Enterococcus faecalis, Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA). In contrast, the N-myristoylated compound, QPyN14Th, showed the lowest MIC (31.25 μM) against Gram-negative, Escherichia coli, uropathogenic Escherichia coli, and Pseudomonas aeruginosa. At 1× MIC, QPyNATh permeabilizes the bacterial membrane, depolarizes the cytoplasmic membranes, and produces excess reactive oxygen species to kill the bacteria, as evidenced by live and dead staining. Interestingly, only QPyNATh containing a palmitoyl acyl chain demonstrated membrane-damaging activity at 2 μM concentrations, suggesting that the optimal hydrophilic-lipophilic balance enables QPyN16Th to selectively kill Gram-positive bacteria at lower doses. S. aureus develops resistance to ciprofloxacin quickly; however, no resistance to QPyN16Th is observed after several passages. As a proof of concept, the animal study revealed that QPyN16Th treatment reduced the bacterial burden in MRSA-infected zebrafish, allowing them to recover from infection and resume normal life. The results imply that lipidation and derivatizing thiazolidine with cationic charge offer an antimicrobial that is selective to treat Gram-positive bacterial infections, biocompatible, and less prone to develop resistance.
Collapse
Affiliation(s)
- Aleena Pious
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University Thanjavur - 613401 Tamil Nadu India
| | - Vignesh Venkatasubramanian
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University Thanjavur - 613401 Tamil Nadu India
| | - Dharshini Karnan Singaravelu
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University Thanjavur - 613401 Tamil Nadu India
| | - Subburethinam Ramesh
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University Thanjavur - 613401 Tamil Nadu India
| | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University Riyadh 11451 Saudi Arabia
| | - Anbazhagan Veerappan
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University Thanjavur - 613401 Tamil Nadu India
| |
Collapse
|
4
|
Xie RH, Liu H, Qi C, He Y. From dysbiosis to homeostasis: Oleic acid matters in the vagina. Cell Host Microbe 2024; 32:1641-1643. [PMID: 39389025 DOI: 10.1016/j.chom.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024]
Abstract
The role of fatty acids in shaping vaginal microbiota remains unclear. In an issue of Cell, Zhu et al. use genomic and transcriptomic analyses to reveal that oleic acid (OA) selectively inhibits L. iners while promoting L. crispatus, suggesting new strategies for the treatment of bacterial vaginosis (BV).
Collapse
Affiliation(s)
- Ri-Hua Xie
- Women and Children Medical Research Center, Foshan Women and Children Hospital, Foshan, Guangdong, China; School of Nursing, Southern Medical University, Guangzhou, Guangdong, China.
| | - Hao Liu
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, China
| | - Cancan Qi
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Zhu M, Frank MW, Radka CD, Jeanfavre S, Xu J, Tse MW, Pacheco JA, Kim JS, Pierce K, Deik A, Hussain FA, Elsherbini J, Hussain S, Xulu N, Khan N, Pillay V, Mitchell CM, Dong KL, Ndung'u T, Clish CB, Rock CO, Blainey PC, Bloom SM, Kwon DS. Vaginal Lactobacillus fatty acid response mechanisms reveal a metabolite-targeted strategy for bacterial vaginosis treatment. Cell 2024; 187:5413-5430.e29. [PMID: 39163861 PMCID: PMC11429459 DOI: 10.1016/j.cell.2024.07.029] [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: 12/20/2023] [Revised: 05/15/2024] [Accepted: 07/18/2024] [Indexed: 08/22/2024]
Abstract
Bacterial vaginosis (BV), a common syndrome characterized by Lactobacillus-deficient vaginal microbiota, is associated with adverse health outcomes. BV often recurs after standard antibiotic therapy in part because antibiotics promote microbiota dominance by Lactobacillus iners instead of Lactobacillus crispatus, which has more beneficial health associations. Strategies to promote L. crispatus and inhibit L. iners are thus needed. We show that oleic acid (OA) and similar long-chain fatty acids simultaneously inhibit L. iners and enhance L. crispatus growth. These phenotypes require OA-inducible genes conserved in L. crispatus and related lactobacilli, including an oleate hydratase (ohyA) and putative fatty acid efflux pump (farE). FarE mediates OA resistance, while OhyA is robustly active in the vaginal microbiota and enhances bacterial fitness by biochemically sequestering OA in a derivative form only ohyA-harboring organisms can exploit. OA promotes L. crispatus dominance more effectively than antibiotics in an in vitro BV model, suggesting a metabolite-based treatment approach.
Collapse
Affiliation(s)
- Meilin Zhu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Matthew W Frank
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christopher D Radka
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | | | - Jiawu Xu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Megan W Tse
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jae Sun Kim
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Kerry Pierce
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amy Deik
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Fatima Aysha Hussain
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Salina Hussain
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Nondumiso Xulu
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Nasreen Khan
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | | | - Caroline M Mitchell
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Krista L Dong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA; Health Systems Trust, Durban, South Africa; Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Thumbi Ndung'u
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa; Africa Health Research Institute, Durban, South Africa; Max Planck Institute for Infection Biology, Berlin, Germany; Division of Infection and Immunity, University College London, London, UK
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles O Rock
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul C Blainey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Seth M Bloom
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA.
| | - Douglas S Kwon
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
DiGianivittorio P, Hinkel LA, Mackinder JR, Schutz K, Klein EA, Wargo MJ. The Pseudomonas aeruginosa sphBC genes are important for growth in the presence of sphingosine by promoting sphingosine metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611043. [PMID: 39282278 PMCID: PMC11398299 DOI: 10.1101/2024.09.03.611043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Sphingoid bases, including sphingosine, are important components of the antimicrobial barrier at epithelial surfaces where they can cause growth inhibition and killing of susceptible bacteria. Pseudomonas aeruginosa is a common opportunistic pathogen that is less susceptible to sphingosine than many Gram-negative bacteria. Here, we determined that deletion of the sphBCD operon reduced growth in the presence of sphingosine. Using deletion mutants, complementation, and growth assays in P. aeruginosa PAO1, we determined that the sphC and sphB genes, encoding a periplasmic oxidase and periplasmic cytochrome c, respectively, were important for growth on sphingosine, while sphD was dispensable under these conditions. Deletion of sphBCD in P. aeruginosa PA14, P. protegens Pf-5, and P. fluorescens Pf01 also showed reduced growth in the presence of sphingosine. The P. aeruginosa sphBC genes were also important for growth in the presence of two other sphingoid bases, phytosphingosine and sphinganine. In wild-type P. aeruginosa, sphingosine is metabolized to an unknown non-inhibitory product, as sphingosine concentrations drop in the culture. However, in the absence of sphBC, sphingosine accumulates, pointing to SphC and SphB as having a role in sphingosine metabolism. Finally, metabolism of sphingosine by wild-type P. aeruginosa protected susceptible cells from full growth inhibition by sphingosine, pointing to a role for sphingosine metabolism as a public good. This work shows that metabolism of sphingosine by P. aeruginosa presents a novel pathway by which bacteria can alter host-derived sphingolipids, but it remains an open question whether SphB and SphC act directly on sphingosine.
Collapse
Affiliation(s)
- Pauline DiGianivittorio
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont
| | - Lauren A. Hinkel
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont
- Biology Department, Rutgers University-Camden
| | - Jacob R. Mackinder
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont
| | - Kristin Schutz
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont
| | | | - Matthew J. Wargo
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont
| |
Collapse
|
7
|
Fu C, Brand HS, Nazmi K, Werner A, van Splunter A, Bikker FJ. Carbon dots combined with phytosphingosine inhibit acid-induced demineralization of hydroxyapatite in vitro. Arch Oral Biol 2024; 160:105911. [PMID: 38335699 DOI: 10.1016/j.archoralbio.2024.105911] [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: 11/08/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVES To study the effects of carbon dots (CDs), in combination with phytosphingosine (PHS), against acid-induced demineralization of hydroxyapatite in vitro. METHODS CDs were generated from citric acid and urea by microwave heating. Transmission electron microscope (TEM), FT-IR, and fluorescence intensity were used to characterize the CDs. A hydroxyapatite (HAp) model was used to investigate the protective effects of CDs, PHS, and their combinations with and without a salivary pellicle against acid-induced demineralization in vitro. Ca2+ release as a parameter to evaluate the inhibition of demineralization was measured by capillary electrophoresis. The interactions between CDs, PHS, and HAp discs were investigated using a fluorescence detector. RESULTS Uniform-sized CDs were synthesized, showing typical optical characteristics. CDs exhibited no inhibition of acid-induced demineralization in vitro, in contrast to PHS. Notably, a pre-coating of CDs increased the protective effects of PHS against acid-induced demineralization, which was not disturbed by the presence of a salivary pellicle and Tween 20. Scanning electron microscope (SEM) confirmed the binding and layers formed of both CDs and PHS to the HAp surfaces. Based on fluorescence spectra CDs binding to HAp seemed to be dependent on Ca2+ and PO43- interactions. CONCLUSIONS CDs combined with PHS showed protective effects against acid-induced demineralization of HAp discs in vitro.
Collapse
Affiliation(s)
- Cuicui Fu
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam 1081LA, the Netherlands.
| | - Henk S Brand
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam 1081LA, the Netherlands
| | - Kamran Nazmi
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam 1081LA, the Netherlands
| | - Arie Werner
- Department of Dental Materials Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam 1081LA, the Netherlands
| | - Annina van Splunter
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam 1081LA, the Netherlands
| | - Floris J Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam 1081LA, the Netherlands
| |
Collapse
|
8
|
Zhu M, Frank MW, Radka CD, Jeanfavre S, Tse MW, Pacheco JA, Pierce K, Deik A, Xu J, Hussain S, Hussain FA, Xulu N, Khan N, Pillay V, Dong KL, Ndung’u T, Clish CB, Rock CO, Blainey PC, Bloom SM, Kwon DS. Vaginal Lactobacillus fatty acid response mechanisms reveal a novel strategy for bacterial vaginosis treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.30.573720. [PMID: 38234804 PMCID: PMC10793477 DOI: 10.1101/2023.12.30.573720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Bacterial vaginosis (BV), a common syndrome characterized by Lactobacillus-deficient vaginal microbiota, is associated with adverse health outcomes. BV often recurs after standard antibiotic therapy in part because antibiotics promote microbiota dominance by Lactobacillus iners instead of Lactobacillus crispatus, which has more beneficial health associations. Strategies to promote L. crispatus and inhibit L. iners are thus needed. We show that oleic acid (OA) and similar long-chain fatty acids simultaneously inhibit L. iners and enhance L. crispatus growth. These phenotypes require OA-inducible genes conserved in L. crispatus and related species, including an oleate hydratase (ohyA) and putative fatty acid efflux pump (farE). FarE mediates OA resistance, while OhyA is robustly active in the human vaginal microbiota and sequesters OA in a derivative form that only ohyA-harboring organisms can exploit. Finally, OA promotes L. crispatus dominance more effectively than antibiotics in an in vitro model of BV, suggesting a novel approach for treatment.
Collapse
Affiliation(s)
- Meilin Zhu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Matthew W. Frank
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Christopher D. Radka
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky
| | | | - Megan W. Tse
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Kerry Pierce
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amy Deik
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jiawu Xu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Salina Hussain
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Fatima Aysha Hussain
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nondumiso Xulu
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Nasreen Khan
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | | | - Krista L. Dong
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Health Systems Trust, Durban, South Africa
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Thumbi Ndung’u
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute (AHRI), Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
- Division of Infection and Immunity, University College London, London, UK
| | | | - Charles O. Rock
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- passed away on September 22, 2023
| | - Paul C. Blainey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Seth M. Bloom
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Douglas S. Kwon
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Mosaddad SA, Hussain A, Tebyaniyan H. Green Alternatives as Antimicrobial Agents in Mitigating Periodontal Diseases: A Narrative Review. Microorganisms 2023; 11:1269. [PMCID: PMC10220622 DOI: 10.3390/microorganisms11051269] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/26/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Periodontal diseases and dental caries are the most common infectious oral diseases impacting oral health globally. Oral cavity health is crucial for enhancing life quality since it serves as the entranceway to general health. The oral microbiome and oral infectious diseases are strongly correlated. Gram-negative anaerobic bacteria have been associated with periodontal diseases. Due to the shortcomings of several antimicrobial medications frequently applied in dentistry, the lack of resources in developing countries, the prevalence of oral inflammatory conditions, and the rise in bacterial antibiotic resistance, there is a need for reliable, efficient, and affordable alternative solutions for the prevention and treatment of periodontal diseases. Several accessible chemical agents can alter the oral microbiota, although these substances also have unfavorable symptoms such as vomiting, diarrhea, and tooth discoloration. Natural phytochemicals generated from plants that have historically been used as medicines are categorized as prospective alternatives due to the ongoing quest for substitute products. This review concentrated on phytochemicals or herbal extracts that impact periodontal diseases by decreasing the formation of dental biofilms and plaques, preventing the proliferation of oral pathogens, and inhibiting bacterial adhesion to surfaces. Investigations examining the effectiveness and safety of plant-based medicines have also been presented, including those conducted over the past decade.
Collapse
Affiliation(s)
- Seyed Ali Mosaddad
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran;
| | - Ahmed Hussain
- School of Dentistry, Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Hamid Tebyaniyan
- Science and Research Branch, Islimic Azade University, Tehran 14878-92855, Iran
| |
Collapse
|
10
|
de Arruda CNF, Vivanco RG, Amorim AA, Ferreira AC, Tonani-Torrieri R, Bikker FJ, Pires-de-Souza FDCP. The effect of phytosphingosine associated with tooth brushing on color change, surface roughness, and microhardness of dental enamel - an in vitro and in situ study. Clin Oral Investig 2023; 27:849-858. [PMID: 35831626 DOI: 10.1007/s00784-022-04619-2] [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: 04/18/2022] [Accepted: 07/07/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study evaluated the in vitro and in situ effects of phytosphingosine (PHS) associated with tooth brushing on color stability, surface roughness, and microhardness of dental enamel. METHODS Sixty-four specimens of bovine teeth (6 × 6 × 2 mm) were separated into 8 groups (n = 8): S + TB: PHS (spray) + tooth brushing; TB + S: tooth brushing + PHS (spray); I + TB: PHS (immersion) + tooth brushing; TB + I: tooth brushing + PHS (immersion); TB: tooth brushing; S: PHS spray; I: immersion in PHS solution, and Saliva: immersion in saliva. Tooth brushing simulation (Mavtec, Brazil) was performed (356 rpm on 3.8 cm area by the toothbrush - Soft Tek) for 1, 7, 15, and 30 days. PHS remained in contact with specimens for 15 min. The specimens were evaluated before and after tooth brushing for color alteration (Easy Shade, VITA), and surface roughness (Model SJ-201P Mitutoyo), and Knoop microhardness (HMV-2, Shimadzu Corporation). For the in situ analyses, 8 participants were recruited and received an intraoral device with 6 fragments of bovine enamel (6 × 6 × 2 mm). The properties evaluated were the same as those of the in vitro study. Participants were randomized following best results of in vitro tested protocols, for 15 days: TB, TB + S, I + TB. Data obtained by in vitro (two-way ANOVA, Tukey, p < .05) and in situ (one-way ANOVA, Tukey, p < .05) studies were analyzed. RESULTS The in vitro study showed that greater color change was found after 30 days. The greatest differences in surface roughness occurred between the initial value and after 1 day. Regarding microhardness, the highest values occurred after 15 and 30 days, which showed similar results. The in situ study showed greater color changes for the TB and I + TB, and greater surface roughness changes for TB as well as a similar increase in microhardness for the PHS protocols, which were higher than TB. CONCLUSIONS Phytosphingosine leads to an increase in performance regarding color stability, surface roughness, and microhardness when applied. In general, the application of PHS after brushing showed a positive impact on its performance. CLINICAL RELEVANCE Phytosphingosine proved to be interesting for compound prevention formulations in the dentistry field.
Collapse
Affiliation(s)
- Carolina Noronha Ferraz de Arruda
- Department of Dental Prosthodontics, Dental School of Rio de Janeiro State University, Boulevard 28 de Setembro, 157, Rio de Janeiro, RJ, 20551-030, Brazil.
| | - Rocio Geng Vivanco
- Department of Dental Materials and Prosthodontics, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ayodele Alves Amorim
- Department of Dental Materials and Prosthodontics, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Adriana Cavalcanti Ferreira
- Department of Dental Materials and Prosthodontics, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaella Tonani-Torrieri
- Department of Dental Materials and Prosthodontics, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Floris Jacob Bikker
- Department of Periodontology and Oral Biochemistry, Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit and Universiteit Van Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
11
|
Li X, Ma M, Zhao B, Li N, Fang L, Wang D, Luan T. Chlorinated Polycyclic Aromatic Hydrocarbons Induce Immunosuppression in THP-1 Macrophages Characterized by Disrupted Amino Acid Metabolism. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:16012-16023. [PMID: 36282008 DOI: 10.1021/acs.est.2c06471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Frequent chlorinated polycyclic aromatic hydrocarbon (Cl-PAH) occurrence in environmental samples and emerging detection in human serum have warned of their underestimated risks. Studies showed that some Cl-PAHs exhibit dioxin-like properties, implying immunotoxic potential but lacking direct evidence and specific mechanisms. Here, we integrated a high-content screening (HCS) system and high-resolution mass spectrometry to investigate the immune dysfunction and metabolic disruption induced by Cl-PAHs and their parent PAHs (PPAHs) in THP-1 macrophages. Both 9-chloroanthracene and 2,7-dichlorofluorene exerted clear immunosuppression on THP-1 mφs, while their PPAHs exhibited different immune disturbances. Interestingly, Cl-PAH/PPAHs induced complex alterations in the multicytokine/chemokine network, including biphasic alterations with initial inhibition and later enhancement. Furthermore, the protein-protein interaction results revealed that inflammatory cytokines are the core of this complicated network regulation. Connecting immune phenotypes and metabolomics, amino acid metabolism reprogramming was identified as a potential cause of Cl-PAH/PAH-induced immunotoxicity. Phytosphingosine and l-kynurenine were proposed as candidate immunosuppression biomarkers upon Cl-PAH exposure. This article provides direct immunotoxicity evidence of Cl-PAHs without activating AhR for the first time and discusses the contribution of metabolites to Cl-PAH/PPAH-induced immune responses in macrophages, highlighting the potential of developing new methods based on immunometabolism mechanisms for toxic risk evaluation of environmental chemicals.
Collapse
Affiliation(s)
- Xinyan Li
- Guangdong Provincial Key Laboratory of Water Quality Improvement and Ecological Restoration for Watersheds, School of Ecology, Environment and Resources, Guangdong University of Technology, Guangzhou510006, China
- Jieyang Branch of Chemistry and Chemical Engineering Guangdong Laboratory (Rongjiang Laboratory), Jieyang515200, China
| | - Mei Ma
- China Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Bilin Zhao
- Guangdong Provincial Key Laboratory of Water Quality Improvement and Ecological Restoration for Watersheds, School of Ecology, Environment and Resources, Guangdong University of Technology, Guangzhou510006, China
| | - Na Li
- China Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Ling Fang
- Instrumental Analysis & Research Center, Sun Yat-Sen University, Guangzhou510275, China
| | - Donghong Wang
- China Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Tiangang Luan
- Guangdong Provincial Key Laboratory of Water Quality Improvement and Ecological Restoration for Watersheds, School of Ecology, Environment and Resources, Guangdong University of Technology, Guangzhou510006, China
- Jieyang Branch of Chemistry and Chemical Engineering Guangdong Laboratory (Rongjiang Laboratory), Jieyang515200, China
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou510275, China
| |
Collapse
|
12
|
Faisal Madhloom A, Bashir Hashim Al-Taweel F, Sha AM, Raad Abdulbaqi H. Antimicrobial Effect of Moringa Oleifera L. and Red Pomegranate against Clinically Isolated Porphyromonas gingivalis: in vitro Study. ARCHIVES OF RAZI INSTITUTE 2022; 77:1405-1419. [PMID: 36883151 PMCID: PMC9985785 DOI: 10.22092/ari.2022.357513.2051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/01/2022] [Indexed: 03/09/2023]
Abstract
Moringa oleifera L. and red pomegranate extracts have been reported to inhibit gram-positive facultative anaerobe growth and inhibit the formation of biofilm on tooth surfaces. The current study aimed to assess the antibacterial effect of M. oleifera L. and red pomegranate extracts and their combinations against Porphyromonas gingivalis. The antimicrobial sensitivity, minimum inhibition concentrations (MIC), and minimum bactericidal concentrations after treatment with the aqueous extracts of M. oleifera L. and red pomegranate as well as their combination against clinically isolated P. gingivalis were determined using agar well diffusion and two-fold serial dilution. The anti-biofilm activity of the extracts and their combination was evaluated using the tube adhesion method. The phytochemical analysis was carried out using gas chromatography-mass spectrometry. It was found that P. gingivalis was sensitive to aqueous extract of M. oleifera L. seeds and red pomegranate albedo, however, not to M. oleifera L. leaves and red pomegranate seeds. The MIC value of M. oleifera L. seeds, red pomegranate albedo, and their combination were obtained at 12.5 mg/ml, 6.25 mg/ml, and 3.12 mg/ml against P. gingivalis, respectively. The extract combination had the highest anti-biofilm effect than M. oleifera L. seeds and red pomegranate albedo aqueous extracts at the minimum concentrations of 6.25 mg/ml, 25 mg/ml, and 12.5 mg/ml, respectively. The combination of red pomegranate albedo and M. oleifera L. seeds showed superior antibacterial and anti-biofilm effects against P. gingivalis, followed by red pomegranate albedo and M. oleifera L. seeds. This may highlight a promising alternative to the traditional chemicals that can be used as an adjunct in the treatment of periodontal diseases.
Collapse
Affiliation(s)
- A Faisal Madhloom
- Department of Periodontics, College of Dentistry, University of AlKafeel, Najaf, Iraq
| | | | - A M Sha
- Department of Periodontics, College of Dentistry, University of Sulaimani, Sulaymaniyah, Iraq.,Smart Health Tower, Sulaymaniyah, Iraq
| | - H Raad Abdulbaqi
- Department of Periodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
13
|
Yoshino N, Ikeda T, Nakao R. Dual Inhibitory Activity of Petroselinic Acid Enriched in Fennel Against Porphyromonas gingivalis. Front Microbiol 2022; 13:816047. [PMID: 35663901 PMCID: PMC9161081 DOI: 10.3389/fmicb.2022.816047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/15/2022] [Indexed: 11/26/2022] Open
Abstract
Increasing evidence has shown that a major periodontal pathobiont, Porphyromonas gingivalis, triggers oral dysbiosis leading to deterioration not only of periodontal health, but also of several systemic conditions. In the present study we identified remarkable anti-P. gingivalis activity of Foeniculum vulgare (fennel), an herbal plant used in Asian cuisine as well as in traditional medicine, by screening of 92 extracts prepared from 23 edible plants. The n-hexane-extracted fennel (HEF) showed a rapid lethal action toward P. gingivalis, while it was rather ineffective with a wide range of other oral commensal bacterial species. Morphological analysis using both high-speed atomic force microscopy and field emission scanning electron microscopy revealed that a low concentration of HEF (8 μg/mL) resulted in formation of protruding nanostructures composed of outer membrane vesicle (OMV)-like particles, while a high concentration of HEF (64 μg/mL) induced bacteriolysis with overproduction of OMVs with unusual surface properties. Interestingly, HEF treatment resulted in deprivation of two outer membrane transporter proteins, RagA and RagB, which is essential for nutrient acquisition in P. gingivalis, by extracellularly releasing RagA/RagB-enriched OMVs. Furthermore, HEF showed gingipain-inhibitory activity toward both arginine-specific (Rgps) and lysine-specific (Kgp) gingipains, resulting in blocking oral epithelial cell rounding and the subsequent detachment from culture dishes. Finally, we isolated petroselinic acid as a major bactericide as well as a gingipain inhibitor through a bioassay-guided fractionation of HEF. Taken together, our findings suggest clinical applicability of HEF and petroselinic acid for periodontitis therapy to eliminate P. gingivalis and its major virulence factors on the basis of the dual anti-P. gingivalis activity, i.e., rapid bacteriolysis and gingipain inhibition.
Collapse
Affiliation(s)
- Nanami Yoshino
- Department of Bacteriology, National Institute of Infectious Diseases, Tokyo, Japan
- Research and Analysis Center, S&B Foods Inc., Tokyo, Japan
| | - Tsuyoshi Ikeda
- Department of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Ryoma Nakao
- Department of Bacteriology, National Institute of Infectious Diseases, Tokyo, Japan
- *Correspondence: Ryoma Nakao,
| |
Collapse
|
14
|
Mudgil P. Antimicrobial Tear Lipids in the Ocular Surface Defense. Front Cell Infect Microbiol 2022; 12:866900. [PMID: 35433501 PMCID: PMC9008483 DOI: 10.3389/fcimb.2022.866900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/05/2022] Open
Abstract
The concept of antimicrobial lipids as effectors of innate host defense is an emerging field. There is limited knowledge on the antimicrobial role of lipids in the ocular environment. Tears act as first line of defense to protect the ocular surface from infections. Antimicrobial effects of tear lipids have been demonstrated using meibomian lipids that are the source of majority of lipids in tears. This article describes the knowledge available on the antimicrobial role of tear lipids at the ocular surface and the antimicrobial potential of various lipid classes present in tears that can contribute to antimicrobial protection of the eye. Like other mucosal secretions, tears contain many proteins and lipids with known antimicrobial effects. The antimicrobial defense of tears is far stronger than can be demonstrated by the effects of individual compounds many of which are present in low concentrations but synergistic and additive interactions between them provide substantial antimicrobial protection to the ocular surface. It is inferred that antimicrobial lipids play important role in innate defense of tears, and cooperative interactions between various antimicrobial lipids and proteins in tears provide a potent host defense mechanism that is effective against a broad spectrum of pathogens and renders self-sterilizing properties to tears for keeping the microbial load low at the ocular surface.
Collapse
|
15
|
Sa’ad MA, Kavitha R, Fuloria S, Fuloria NK, Ravichandran M, Lalitha P. Synthesis, Characterization and Biological Evaluation of Novel Benzamidine Derivatives: Newer Antibiotics for Periodontitis Treatment. Antibiotics (Basel) 2022; 11:antibiotics11020207. [PMID: 35203811 PMCID: PMC8868241 DOI: 10.3390/antibiotics11020207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/16/2022] Open
Abstract
Periodontal disease (PD) is complex polymicrobial disease which destroys tooth-supporting tissue. Although various synthetic inhibitors of periodontitis-triggering pathogens have been recognized, their undesirable side effects limit their application. Hence, the present study intended to perform the synthesis, characterization, antimicrobial evaluation, and cytotoxicity analysis of novel benzamidine analogues (NBA). This study involved the synthesis of novel imino bases of benzamidine (4a–c), by reacting different aromatic aldehydes with 2-(4-carbamimidoylphenoxy) acetohydrazide (3), which was synthesized by the hydrazination of ethyl 2-(4-carbamimidoylphenoxy) acetate (2), the derivative of 4-hydroxybenzene carboximidamide (1). This was followed by characterization using FTIR, 1H, 13C NMR and mass spectrometry. All synthesized compounds were further tested for antimicrobial potential against PD-triggering pathogens by the micro broth dilution method. The cytotoxicity analysis of the NBA against HEK 293 cells was conducted using an MTT assay. The present study resulted in a successful synthesis of NBA and elucidated their structures. The synthesized NBA exhibited significant antimicrobial activity values between 31.25 and 125 µg/mL against tested pathogens. All NBA exhibited weak cytotoxicity against HEK 293 cells at 7.81 µg, equally to chlorhexidine at 0.2%. The significant antimicrobial activity of NBA against PD-triggering pathogens supports their potential application in periodontitis treatment.
Collapse
Affiliation(s)
- Mohammad Auwal Sa’ad
- Department of Biochemistry, Faculty of Medicine, AIMST University, Bedong 08100, Kedah, Malaysia;
- Centre of Excellence for Vaccine Development (CoEVD), Faculty of Applied Science, AIMST University, Bedong 08100, Kedah, Malaysia
| | - Ramasamy Kavitha
- Department of Biotechnology, Faculty of Applied Science, AIMST University, Bedong 08100, Kedah, Malaysia;
| | - Shivkanya Fuloria
- Centre of Excellence for Biomaterials Engineering, Faculty of Pharmacy, AIMST University, Bedong 08100, Kedah, Malaysia;
| | - Neeraj Kumar Fuloria
- Centre of Excellence for Biomaterials Engineering, Faculty of Pharmacy, AIMST University, Bedong 08100, Kedah, Malaysia;
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospital, Saveetha University, Chennai 600077, India
- Correspondence: (N.K.F.); (M.R.); (P.L.); Tel.: +60-143-034-057 (N.K.F.)
| | - Manickam Ravichandran
- Centre of Excellence for Vaccine Development (CoEVD), Faculty of Applied Science, AIMST University, Bedong 08100, Kedah, Malaysia
- Correspondence: (N.K.F.); (M.R.); (P.L.); Tel.: +60-143-034-057 (N.K.F.)
| | - Pattabhiraman Lalitha
- Department of Biochemistry, Faculty of Medicine, AIMST University, Bedong 08100, Kedah, Malaysia;
- Correspondence: (N.K.F.); (M.R.); (P.L.); Tel.: +60-143-034-057 (N.K.F.)
| |
Collapse
|
16
|
Push Out Bond Strength of a Glass Fibre Post to Root Dentine Pretreated with Proanthocyanidin and Phytosphingosine - An In Vitro Study. Eur Endod J 2021; 6:230-234. [PMID: 34650018 PMCID: PMC8461486 DOI: 10.14744/eej.2021.22931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE To evaluate the push out bond strength of a glass fibre post to root dentine pretreated with 6.5% proanthocyanidin (PAC) and 0.02% phytosphingosine (PHS). METHODS Thirty-three freshly extracted single rooted human teeth were decoronated to a length of 14 mm. Root canals were prepared using rotary NiTi files and obturated with gutta percha and resin sealer. Post space was prepared using peeso reamers, retaining 5 mm of apical gutta percha. Following smear layer removal and acid etching of the post space, samples were randomly assigned to 3 groups based on the dentine pretreatment, namely the control (no pretreatment) group, 6.5% PAC group, and 0.02% PHS group. A glass fibre post was luted using a dual cure adhesive and luting cement. 1 mm thick root slices were sectioned from coronal, middle and apical levels of the post and their push out bond strength was evaluated using a universal testing machine. Data was analysed with one-way ANOVA and Games-Howell post hoc test (P<0.05). RESULTS At all levels, PHS showed higher push out bond strength than PAC and control groups, with a significant difference between the experimental groups at the middle and apical thirds (P<0.05). The push out bond strength of PAC group was significantly higher than the control group in the coronal and apical thirds (P<0.05). CONCLUSION Both PAC and PHS improved the push out bond strength of a glass fibre post to dentine.
Collapse
|
17
|
Abstract
The nares of one in three humans are colonized by Staphylococcus aureus. In these environments, and arguably on all mucosal surfaces, bacteria encounter fatty acids with antimicrobial properties. Our study uncovers that S. aureus releases membrane vesicles (MVs) that act as decoys to protect the bacterium against antimicrobial fatty acids (AFAs). The AFA-neutralizing effects of MVs were neither strain specific nor restricted to one particular AFA. Hence, MVs may represent “public goods” playing an overlooked role in shaping bacterial communities in AFA-rich environments such as the skin and nose. Intriguingly, in addition to MV biogenesis, S. aureus modulates MV composition in response to exposure to AFAs, including an increased release of lipoproteins. These MVs strongly stimulate the innate immunity via Toll-like receptor 2 (TLR2). TLR2-mediated inflammation, which helps to fight infections, may exacerbate inflammatory disorders like atopic dermatitis. Our study highlights intricate immune responses preventing infections from colonizing bacteria. Staphylococcus aureus is a major pathogen, which colonizes one in three otherwise healthy humans. This significant spread of S. aureus is largely due to its ability to circumvent innate immune responses, including antimicrobial fatty acids (AFAs) on the skin and in nasal secretions. In response to AFAs, S. aureus swiftly induces resistance mechanisms, which have yet to be completely elucidated. Here, we identify membrane vesicle (MV) release as a resistance strategy used by S. aureus to sequester host-specific AFAs. MVs protect S. aureus against a wide array of AFAs. Strikingly, beside MV production, S. aureus modulates MV composition upon exposure to AFAs. MVs purified from bacteria grown in the presence of linoleic acid display a distinct protein content and are enriched in lipoproteins, which strongly activate Toll-like receptor 2 (TLR2). Cumulatively, our findings reveal the protective capacities of MVs against AFAs, which are counteracted by an increased TLR2-mediated innate immune response. IMPORTANCE The nares of one in three humans are colonized by Staphylococcus aureus. In these environments, and arguably on all mucosal surfaces, bacteria encounter fatty acids with antimicrobial properties. Our study uncovers that S. aureus releases membrane vesicles (MVs) that act as decoys to protect the bacterium against antimicrobial fatty acids (AFAs). The AFA-neutralizing effects of MVs were neither strain specific nor restricted to one particular AFA. Hence, MVs may represent “public goods” playing an overlooked role in shaping bacterial communities in AFA-rich environments such as the skin and nose. Intriguingly, in addition to MV biogenesis, S. aureus modulates MV composition in response to exposure to AFAs, including an increased release of lipoproteins. These MVs strongly stimulate the innate immunity via Toll-like receptor 2 (TLR2). TLR2-mediated inflammation, which helps to fight infections, may exacerbate inflammatory disorders like atopic dermatitis. Our study highlights intricate immune responses preventing infections from colonizing bacteria.
Collapse
|
18
|
Antimicrobial Prosthetic Surfaces in the Oral Cavity-A Perspective on Creative Approaches. Microorganisms 2020; 8:microorganisms8081247. [PMID: 32824437 PMCID: PMC7463865 DOI: 10.3390/microorganisms8081247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
Replacement of missing teeth is an essential component of comprehensive dental care for patients suffering of edentulism. A popular option is implant-supported restorations. However, implant surfaces can become colonized with polymicrobial biofilms containing Candida species that may compromise peri-implant health. To prevent this, implant components may be treated with a variety of coatings to create surfaces that either repel the attachment of viable microorganisms or kill microorganisms on contact. These coatings may consist of nanoparticles of pure elements (more commonly silver, copper, and zinc), sanitizing agents and disinfectants (quaternary ammonium ions and chlorhexidine), antibiotics (cefalotin, vancomycin, and gentamicin), or antimicrobial peptides (AMPs). AMPs in bioactive coatings have a number of advantages. They elicit a protective action against pathogens, inhibit the formation of biofilms, are less toxic to host tissues, and do not prompt inflammatory responses. Furthermore, many of these coatings may involve unique delivery systems to direct their antimicrobial capacity against pathogens, but not commensals. Coatings may also contain multiple antimicrobial substances to widen antimicrobial activity across multiple microbial species. Here, we compiled relevant information about a variety of creative approaches used to generate antimicrobial prosthetic surfaces in the oral cavity with the purpose of facilitating implant integration and peri-implant tissue health.
Collapse
|
19
|
Amorim AA, de Arruda CNF, Vivanco RG, Bikker F, de Pires-de-Souza FCP. Effect of phytosphingosine on staining resistance and microhardness of tooth enamel. J ESTHET RESTOR DENT 2020; 33:294-302. [PMID: 32445541 DOI: 10.1111/jerd.12599] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/23/2020] [Accepted: 05/08/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To evaluate the protective effect of phytosphingosine (PHS) against staining on dental enamel. MATERIALS AND METHODS Ninety-six specimens of bovine teeth (6 mm × 6 mm × 2 mm) were cut, and initial color (Easyshade, VITA), microhardness (HMV-2, Shimadzu) and fluorescence (Matlabs software, Matworks) measurements were performed. Specimens were separated into four groups according to the treatments: Distilled water (control); Human saliva (HS); PHS; PHS + HS. Specimens (n = 6) were submitted to staining procedures: Distilled water (immersion for 30 days-control); Coffee (15 minutes, twice a day/for 15 days); Black tea (as described for Coffee) and cigarette smoke (20 cigarettes/sample). Final measurements were performed, and data were analyzed (Color-CIEDE2000, fluorescence-2-way ANOVA, Tukey, and microhardness-Kruskal-Wallis Dunn, P < .05). RESULTS Coffee caused the highest color change (ΔE00 ), followed by black tea, regardless of the treatment employed. Distilled water and cigarette smoke produced similar color changes (P > .05) for the groups control (water = 1.0 + - 0.5/ cigarette = 2.3 0.3) and PHS (water = 0.8 0.4/ cigarette = 2.3 0.4). PHS + HS demonstrated intermediate means than PHS and HS when stained with coffee and tea. After treatments, the least fluorescence alterations occurred for the groups treated with distilled water and cigarette, regardless of the treatment, with no difference (P > .05) between them. There was a significant difference (P < .05) on microhardness between all the groups, as PHS + HS > PHS > HS > Distilled water. CONCLUSIONS It was concluded that PHS treatment did not protect the staining of the enamel by coffee and tea, but increased the microhardness, both in the presence and absence of a salivary pellicle. PRACTICAL IMPLICATIONS Phytoshingosine is a novel agent and considered a promising component for anti-biofilm and anti-erosion properties by the formation of a diffusion barrier on the dental enamel. In line, PHS might be considered for anti-staining purposes.
Collapse
Affiliation(s)
- Ayodele Alves Amorim
- Department of Dental Materials and Prosthodontics, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo., Ribeirão Preto, São Paulo, Brazil
| | - Carolina Noronha Ferraz de Arruda
- Department of Dental Materials and Prosthodontics, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo., Ribeirão Preto, São Paulo, Brazil
| | - Rocio Geng Vivanco
- Department of Dental Materials and Prosthodontics, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo., Ribeirão Preto, São Paulo, Brazil
| | - Floris Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | | |
Collapse
|
20
|
Fischer CL. Antimicrobial Activity of Host-Derived Lipids. Antibiotics (Basel) 2020; 9:E75. [PMID: 32054068 PMCID: PMC7168235 DOI: 10.3390/antibiotics9020075] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023] Open
Abstract
Host-derived lipids are increasingly recognized as antimicrobial molecules that function in innate immune activities along with antimicrobial peptides. Sphingoid bases and fatty acids found on the skin, in saliva and other body fluids, and on all mucosal surfaces, including oral mucosa, exhibit antimicrobial activity against a variety of Gram positive and Gram negative bacteria, viruses, and fungi, and reduce inflammation in animal models. Multiple studies demonstrate that the antimicrobial activity of lipids is both specific and selective. There are indications that the site of action of antimicrobial fatty acids is the bacterial membrane, while the long-chain bases may inhibit cell wall synthesis as well as interacting with bacterial membranes. Research in this area, although still sporadic, has slowly increased in the last few decades; however, we still have much to learn about antimicrobial lipid mechanisms of activity and their potential use in novel drugs or topical treatments. One important potential benefit for the use of innate antimicrobial lipids (AMLs) as antimicrobial agents is the decreased likelihood side effects with treatment. Multiple studies report that endogenous AML treatments do not induce damage to cells or tissues, often decrease inflammation, and are active against biofilms. The present review summarizes the history of antimicrobial lipids from the skin surface, including both fatty acids and sphingoid bases, in multiple human body systems and summarizes their relative activity against various microorganisms. The range of antibacterial activities of lipids present at the skin surface and in saliva is presented. Some observations relevant to mechanisms of actions are discussed, but are largely still unknown. Multiple recent studies examine the therapeutic and prophylactic uses of AMLs. Although these lipids have been repeatedly demonstrated to act as innate effector molecules, they are not yet widely accepted as such. These compiled data further support fatty acid and sphingoid base inclusion as innate effector molecules.
Collapse
Affiliation(s)
- Carol L Fischer
- Biology Department, Waldorf University, Forest City, IA 50436, USA
| |
Collapse
|
21
|
Beck S, Sehl C, Voortmann S, Verhasselt HL, Edwards MJ, Buer J, Hasenberg M, Gulbins E, Becker KA. Sphingosine is able to prevent and eliminate Staphylococcus epidermidis biofilm formation on different orthopedic implant materials in vitro. J Mol Med (Berl) 2019; 98:209-219. [PMID: 31863153 PMCID: PMC7007894 DOI: 10.1007/s00109-019-01858-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/11/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022]
Abstract
Abstract Periprosthetic infection (PPI) is a devastating complication in joint replacement surgery. On the background of an aging population, the number of joint replacements and associated complications is expected to increase. The capability for biofilm formation and the increasing resistance of different microbes to antibiotics have complicated the treatment of PPI, requiring the need for the development of alternative treatment options. The bactericidal effect of the naturally occurring amino alcohol sphingosine has already been reported. In our study, we demonstrate the antimicrobial efficacy of sphingosine on three different strains of biofilm producing Staphylococcus epidermidis, representing one of the most frequent microbes involved in PPI. In an in vitro analysis, sphingosine’s capability for prevention and treatment of biofilm-contamination on different common orthopedic implant surfaces was tested. Coating titanium implant samples with sphingosine not only prevented implant contamination but also revealed a significant reduction of biofilm formation on the implant surfaces by 99.942%. When testing the antimicrobial efficacy of sphingosine on sessile biofilm-grown Staphylococcus epidermidis, sphingosine solution was capable to eliminate 99.999% of the bacteria on the different implant surfaces, i.e., titanium, steel, and polymethylmethacrylate. This study provides evidence on the antimicrobial efficacy of sphingosine for both planktonic and sessile biofilm-grown Staphylococcus epidermidis on contaminated orthopedic implants. Sphingosine may provide an effective and cheap treatment option for prevention and reduction of infections in joint replacement surgery. Key messages • Here we established a novel technology for prevention of implant colonization by sphingosine-coating of orthopedic implant materials. • Sphingosine-coating of orthopedic implants prevented bacterial colonization and significantly reduced biofilm formation on implant surfaces by 99.942%. • Moreover, sphingosine solution was capable to eliminate 99.999% of sessile biofilm-grown Staphylococcus epidermidis on different orthopedic implant surfaces.
Collapse
Affiliation(s)
- Sascha Beck
- Department of Molecular Biology, Medical School Essen, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany. .,Department of Orthopedics and Orthopedic Surgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany.
| | - Carolin Sehl
- Department of Molecular Biology, Medical School Essen, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Sylvia Voortmann
- Institute for Experimental Immunology and Imaging, Medical Research Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hedda Luise Verhasselt
- Institute of Medical Microbiology, Medical School Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Jan Buer
- Institute of Medical Microbiology, Medical School Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mike Hasenberg
- Institute for Experimental Immunology and Imaging, Medical Research Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, Medical School Essen, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Department of Surgery, University of Cincinnati, Cincinnati, USA
| | - Katrin Anne Becker
- Department of Molecular Biology, Medical School Essen, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
22
|
Protective properties of milk sphingomyelin against dysfunctional lipid metabolism, gut dysbiosis, and inflammation. J Nutr Biochem 2019; 73:108224. [DOI: 10.1016/j.jnutbio.2019.108224] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/25/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022]
|
23
|
Ikeda E, Shiba T, Ikeda Y, Suda W, Nakasato A, Takeuchi Y, Azuma M, Hattori M, Izumi Y. Japanese subgingival microbiota in health vs disease and their roles in predicted functions associated with periodontitis. Odontology 2019; 108:280-291. [PMID: 31502122 DOI: 10.1007/s10266-019-00452-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
The present study aimed to identify and compare the microbial signatures between periodontally healthy and periodontitis subjects using 454 sequences of 16S rRNA genes. Subgingival plaque samples were collected from ten periodontally healthy subjects and ten matched chronic periodontitis patients. Bacterial DNA was extracted and next-generation sequencing of 16S rRNA genes was performed. The microbial composition differed between healthy subjects and periodontitis patients at all phylogenetic levels. Particularly, 16 species, including Lautropia mirabilis and Neisseria subflava predominated in healthy subjects, whereas nine species, including Porphyromonas gingivalis and Filifactor alocis predominated in periodontitis. UniFrac, a principal coordinate and network analysis, confirmed distinct community profiles in healthy subjects and periodontitis patients. Using predicted function profiling, pathways involved in phenylpropanoid, GPI-anchor biosynthesis, and metabolism of alanine, arginine, aspartate, butanoate, cyanoamino acid, fatty acid, glutamate, methane, proline, and vitamin B6 were significantly over-represented in periodontitis patients. These results highlight the oral microbiota alterations in microbial composition in periodontitis and suggest the genes and metabolic pathways associated with health and periodontitis. Our findings help to further elucidate microbial composition and interactions in health and periodontitis.
Collapse
Affiliation(s)
- Eri Ikeda
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Takahiko Shiba
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Yuichi Ikeda
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-0016, Japan
| | - Akinori Nakasato
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Yasuo Takeuchi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan.
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Masahira Hattori
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Faculty of Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo, 169-8555, Japan
| | - Yuichi Izumi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan.,Oral Care Perio Center, Southern TOHOKU General Hospital, Southern TOHOKU Research Institute for Neuroscience, 7-115 Yatsuyamada, Koriyama, Fukushima, 963-8052, Japan
| |
Collapse
|
24
|
Seitz AP, Schumacher F, Baker J, Soddemann M, Wilker B, Caldwell CC, Gobble RM, Kamler M, Becker KA, Beck S, Kleuser B, Edwards MJ, Gulbins E. Sphingosine-coating of plastic surfaces prevents ventilator-associated pneumonia. J Mol Med (Berl) 2019; 97:1195-1211. [PMID: 31222488 PMCID: PMC6647234 DOI: 10.1007/s00109-019-01800-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/01/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
Ventilator-associated pneumonia (VAP) is a major cause of morbidity and mortality in critically ill patients. Here, we employed the broad antibacterial effects of sphingosine to prevent VAP by developing a novel method of coating surfaces of endotracheal tubes with sphingosine and sphingosine analogs. Sphingosine and phytosphingosine coatings of endotracheal tubes prevent adherence and mediate killing of Pseudomonas aeruginosa, Acinetobacter baumannii, and Staphylococcus aureus, even in biofilms. Most importantly, sphingosine-coating of endotracheal tubes also prevented P. aeruginosa and S. aureus pneumonia in vivo. Coating of the tubes with sphingosine was stable, without obvious side effects on tracheal epithelial cells and did not induce inflammation. In summary, we describe a novel method to coat plastic surfaces and provide evidence for the application of sphingosine and phytosphingosine as novel antimicrobial coatings to prevent bacterial adherence and induce killing of pathogens on the surface of endotracheal tubes with potential to prevent biofilm formation and VAP. KEY MESSAGES: Novel dip-coating method to coat plastic surfaces with lipids. Sphingosine and phytosphingosine as novel antimicrobial coatings on plastic surface. Sphingosine coatings of endotracheal tubes prevent bacterial adherence and biofilms. Sphingosine coatings of endotracheal tubes induce killing of pathogens. Sphingosine coatings of endotracheal tubes ventilator-associated pneumonia.
Collapse
Affiliation(s)
- Aaron P Seitz
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA.
| | - Fabian Schumacher
- Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.,Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Jennifer Baker
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA
| | - Matthias Soddemann
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Barbara Wilker
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Charles C Caldwell
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA.,Division of Research, Shriners Hospital for Children, Cincinnati, OH, 45229, USA
| | - Ryan M Gobble
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA
| | - Markus Kamler
- Thoracic Transplantation, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Sascha Beck
- Orthopedic Surgery, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Burkhard Kleuser
- Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Michael J Edwards
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA
| | - Erich Gulbins
- Department of Surgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way ML 0558, Cincinnati, OH, 45267, USA. .,Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
25
|
Baker JE, Boudreau RM, Seitz AP, Caldwell CC, Gulbins E, Edwards MJ. Sphingolipids and Innate Immunity: A New Approach to Infection in the Post-Antibiotic Era? Surg Infect (Larchmt) 2018; 19:792-803. [DOI: 10.1089/sur.2018.187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Jennifer E. Baker
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Ryan M. Boudreau
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Aaron P. Seitz
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Charles C. Caldwell
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
- Division of Research, Shriners Hospital for Children, Cincinnati, Ohio
| | - Erich Gulbins
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael J. Edwards
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
26
|
Wang Y, Zhang Y, Shi YQ, Pan XH, Lu YH, Cao P. Antibacterial effects of cinnamon (Cinnamomum zeylanicum) bark essential oil on Porphyromonas gingivalis. Microb Pathog 2018; 116:26-32. [PMID: 29325862 DOI: 10.1016/j.micpath.2018.01.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 12/23/2022]
Abstract
The objective of this study was to investigate the antibacterial effects of cinnamon (Cinnamomum zeylanicum) bark essential oil (CBEO) and its principal constituent cinnamaldehyde against Porphyromonas gingivalis and to elucidate the antibacterial mechanism. GC-MS analysis showed that cinnamaldehyde was the major constituent in CBEO (57.97%). The minimum inhibition concentrations (MICs) of CBEO and cinnamaldehyde were 6.25 μg/mL and 2.5 μM for P. gingivalis, respectively. Nucleic acid and protein leakage was observed with increasing concentrations of CBEO and cinnamaldehyde. Additionally, propidium iodide uptake assays revealed CBEO and cinnamaldehyde at 1 × MIC impaired P. gingivalis membrane integrity by enhancing cell permeability. Morphological changes in P. gingivalis cells were observed by scanning electron microscopy, which indicated cell membrane destruction. To further determine the anti-biofilm effect, relative biofilm formation and established biofilms were examined, which demonstrated that both CBEO and cinnamaldehyde at sub-MIC levels inhibited P. gingivalis biofilm formation by 74.5% and 67.3% separately, but only CBEO slightly decreased established biofilms by 33.5% at 4 × MIC. These results suggest the potential of CBEO as a natural antimicrobial agent against periodontal disease. Furthermore, cinnamaldehyde was confirmed to be the antibacterial substance of CBEO with inhibitory action against P. gingivalis.
Collapse
Affiliation(s)
- Yue Wang
- Shanghai Institute of Technology, Shanghai 201418, China
| | - Yi Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yan-Qin Shi
- Shanghai Institute of Technology, Shanghai 201418, China
| | - Xian-Hua Pan
- Shanghai Institute of Technology, Shanghai 201418, China
| | - Yan-Hua Lu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ping Cao
- Shanghai Jahwa (Group) Co., Ltd., Shanghai 200082, China.
| |
Collapse
|
27
|
Das UN. Arachidonic acid and other unsaturated fatty acids and some of their metabolites function as endogenous antimicrobial molecules: A review. J Adv Res 2018; 11:57-66. [PMID: 30034876 PMCID: PMC6052656 DOI: 10.1016/j.jare.2018.01.001] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/01/2018] [Accepted: 01/01/2018] [Indexed: 12/13/2022] Open
Abstract
Our body is endowed with several endogenous anti-microbial compounds such as interferon, cytokines, free radicals, etc. However, little attention has been paid to the possibility that lipids could function as antimicrobial compounds. In this short review, the antimicrobial actions of various polyunsaturated fatty acids (PUFAs, mainly free acids) and their putative mechanisms of action are described. In general, PUFAs kill microbes by their direct action on microbial cell membranes, enhancing generation of free radicals, augmenting the formation of lipid peroxides that are cytotoxic, and by increasing the formation of their bioactive metabolites, such as prostaglandins, lipoxins, resolvins, protectins and maresins that enhance the phagocytic action of leukocytes and macrophages. Higher intakes of α-linolenic and cis-linoleic acids (ALA and LA respectively) and fish (a rich source of eicosapentaenoic acid and docosahexaenoic acid) might reduce the risk pneumonia. Previously, it was suggested that polyunsaturated fatty acids (PUFAs): linoleic, α-linolenic, γ-linolenic (GLA), dihomo-GLA (DGLA), arachidonic (AA), eicosapentaenoic (EPA), and docosahexaenoic acids (DHA) function as endogenous anti-bacterial, anti-fungal, anti-viral, anti-parasitic, and immunomodulating agents. A variety of bacteria are sensitive to the growth inhibitory actions of LA and ALA in vitro. Hydrolyzed linseed oil can kill methicillin-resistant Staphylococcus aureus. Both LA and AA have the ability to inactivate herpes, influenza, Sendai, and Sindbis virus within minutes of contact. AA, EPA, and DHA induce death of Plasmodium falciparum both in vitro and in vivo. Prostaglandin E1 (PGE1) and prostaglandin A (PGA), derived from DGLA, AA, and EPA inhibit viral replication and show anti-viral activity. Oral mucosa, epidermal cells, lymphocytes and macrophages contain and release significant amounts of PUFAs on stimulation. PUFAs stimulate NADPH-dependent superoxide production by macrophages, neutrophils and lymphocytes to kill the invading microorganisms. Cytokines induce the release of PUFAs from cell membrane lipid pool, a potential mechanism for their antimicrobial action. AA, EPA, and DHA give rise to lipoxins (LXs), resolvins, protectins, and maresins that limit and resolve inflammation and have antimicrobial actions. Thus, PUFAs and their metabolites have broad antimicrobial actions.
Collapse
|
28
|
Bikker FJ, Hoogenkamp MA, Malhaoui A, Nazmi K, Neilands J, Krom BP. Phytosphingosine Prevents the Formation of Young Salivary Biofilms in vitro. Caries Res 2017; 52:7-13. [DOI: 10.1159/000480712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022] Open
Abstract
Dental biofilms are formed in a multistep process that is initiated by the adhesion of oral bacteria to the dental hard surface. As dental biofilms are associated with oral diseases their control is necessary in order to maintain oral health. Recently, it was revealed that phytosphingosine (PHS)-treated hydroxyapatite discs showed anti-adhesive activity in a static in vitro biofilm model against Streptococcus mutans. The goal of the present study was to further unravel the anti-adhesive and anti-biofilm properties of PHS in both static and dynamic in vitro biofilm models against a full salivary inoculum. After 3 h under static conditions, bacterial adherence on PHS-treated cover glass slides was reduced by 60% compared to the untreated surface. After 6 and 24 h under static conditions, no significant differences in bacterial adherence were observed between PHS-treated and untreated cover glass slides. However, under dynamic conditions, i.e., the presence of shear forces, virtually no bacterial adherence was observed for up to 16 h on PHS-coated surfaces. Besides, PHS showed a strong bactericidal activity on salivary biofilms. Treatment of a 3- and 6-h statically grown biofilm resulted in a 99 and 94% reduction of viable cells, respectively, which was effectuated within minutes. In principle, these anti-adherence and anti-biofilm properties make PHS a promising candidate ingredient for use in oral care products aimed at oral microbial control.
Collapse
|
29
|
Garaicoa JL, Fischer CL, Bates AM, Holloway J, Avila-Ortiz G, Guthmiller JM, Johnson GK, Stanford C, Brogden KA. Promise of Combining Antifungal Agents in Denture Adhesives to Fight Candida Species Infections. J Prosthodont 2016; 27:755-762. [PMID: 27870138 DOI: 10.1111/jopr.12565] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2016] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Several complications may arise in patients wearing complete prosthetic appliances, including denture-associated infections and mucosal stomatitis due to Candida species. This study evaluated the activity of anti-Candida agents in denture adhesive and the cytotoxicities of these preparations for primary human gingival epithelial (GE) keratinocytes. MATERIALS AND METHODS The anti-Candida activities of antimicrobial peptides, antimicrobial lipids, and antifungal agents against C. albicans ATCC 64124 or HMV4C were assessed in microdilution assays containing water or 1% denture adhesive. The minimal inhibitory concentrations (MIC) and the minimal bactericidal concentrations (MBC) were determined. The cytotoxicities of denture adhesive compounded with these agents were assessed in 1.0 × 105 primary GE keratinocytes in LGM-3 media with resazurin. RESULTS Lactoferricin B, SMAP28, sphingosine, dihydrosphingosine, and phytosphingosine in 1% denture adhesive lost antimicrobial activity for C. albicans (p < 0.05). Amphotericin B, chlorhexidine dihydrochloride, chlorhexidine gluconate, fluconazole, and nystatin in 1% denture adhesive or compounded directly into denture adhesive and then diluted to 1% adhesive, did not lose antimicrobial activity. Compounded formulations were not cytotoxic (LD50 > 100.0 μg/ml) against primary human GE keratinocytes. CONCLUSIONS Antimicrobial peptides and antimicrobial lipids had diminished activities in 1% adhesive, suggesting that components in adhesives may inactivate local innate immune factors in the oral cavity, possibly predisposing denture wearers to Candida species infections. More importantly, antifungal agents retained their anti-C. albicans activities in denture adhesive, strongly suggesting that antifungal agents could be candidates for inclusion in adhesive formulations and used as prescribed topical treatments for individuals with denture stomatitis.
Collapse
Affiliation(s)
- Jorge L Garaicoa
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR
| | - Carol L Fischer
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA
| | - Amber M Bates
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA
| | - Julie Holloway
- Department of Prosthodontics, College of Dentistry, The University of Iowa, Iowa City, IA
| | - Gustavo Avila-Ortiz
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA
| | - Janet M Guthmiller
- College of Dentistry, University of Nebraska Medical Center, Lincoln, NE
| | - Georgia K Johnson
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA
| | - Clark Stanford
- College of Dentistry, University of Illinois at Chicago, IL
| | - Kim A Brogden
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA
| |
Collapse
|
30
|
Qiu W, Ren B, Dai H, Zhang L, Zhang Q, Zhou X, Li Y. Clotrimazole and econazole inhibit Streptococcus mutans biofilm and virulence in vitro. Arch Oral Biol 2016; 73:113-120. [PMID: 27764679 DOI: 10.1016/j.archoralbio.2016.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 09/01/2016] [Accepted: 10/11/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The aim of this study was to determine the inhibitory effect of eight antifungal drugs on S. mutans growth, biofilm formation and virulence factors. METHODS The actions of antifungal drugs on S. mutans were determined by recovery plates and survival kinetic curves. Biofilms were observed by scanning electron microscopy and the viable cells were recovered on BHI plates, meanwhile biofilms were stained by BacLight live/dead kit to investigate the biofilm viability. Bacteria/extracellular polysaccharides staining assays were performed to determine the EPS production of S. mutans biofilms. Acidogenicity and acidurity of S. mutans were determined using pH drop and acid tolerance assays, and the expression of ldh gene was evaluated using qPCR. RESULTS We found that clotrimazole (CTR) and econazole (ECO) showed antibacterial activities on S. mutans UA159 and S. mutans clinical isolates at 12.5 and 25mg/L, respectively. CTR and ECO could also inhibit S. mutans biofilm formation and reduce the viability of preformed biofilm. CTR and ECO affected the live/dead ratio and the EPS/bacteria ratio of S. mutans biofilms. CTR and ECO also inhibited the pH drop, lactate acid production, and acid tolerance. The abilities of CTR and ECO to inhibit S. mutans ldh expression were also confirmed. CONCLUSIONS We found that two antifungal azoles, CTR and ECO, had the abilities to inhibit the growth and biofilm formation of S. mutans and more importantly, they could also inhibit the virulence factors of S. mutans.
Collapse
Affiliation(s)
- Wei Qiu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huanqin Dai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lixin Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Qiong Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
31
|
Chen W, Gao B, Hao L, Zhu G, Jules J, Macdougall MJ, Han X, Zhou X, Li YP. The silencing of cathepsin K used in gene therapy for periodontal disease reveals the role of cathepsin K in chronic infection and inflammation. J Periodontal Res 2016; 51:647-60. [PMID: 26754272 PMCID: PMC5482270 DOI: 10.1111/jre.12345] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2015] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVE Periodontitis is a severe chronic inflammatory disease and one of the most prevalent non-communicable chronic diseases that affects the majority of the world's adult population. While great efforts have been devoted toward understanding the pathogenesis of periodontitis, there remains a pressing need for developing potent therapeutic strategies for targeting this dreadful disease. In this study, we utilized adeno-associated virus (AAV) expressing cathepsin K (Ctsk) small hairpin (sh)RNA (AAV-sh-Ctsk) to silence Ctsk in vivo and subsequently evaluated its impact in periodontitis as a potential therapeutic strategy for this disease. MATERIAL AND METHODS We used a known mouse model of periodontitis, in which wild-type BALB/cJ mice were infected with Porphyromonas gingivalis W50 in the maxillary and mandibular periodontium to induce the disease. AAV-sh-Ctsk was then administrated locally into the periodontal tissues in vivo, followed by analyses to assess progression of the disease. RESULTS AAV-mediated Ctsk silencing drastically protected mice (> 80%) from P. gingivalis-induced bone resorption by osteoclasts. In addition, AAV-sh-Ctsk administration drastically reduced inflammation by impacting the expression of many inflammatory cytokines as well as T-cell and dendritic cell numbers in periodontal lesions. CONCLUSION AAV-mediated Ctsk silencing can simultaneously target both the inflammation and bone resorption associated with periodontitis through its inhibitory effect on immune cells and osteoclast function. Thereby, AAV-sh-Ctsk administration can efficiently protect against periodontal tissue damage and alveolar bone loss, establishing this AAV-mediated local silencing of Ctsk as an important therapeutic strategy for effectively treating periodontal disease.
Collapse
Affiliation(s)
- Wei Chen
- Department of Pathology, University of Alabama at Birmingham, SHEL 810, 1825 University Blvd, Birmingham AL 35294-2182, USA
| | - Bo Gao
- Department of Pathology, University of Alabama at Birmingham, SHEL 810, 1825 University Blvd, Birmingham AL 35294-2182, USA
- The State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Liang Hao
- Department of Pathology, University of Alabama at Birmingham, SHEL 810, 1825 University Blvd, Birmingham AL 35294-2182, USA
- The State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Guochun Zhu
- Department of Pathology, University of Alabama at Birmingham, SHEL 810, 1825 University Blvd, Birmingham AL 35294-2182, USA
| | - Joel Jules
- Department of Pathology, University of Alabama at Birmingham, SHEL 810, 1825 University Blvd, Birmingham AL 35294-2182, USA
| | - Mary J. Macdougall
- Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, SDB Room 702, 1919 7 Avenue South, Birmingham AL 35233, USA
| | - Xiaozhe Han
- Department of Immunology and Infectious Disease, The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA
| | - Xuedong Zhou
- The State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yi-Ping Li
- Department of Pathology, University of Alabama at Birmingham, SHEL 810, 1825 University Blvd, Birmingham AL 35294-2182, USA
| |
Collapse
|
32
|
Antibacterial effects of Lactobacillus and bacteriocin PLNC8 αβ on the periodontal pathogen Porphyromonas gingivalis. BMC Microbiol 2016; 16:188. [PMID: 27538539 PMCID: PMC4990846 DOI: 10.1186/s12866-016-0810-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/12/2016] [Indexed: 11/18/2022] Open
Abstract
Background The complications in healthcare systems associated with antibiotic-resistant microorganisms have resulted in an intense search for new effective antimicrobials. Attractive substances from which novel antibiotics may be developed are the bacteriocins. These naturally occurring peptides are generally considered to be safe and efficient at eliminating pathogenic bacteria. Among specific keystone pathogens in periodontitis, Porphyromonas gingivalis is considered to be the most important pathogen in the development and progression of chronic inflammatory disease. The aim of the present study was to investigate the antimicrobial effects of different Lactobacillus species and the two-peptide bacteriocin PLNC8 αβ on P. gingivalis. Results Growth inhibition of P. gingivalis was obtained by viable Lactobacillus and culture media from L. plantarum NC8 and 44048, but not L. brevis 30670. The two-peptide bacteriocin from L. plantarum NC8 (PLNC8 αβ) was found to be efficient against P. gingivalis through binding followed by permeabilization of the membranes, using Surface plasmon resonance analysis and DNA staining with Sytox Green. Liposomal systems were acquired to verify membrane permeabilization by PLNC8 αβ. The antimicrobial activity of PLNC8 αβ was found to be rapid (1 min) and visualized by TEM to cause cellular distortion through detachment of the outer membrane and bacterial lysis. Conclusion Soluble or immobilized PLNC8 αβ bacteriocins may be used to prevent P. gingivalis colonization and subsequent pathogenicity, and thus supplement the host immune system against invading pathogens associated with periodontitis. Electronic supplementary material The online version of this article (doi:10.1186/s12866-016-0810-8) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Holdren GO, Rosenthal DJ, Yang J, Bates AM, Fischer CL, Zhang Y, Brogden NK, Brogden KA. Antimicrobial Activity of Chemokine CXCL10 for Dermal and Oral Microorganisms. Antibiotics (Basel) 2016; 3:527-39. [PMID: 25859394 PMCID: PMC4387564 DOI: 10.3390/antibiotics3040527] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
CXCL10 (IP-10) is a small 10 kDa chemokine with antimicrobial activity. It is induced by IFN-γ, chemoattracts mononuclear cells, and promotes adhesion of T cells. Recently, we detected CXCL10 on the surface of the skin and in the oral cavity. In the current study, we used broth microdilution and radial diffusion assays to show that CXCL10 inhibits the growth of Escherichia coli, Staphylococcus aureus, Corynebacterium jeikeium, Corynebacterium striatum, and Candida albicans HMV4C, but not Corynebacterium bovis, Streptococcus mutans, Streptococcus mitis, Streptococcus sanguinis, Fusobacterium nucleatum, Aggregatibacter actinomycetemcomitans, Poryphromonas gingivalis, or C. albicans ATCC 64124. The reason for the selective antimicrobial activity is not yet known. However, antimicrobial activity of CXCL10 may be related to its composition and structure, as a cationic 98 amino acid residue molecule with 10 lysine residues, 7 arginine residues, a total net charge of +11, and a theoretical pI of 9.93. Modeling studies revealed that CXCL10 contains an α-helix at the N-terminal, three anti-parallel β-strands in the middle, and an α-helix at the C-terminal. Thus, CXCL10, when produced on the surface of the skin or in the oral cavity, likely has antimicrobial activity and may enhance innate antimicrobial and cellular responses to the presence of select commensal or opportunistic microorganisms.
Collapse
Affiliation(s)
- Grant O. Holdren
- Division of Pharmaceutics and Translational Therapeutics, Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA; E-Mails: (G.O.H.); (N.K.B.)
| | - David J. Rosenthal
- Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA; E-Mails: (D.J.R.); (A.M.B.); (C.L.F.)
| | - Jianyi Yang
- Department of Computational Medicine and Bioinformatics, The University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA; E-Mails: (J.Y.); (Y.Z.)
| | - Amber M. Bates
- Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA; E-Mails: (D.J.R.); (A.M.B.); (C.L.F.)
| | - Carol L. Fischer
- Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA; E-Mails: (D.J.R.); (A.M.B.); (C.L.F.)
| | - Yang Zhang
- Department of Computational Medicine and Bioinformatics, The University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA; E-Mails: (J.Y.); (Y.Z.)
| | - Nicole K. Brogden
- Division of Pharmaceutics and Translational Therapeutics, Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA; E-Mails: (G.O.H.); (N.K.B.)
| | - Kim A. Brogden
- Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA; E-Mails: (D.J.R.); (A.M.B.); (C.L.F.)
- Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-319-335-8077; Fax: +1-319-335-8895
| |
Collapse
|
34
|
Inactivation of Escherichia coli O157:H7 in vitro and on the surface of spinach leaves by biobased antimicrobial surfactants. Food Control 2016. [DOI: 10.1016/j.foodcont.2015.07.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
35
|
Qiu W, Zheng X, Wei Y, Zhou X, Zhang K, Wang S, Cheng L, Li Y, Ren B, Xu X, Li Y, Li M. d-Alanine metabolism is essential for growth and biofilm formation of Streptococcus mutans. Mol Oral Microbiol 2015; 31:435-44. [PMID: 26526529 DOI: 10.1111/omi.12146] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2015] [Indexed: 02/05/2023]
Abstract
Part of the d-alanine (d-Ala) metabolic pathway in bacteria involves the conversion of l-alanine to d-Ala by alanine racemase and the formation of d-alanyl-d-alanine by d-alanine-d-alanine ligase, the product of which is involved in cell wall peptidoglycan synthesis. At present, drugs that target the metabolic pathway of d-Ala are already in clinical use - e.g. d-cycloserine (DCS) is used as an antibiotic against Mycobacterium tuberculosis. Streptococcus mutans is the main cariogenic bacterium in the oral cavity. Its d-Ala metabolism-associated enzymes alanine racemase and d-alanine-d-alanine ligase are encoded by the genes smu.1834 and smu.599, respectively, which may be potential targets for inhibitors. In this study, the addition of DCS blocked the d-Ala metabolic pathway in S. mutans, leading to bacterial cell wall defects, significant inhibition of bacterial growth and biofilm formation, and reductions in extracellular polysaccharide production and bacterial adhesion. However, the exogenous addition of d-Ala could reverse the inhibitory effect of DCS. Through the means of drug regulation, our study demonstrated, for the first time, the importance of d-Ala metabolism in the survival and biofilm formation of S. mutans. If the growth of S. mutans can be specifically inhibited by designing drugs that target d-Ala metabolism, then this may serve as a potential new treatment for dental caries.
Collapse
Affiliation(s)
- W Qiu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - X Zheng
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Wei
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Zhou
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - K Zhang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - S Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Cheng
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - B Ren
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - X Xu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - M Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Protein Analysis of Sapienic Acid-Treated Porphyromonas gingivalis Suggests Differential Regulation of Multiple Metabolic Pathways. J Bacteriol 2015; 198:157-67. [PMID: 26483519 DOI: 10.1128/jb.00665-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/02/2015] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Lipids endogenous to skin and mucosal surfaces exhibit potent antimicrobial activity against Porphyromonas gingivalis, an important colonizer of the oral cavity implicated in periodontitis. Our previous work demonstrated the antimicrobial activity of the fatty acid sapienic acid (C(16:1Δ6)) against P. gingivalis and found that sapienic acid treatment alters both protein and lipid composition from those in controls. In this study, we further examined whole-cell protein differences between sapienic acid-treated bacteria and untreated controls, and we utilized open-source functional association and annotation programs to explore potential mechanisms for the antimicrobial activity of sapienic acid. Our analyses indicated that sapienic acid treatment induces a unique stress response in P. gingivalis resulting in differential expression of proteins involved in a variety of metabolic pathways. This network of differentially regulated proteins was enriched in protein-protein interactions (P = 2.98 × 10(-8)), including six KEGG pathways (P value ranges, 2.30 × 10(-5) to 0.05) and four Gene Ontology (GO) molecular functions (P value ranges, 0.02 to 0.04), with multiple suggestive enriched relationships in KEGG pathways and GO molecular functions. Upregulated metabolic pathways suggest increases in energy production, lipid metabolism, iron acquisition and processing, and respiration. Combined with a suggested preferential metabolism of serine, which is necessary for fatty acid biosynthesis, these data support our previous findings that the site of sapienic acid antimicrobial activity is likely at the bacterial membrane. IMPORTANCE P. gingivalis is an important opportunistic pathogen implicated in periodontitis. Affecting nearly 50% of the population, periodontitis is treatable, but the resulting damage is irreversible and eventually progresses to tooth loss. There is a great need for natural products that can be used to treat and/or prevent the overgrowth of periodontal pathogens and increase oral health. Sapienic acid is endogenous to the oral cavity and is a potent antimicrobial agent, suggesting a potential therapeutic or prophylactic use for this fatty acid. This study examines the effects of sapienic acid treatment on P. gingivalis and highlights the membrane as the likely site of antimicrobial activity.
Collapse
|
37
|
Li S, Hao L, Wang L, Lu Y, Li Q, Zhu Z, Shao JZ, Chen W. Targeting Atp6v1c1 Prevents Inflammation and Bone Erosion Caused by Periodontitis and Reveals Its Critical Function in Osteoimmunology. PLoS One 2015; 10:e0134903. [PMID: 26274612 PMCID: PMC4537256 DOI: 10.1371/journal.pone.0134903] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 07/16/2015] [Indexed: 01/08/2023] Open
Abstract
Periodontal disease (Periodontitis) is a serious disease that affects a majority of adult Americans and is associated with other systemic diseases, including diabetes, rheumatoid arthritis, and other inflammatory diseases. While great efforts have been devoted toward understanding the pathogenesis of periodontitis, there remains a pressing need for developing potent therapeutic strategies for targeting this pervasive and destructive disease. In this study, we utilized novel adeno-associated virus (AAV)-mediated Atp6v1c1 knockdown gene therapy to treat bone erosion and inflammatory caused by periodontitis in mouse model. Atp6v1c1 is a subunit of the V-ATPase complex and regulator of the assembly of the V0 and V1 domains of the V-ATPase complex. We demonstrated previously that Atp6v1c1 has an essential function in osteoclast mediated bone resorption. We hypothesized that Atp6v1c1 may be an ideal target to prevent the bone erosion and inflammation caused by periodontitis. To test the hypothesis, we employed AAV RNAi knockdown of Atp6v1c1 gene expression to prevent bone erosion and gingival inflammation simultaneously. We found that lesion-specific injection of AAV-shRNA-Atp6v1c1 into the periodontal disease lesions protected against bone erosion (>85%) and gingival inflammation caused by P. gingivalis W50 infection. AAV-mediated Atp6v1c1 knockdown dramatically reduced osteoclast numbers and inhibited the infiltration of dendritic cells and macrophages in the bacteria-induced inflammatory lesions in periodontitis. Silencing of Atp6v1c1 expression also prevented the expressions of osteoclast-related genes and pro-inflammatory cytokine genes. Our data suggests that AAV-shRNA-Atp6v1c1 treatment can significantly attenuate the bone erosion and inflammation caused by periodontitis, indicating the dual function of AAV-shRNA-Atp6v1c1 as an inhibitor of bone erosion mediated by osteoclasts, and as an inhibitor of inflammation through down-regulation of pro-inflammatory cytokine expression. This study demonstrated that Atp6v1c1 RNAi knockdown gene therapy mediated by AAV-shRNA-Atp6v1c1 is a promising novel therapeutic approach for the treatment of bone erosion and inflammatory related diseases, such as periodontitis and rheumatoid arthritis.
Collapse
Affiliation(s)
- Sheng Li
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, United States of America
- College of Stomatology, Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Liang Hao
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, United States of America
| | - Lin Wang
- College of Stomatology, Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Yun Lu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, United States of America
| | - Qian Li
- Life Science College, Zhejiang University, 388 Yuhang Road, Hangzhou, 310058, People's Republic of China
| | - Zheng Zhu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, United States of America
- College of Stomatology, Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Jian-Zhong Shao
- Life Science College, Zhejiang University, 388 Yuhang Road, Hangzhou, 310058, People's Republic of China
| | - Wei Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, United States of America
- * E-mail:
| |
Collapse
|
38
|
Poulsen C, Mehalick LA, Fischer CL, Lanzel EA, Bates AM, Walters KS, Cavanaugh JE, Guthmiller JM, Johnson GK, Wertz PW, Brogden KA. Differential cytotoxicity of long-chain bases for human oral gingival epithelial keratinocytes, oral fibroblasts, and dendritic cells. Toxicol Lett 2015; 237:21-9. [PMID: 26005054 DOI: 10.1016/j.toxlet.2015.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/15/2015] [Accepted: 05/17/2015] [Indexed: 01/25/2023]
Abstract
Long-chain bases are present in the oral cavity. Previously we determined that sphingosine, dihydrosphingosine, and phytosphingosine have potent antimicrobial activity against oral pathogens. Here, we determined the cytotoxicities of long-chain bases for oral cells, an important step in considering their potential as antimicrobial agents for oral infections. This information would clearly help in establishing prophylactic or therapeutic doses. To assess this, human oral gingival epithelial (GE) keratinocytes, oral gingival fibroblasts (GF), and dendritic cells (DC) were exposed to 10.0-640.0 μM long-chain bases and glycerol monolaurate (GML). The effects of long-chain bases on cell metabolism (conversion of resazurin to resorufin), membrane permeability (uptake of propidium iodide or SYTOX-Green), release of cellular contents (LDH), and cell morphology (confocal microscopy) were all determined. GE keratinocytes were more resistant to long-chain bases as compared to GF and DC, which were more susceptible. For DC, 0.2-10.0 μM long-chain bases and GML were not cytotoxic; 40.0-80.0 μM long-chain bases, but not GML, were cytotoxic; and 80.0 μM long-chain bases induced cellular damage and death in less than 20 min. The LD50 of long-chain bases for GE keratinocytes, GF, and DC were considerably higher than their minimal inhibitory concentrations for oral pathogens, a finding important to pursuing their future potential in treating periodontal and oral infections.
Collapse
Affiliation(s)
- Christopher Poulsen
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Leslie A Mehalick
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Carol L Fischer
- Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Emily A Lanzel
- Department of Oral Pathology, Radiology and Medicine, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Amber M Bates
- Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Katherine S Walters
- Central Microscopy Research Facility, The University of Iowa, Iowa City, IA 52242, USA
| | - Joseph E Cavanaugh
- Department of Biostatistics, College of Public Health, The University of Iowa, Iowa City, IA 52242, USA
| | - Janet M Guthmiller
- College of Dentistry, University of Nebraska Medical Center, 40th and Holdrege, Lincoln, NE 68583, USA
| | - Georgia K Johnson
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Philip W Wertz
- Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA; Department of Oral Pathology, Radiology and Medicine, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Kim A Brogden
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA; Dows Institute for Dental Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
39
|
Deficiency of cathepsin K prevents inflammation and bone erosion in rheumatoid arthritis and periodontitis and reveals its shared osteoimmune role. FEBS Lett 2015; 589:1331-1339. [PMID: 25896020 DOI: 10.1016/j.febslet.2015.04.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/06/2015] [Accepted: 04/08/2015] [Indexed: 12/22/2022]
Abstract
Using rheumatoid arthritis (RA) and periodontitis mouse models, we demonstrate that RA and periodontitis share many pathological features, such as deregulated cytokine production, increased immune-cell infiltration, increased expression of Toll-like receptors (TLRs), and enhanced osteoclast activity and bone erosion. We reveal that genetic deletion of cathepsin K (Ctsk) caused a radical reduction in inflammation and bone erosion within RA joint capsules and periodontal lesions, a drastic decrease in immune-cell infiltration, and a significant reduction in osteoclasts, macrophages, dendritic and T-cells. Deficiency of Ctsk greatly decreased the expression of TLR-4, 5, and 9 and their downstream cytokines in periodontal gingival epithelial lesions and synovial RA lesions. Hence, Ctsk may be targeted to treat RA and periodontitis simultaneously due to its shared osteoimmune role.
Collapse
|
40
|
Valentijn-Benz M, van ''t Hof W, Bikker FJ, Nazmi K, Brand HS, Sotres J, Lindh L, Arnebrant T, Veerman EC. Sphingoid Bases Inhibit Acid-Induced Demineralization of Hydroxyapatite. Caries Res 2014; 49:9-17. [DOI: 10.1159/000362096] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 03/03/2014] [Indexed: 11/19/2022] Open
Abstract
Calcium hydroxyapatite (HAp), the main constituent of dental enamel, is inherently susceptible to the etching and dissolving action of acids, resulting in tooth decay such as dental caries and dental erosion. Since the prevalence of erosive wear is gradually increasing, there is urgent need for agents that protect the enamel against erosive attacks. In the present study we studied in vitro the anti-erosive effects of a number of sphingolipids and sphingoid bases, which form the backbone of sphingolipids. Pretreatment of HAp discs with sphingosine, phytosphingosine (PHS), PHS phosphate and sphinganine significantly protected these against acid-induced demineralization by 80 ± 17%, 78 ± 17%, 78 ± 7% and 81 ± 8%, respectively (p < 0.001). On the other hand, sphingomyelin, acetyl PHS, octanoyl PHS and stearoyl PHS had no anti-erosive effects. Atomic force measurement revealed that HAp discs treated with PHS were almost completely and homogeneously covered by patches of PHS. This suggests that PHS and other sphingoid bases form layers on the surface of HAp, which act as diffusion barriers against H+ ions. In principle, these anti-erosive properties make PHS and related sphingosines promising and attractive candidates as ingredients in oral care products. © 2014 S. Karger AG, Basel
Collapse
|
41
|
Gentile E, Di Stasio D, Santoro R, Contaldo M, Salerno C, Serpico R, Lucchese A. In vivo microstructural analysis of enamel in permanent and deciduous teeth. Ultrastruct Pathol 2014; 39:131-4. [PMID: 25268300 DOI: 10.3109/01913123.2014.960544] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Confocal microscope was used to analyze human enamel from 10 deciduous and 10 permanent teeth. Optically sectioned images were obtained. A more intense autofluorescence was found in primary teeth. This finding might be due to the greater presence of organic substances in deciduous enamel. The mean prism diameter measurement in permanent teeth enamel was 3.150 µm and 2.602 µm in deciduous teeth. The mean prism diameter in deciduous teeth was statistically least. The results indicate that a confocal microscope may be of help in analyzing and defining the microscopic features of human enamel.
Collapse
Affiliation(s)
- Enrica Gentile
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, Second University of Naples , Naples , Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Lee JT, Escobar OH, Anouseyan R, Janisiewicz A, Eivers E, Blackwell KE, Keschner DB, Garg R, Porter E. Assessment of epithelial innate antimicrobial factors in sinus tissue from patients with and without chronic rhinosinusitis. Int Forum Allergy Rhinol 2014; 4:893-900. [PMID: 25196914 DOI: 10.1002/alr.21404] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/27/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Airway secretions contain endogenous antimicrobial factors (AMFs) that contribute to the innate host defense of the respiratory tract. Antibacterial peptides as well as host-derived lipids including cholesteryl esters have been detected in maxillary lavage fluid. Sterol O-acyltransferase 1 (SOAT1) is a key enzyme in cholesteryl ester production. The purpose of this study is to determine if such intrinsic microbicidal molecules are acutely expressed within sinus tissue and to compare levels of expression between patients with and without chronic rhinosinusitis (CRS). METHODS Sinus tissue was obtained from subjects with (24) and without (9) a history of CRS. Six CRS patients had nasal polyposis (CRSwNP). Immunofluorescence staining for human neutrophil peptide (HNP) was done as a marker for inflammation. Real-time polymerase chain reaction (RT-PCR) following RNA extraction was used to quantify the expression of SOAT-1, the epithelial beta-defensins (HBD2 and HBD3), and the cathelicidin LL37 with ribosomal protein, large, P0 (RPLP0) as the housekeeping gene. RESULTS Immunofluorescence showed significant increase in HNP staining in CRS patients without nasal polyposis (CRSsNP) vs non-CRS specimens (p = 0.010), in agreement with clinical inflammation status. SOAT1 messenger RNA (mRNA) expression was also upregulated in CRSsNP compared to non-CRS (p = 0.041) and CRSwNP (p = 0.005) patients, whereas increases for HBD2 and HBD3 were less prominent. LL37 was either absent or expressed at very low levels in all samples. CONCLUSION Increased biosynthesis of SOAT1, a key enzyme for antimicrobial cholesteryl ester production, was observed in the sinus tissue of CRSsNP patients but not in CRSwNP patients. This further supports the novel concept of lipid-mediated innate mucosal defense and delineates CRS with and without nasal polyposis as distinct subtypes.
Collapse
Affiliation(s)
- Jivianne T Lee
- Orange County Sinus Institute, Southern California Permanente Medical Group (SCPMG), Irvine, CA; Department of Otolaryngology-Head and Neck Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Liu R, Li J, Zhang T, Zou L, Chen Y, Wang K, Lei Y, Yuan K, Li Y, Lan J, Cheng L, Xie N, Xiang R, Nice EC, Huang C, Wei Y. Itraconazole suppresses the growth of glioblastoma through induction of autophagy: involvement of abnormal cholesterol trafficking. Autophagy 2014; 10:1241-55. [PMID: 24905460 DOI: 10.4161/auto.28912] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma is one of the most aggressive human cancers with poor prognosis, and therefore a critical need exists for novel therapeutic strategies for management of glioblastoma patients. Itraconazole, a traditional antifungal drug, has been identified as a novel potential anticancer agent due to its inhibitory effects on cell proliferation and tumor angiogenesis; however, the molecular mechanisms involved are still unclear. Here, we show that itraconazole inhibits the proliferation of glioblastoma cells both in vitro and in vivo. Notably, we demonstrate that treatment with itraconazole induces autophagic progression in glioblastoma cells, while blockage of autophagy markedly reverses the antiproliferative activities of itraconazole, suggesting an antitumor effect of autophagy in response to itraconazole treatment. Functional studies revealed that itraconazole retarded the trafficking of cholesterol from late endosomes and lysosomes to the plasma membrane by reducing the levels of SCP2, resulting in repression of AKT1-MTOR signaling, induction of autophagy, and finally inhibition of cell proliferation. Together, our studies provide new insights into the molecular mechanisms regarding the antitumor activities of itraconazole, and may further assist both the pharmacological investigation and rational use of itraconazole in potential clinical applications.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu, China
| | - Jingyi Li
- School of Biomedical Sciences; Chengdu Medical College; Chengdu, China
| | - Tao Zhang
- School of Biomedical Sciences; Chengdu Medical College; Chengdu, China
| | - Linzhi Zou
- College of Life Sciences; Sichuan University; Chengdu, China
| | - Yi Chen
- Department of Gastrointestinal Surgery; State Key Laboratory of Biotherapy; West China Hospital, Sichuan University, Chengdu, China
| | - Kui Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center; Chongqing Medical University; Chongqing, China
| | - Kefei Yuan
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| | - Yi Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| | - Jiang Lan
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| | - Na Xie
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| | - Rong Xiang
- School of Medicine/Collaborative Innovation Center of Biotherapy; Nankai University; Tianjin, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology; Monash University; Clayton, Victoria Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; Sichuan University; Chengdu, China
| |
Collapse
|
44
|
Li Y, He J, He Z, Zhou Y, Yuan M, Xu X, Sun F, Liu C, Li J, Xie W, Deng Y, Qin Y, VanNostrand JD, Xiao L, Wu L, Zhou J, Shi W, Zhou X. Phylogenetic and functional gene structure shifts of the oral microbiomes in periodontitis patients. ISME JOURNAL 2014; 8:1879-91. [PMID: 24671083 DOI: 10.1038/ismej.2014.28] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 12/09/2013] [Accepted: 01/25/2014] [Indexed: 02/05/2023]
Abstract
Determining the composition and function of subgingival dental plaque is crucial to understanding human periodontal health and disease, but it is challenging because of the complexity of the interactions between human microbiomes and human body. Here, we examined the phylogenetic and functional gene differences between periodontal and healthy individuals using MiSeq sequencing of 16S rRNA gene amplicons and a specific functional gene array (a combination of GeoChip 4.0 for biogeochemical processes and HuMiChip 1.0 for human microbiomes). Our analyses indicated that the phylogenetic and functional gene structure of the oral microbiomes were distinctly different between periodontal and healthy groups. Also, 16S rRNA gene sequencing analysis indicated that 39 genera were significantly different between healthy and periodontitis groups, and Fusobacterium, Porphyromonas, Treponema, Filifactor, Eubacterium, Tannerella, Hallella, Parvimonas, Peptostreptococcus and Catonella showed higher relative abundances in the periodontitis group. In addition, functional gene array data showed that a lower gene number but higher signal intensity of major genes existed in periodontitis, and a variety of genes involved in virulence factors, amino acid metabolism and glycosaminoglycan and pyrimidine degradation were enriched in periodontitis, suggesting their potential importance in periodontal pathogenesis. However, the genes involved in amino acid synthesis and pyrimidine synthesis exhibited a significantly lower relative abundance compared with healthy group. Overall, this study provides new insights into our understanding of phylogenetic and functional gene structure of subgingival microbial communities of periodontal patients and their importance in pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinzhi He
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhili He
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Yuan Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengting Yuan
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Xin Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Feifei Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenbo Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ye Deng
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Yujia Qin
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Joy D VanNostrand
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Liying Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liyou Wu
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Jizhong Zhou
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Wenyuan Shi
- 1] State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China [2] UCLA School of Dentistry, Los Angeles, CA, USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|