Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
JDR Clin Trans Res ; 9(1): 72-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-36680313

ABSTRACT

BACKGROUND: With addiction rates and opioid deaths increasing, health care providers are obligated to help stem the opioid crisis. As limited studies examine the comparative effectiveness of fixed-dose combination nonopioid analgesia to opioid-containing analgesia, a comparative effectiveness study was planned and refined by conducting a pilot study. METHODS: The Opioid Analgesic Reduction Study (OARS) pilot, a stratified, randomized, multisite, double-blind clinical trial, was designed to test technology and procedures to be used in the full OARS trial. Participants engaged in the full protocol, enabling the collection of OARS outcome data. Eligible participants reporting to 1 of 5 sites for partial or full bony impacted mandibular third molar extraction were stratified by biologic sex and randomized to 1 of 2 treatment groups, OPIOID or NONOPIOID. OPIOID participants were provided 20 doses of hydrocodone 5 mg/acetaminophen 300 mg. NONOPIOID participants were provided 20 doses of ibuprofen 400 mg/acetaminophen 500 mg. OARS outcomes data, including pain experience, adverse effects, sleep quality, pain interference, overall satisfaction, and remaining opioid tablets available for diversion, were collected via surveys, electronic medication bottles, eDiary, and activity/sleep monitor. RESULTS: Fifty-three participants were randomized with 50 completing the OARS pilot protocol. Across all outcome pain domains, in all but 1 time period, NONOPIOID was better in managing pain than OPIOID (P < 0.05 level). Other outcomes suggest less pain interference, less adverse events, better sleep quality, better overall satisfaction, and fewer opioid-containing tablets available for diversion. DISCUSSION: Results suggest patients requiring impacted mandibular third molar extraction would benefit from fixed-dose combination nonopioid analgesia. KNOWLEDGE TRANSFER STATEMENT: Study results suggest fixed-dose nonopioid combination ibuprofen 400 mg/acetaminophen 500 mg is superior to opioid-containing analgesic (hydrocodone 5 mg/acetaminophen 500 mg). This knowledge should inform surgeons and patients in the selection of postsurgical analgesia.


Subject(s)
Analgesics, Non-Narcotic , Analgesics, Opioid , Humans , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/adverse effects , Acetaminophen/therapeutic use , Acetaminophen/adverse effects , Ibuprofen/therapeutic use , Ibuprofen/adverse effects , Hydrocodone/adverse effects , Pilot Projects , Drug Combinations , Analgesics, Non-Narcotic/therapeutic use , Analgesics, Non-Narcotic/adverse effects , Pain/chemically induced , Pain/drug therapy , Double-Blind Method
2.
JDR Clin Trans Res ; 8(2): 188-197, 2023 04.
Article in English | MEDLINE | ID: mdl-35191352

ABSTRACT

OBJECTIVES: To estimate the association between safety perception on vaccine acceptance and adoptions of risk mitigation strategies among dental health care workers (DHCWs). METHODS: A survey was emailed to DHCWs in the New Jersey area from December 2020 to January 2021. Perceived safety from regular SARS-CoV-2 testing of self, coworkers, and patients and its association with vaccine hesitancy and risk mitigation were ascertained. Risk Mitigation Strategy (RiMS) scores were computed from groupings of office measures: 1) physical distancing (reduced occupancy, traffic flow, donning of masks, minimal room crowding), 2) personal protective equipment (fitted for N95; donning N95 masks; use of face shields; coverings for head, body, and feet), and 3) environmental disinfection (suction, air filtration, ultraviolet, surface wiping). RESULTS: SARS-CoV-2 testing of dental professionals, coworkers, and patients were perceived to provide safety at 49%, 55%, and 68%, respectively. While dentists were least likely to feel safe with regular self-testing for SARS-CoV-2 (P < 0.001) as compared with hygienists and assistants, they were more willing than hygienists (P = 0.004; odds ratio, 1.79 [95% CI, 1.21 to 2.66]) and assistants (P < 0.001; odds ratio, 3.32 [95% CI, 1.93 to 5.71]) to receive the vaccine. RiMS scores ranged from 0 to 19 for 467 participants (mean [SD], 10.9 [2.9]). RiMS scores did not significantly differ among groups of DHCWs; however, mean RiMS scores were higher among those who received or planned to receive the COVID-19 vaccine than those with who did not (P = 0.004). DHCWs who felt safer with regular testing had greater RiMS scores than those who did not (11.0 vs. 10.3, P = 0.01). CONCLUSIONS: Understanding DHCWs' perception of risk and safety is crucial, as it likely influences attitudes toward testing and implementation of office risk mitigation policies. Clinical studies that correlate risk perception and RiMS with SARS-CoV-2 testing are needed to demonstrate the effectiveness of RiMS in dental care settings. KNOWLEDGE TRANSFER STATEMENT: Educators, clinicians, and policy makers can use the results of this study when improving attitudes toward testing and implementation of risk mitigation policies within dental offices, for current and future pandemics.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines , COVID-19 Testing , Delivery of Health Care , Perception
3.
Oral Dis ; 25(3): 652-669, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29656422

ABSTRACT

The oral cavity harbors different taxonomic groups, the evolutionary coexistence of which develops the oral ecosystem. These resident microorganisms can alter the balance between the physiologic and pathologic conditions that affect the host, both locally and systemically. This highly sophisticated nature of the oral cavity poses a significant therapeutic challenge. Numerous human and animal studies have been conducted to potentiate the efficacy and competence of current treatments of pathologic conditions as well as to develop novel therapeutic modalities. One of these studies is the use of the potent antimicrobial agent lactoferrin (LF), which was originally derived from the host immune system. LF is an 80-kDa glycoprotein that has a free iron sequestration mechanism with evident antimicrobial, anti-tumor, and immunomodulatory properties. A wide range of active peptides have been isolated from the N-terminal region of LF, which possess antimicrobial activities. In this review, we discuss the role of LF and LF-derived peptides under a heterogeneous group of oral and maxillofacial conditions, including bacterial, fungal, viral infections; head and neck cancers; xerostomia; and implantology-bone-related manifestations.


Subject(s)
Bacteria/drug effects , Lactoferrin/pharmacology , Mouth Neoplasms/prevention & control , Peptides/pharmacology , Periodontal Diseases/microbiology , Animals , Candida albicans/drug effects , Carcinogenesis/drug effects , Dental Caries/microbiology , Dental Caries/prevention & control , Humans , Lactoferrin/genetics , Lactoferrin/therapeutic use , Peptides/therapeutic use , Polymorphism, Single Nucleotide , Streptococcus mutans/drug effects , Virus Physiological Phenomena/drug effects
5.
Oral Dis ; 22(2): 148-54, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26601919

ABSTRACT

OBJECTIVES: The objective of this study is to evaluate the importance of human lactoferrin (hLF) in an experimental caries induced by Streptococcus mutans in a lactoferrin-knockout (LFKO(-/-)) mouse model compared with C576J/BL wild-type (WT) mice. MATERIALS AND METHODS: The WT and LFKO(-/-) mice were infected with S. mutans (1 × 10(8) cells) and/or sham infection. Furthermore, the effect of hLF administration was evaluated in LFKO(-/-) mice infected with S. mutans. Mice were assessed for colonization, salivary pH, and caries development. RESULTS: The results showed that the lactoferrin-knockout infected (LFKO(-/-) I) mice had significantly higher colonization with S. mutans (P = 0.02), lower salivary pH (P = 0.01), and more carious lesions (P = 0.01) when compared to wild-type infected (WTI) mice. In addition, the administration of hLF did not show any evidence of S. mutans colonization as well as carious lesions (P = 0.001) in LFKO(-/-) I mice when compared to untreated LFKO(-/-) I mice. CONCLUSION: These results show that endogenous LF protects against S. mutans-induced caries and that exogenous hLF can exert a protective effect against caries development.


Subject(s)
Anti-Infective Agents/therapeutic use , Dental Caries/microbiology , Dental Caries/prevention & control , Lactoferrin/therapeutic use , Streptococcal Infections/prevention & control , Streptococcus mutans , Animals , Humans , Hydrogen-Ion Concentration , Lactoferrin/genetics , Male , Mice , Mice, Knockout , Saliva/chemistry
6.
Mol Oral Microbiol ; 31(4): 329-39, 2016 08.
Article in English | MEDLINE | ID: mdl-26280561

ABSTRACT

Among the various proteins expressed by the periodontopathogen Aggregatibacter actinomycetemcomitans, two proteins play important roles for survival in the oral cavity. The autotransporter Aae facilitates the attachment of the pathogen to oral epithelial cells, which act as a reservoir, while the biofilm-degrading glycoside hydrolase dispersin B facilitates the movement of daughter cells from the mature biofilm to a new site. The objective of this study was to use the potential of these two proteins to control biofilms. To this end, we generated a hybrid construct between the Aae C-terminal translocating domain and dispersin B, and mobilized it into Escherichia coli Rosetta (DE3) pLysS cells. Immunofluorescence analysis of the modified E. coli cells confirmed the presence of dispersin B on the surface. Further, the membrane localization of the displayed dispersin B was confirmed with Western blot analysis. The integrity of the E. coli cells displaying the dispersin B was confirmed through FACS analysis. The hydrolytic activity of the surface-displayed dispersin B was confirmed by using 4-methylumbelliferyl-ß-d-glucopyranoside as the substrate. The detachment ability of the dispersin B surface-displaying E. coli cells was shown using Staphylococcus epidermidis and Actinobacillus pleuropneumoniae biofilms in a microtiter assay. We concluded that the Aae ß-domain is sufficient to translocate foreign enzymes in the native folded form and that the method of Aae-mediated translocation of surface displayed enzymes might be useful for control of biofilms.


Subject(s)
Aggregatibacter actinomycetemcomitans/physiology , Bacterial Adhesion , Bacterial Proteins/metabolism , Biofilms/growth & development , Escherichia coli/genetics , Glycoside Hydrolases/metabolism , Type V Secretion Systems/metabolism , Actinobacillus pleuropneumoniae/physiology , Aggregatibacter actinomycetemcomitans/enzymology , Aggregatibacter actinomycetemcomitans/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Surface Display Techniques , Escherichia coli/chemistry , Escherichia coli/metabolism , Flow Cytometry , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Staphylococcus epidermidis/physiology , Type V Secretion Systems/chemistry , Type V Secretion Systems/genetics
7.
J Dent Res ; 94(6): 768-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25784250

ABSTRACT

Lactoferrin is one of a number of multifunctional proteins that are present in or on all mucosal surfaces throughout the body. Levels of lactoferrin are consistently elevated in inflammatory diseases such as arthritis, inflammatory bowel diseases, corneal disease, and periodontitis. Single-nucleotide polymorphisms (SNPs) in lactoferrin have been shown to be present in individuals susceptible to Escherichia coli-induced travelers' diarrhea and in tear fluid derived from virally associated corneal disease. Here, we review data showing a lactoferrin SNP in amino acid position 29 in the antimicrobial region of lactoferrin that acts against caries associated bacteria. This SNP was initially discovered in African American subjects with localized aggressive periodontitis (LAP) who had proximal bone loss but minimal proximal caries. Results were confirmed in a genetic association study of children from Brazil with this same SNP who showed a reduced level of caries. In vitro data indicate that lactoferrin from whole saliva derived from subjects with this SNP, recombinant human lactoferrin containing this SNP, or an 11-mer peptide designed for this SNP kills mutans streptococci associated with caries by >1 log. In contrast, the SNP has minimal effect on Gram-negative species associated with periodontitis. Moreover, periodontally healthy subjects homozygous for this lysine (K) SNP have lactoferrin in their saliva that kills mutans streptococci and have reduced proximal decay. The review summarizes data supporting the ecologic plaque hypothesis and suggests that a genetic variant in lactoferrin with K in position 29 when found in saliva and crevice fluid can influence community biofilm composition. We propose that, for caries, this SNP is ethnicity independent and protective by directly killing caries-provoking bacteria (reducing proximal decay). However, the clinical effect of this SNP in LAP is ethnicity dependent, destructive (increases LAP incidence), and complex with mechanisms still to be determined.


Subject(s)
Aggressive Periodontitis/genetics , Dental Caries/genetics , Lactoferrin/genetics , Anti-Infective Agents/pharmacology , Biofilms/drug effects , Dental Caries/microbiology , Dental Plaque/microbiology , Humans , Lactoferrin/physiology , Lysine/genetics , Polymorphism, Single Nucleotide/genetics , Streptococcus mutans/drug effects
8.
J Appl Microbiol ; 118(1): 212-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25319508

ABSTRACT

AIMS: To determine the role of human lactoferrin (hLF) in protecting the oral cavities of mice against Candida albicans infection in lactoferrin knockout (LFKO(-/-)) mice was compared to wild-type (WT) mice. We also aim to determine the protective role of hLF in LFKO(-/-) mice. METHODS AND RESULTS: Antibiotic-treated immunosuppressed mice were inoculated with C. albicans (or sham infection) by oral swab and evaluated for the severity of infection after 7 days of infection. To determine the protective role of hLF, we added 0·3% solution of hLF to the drinking water given to some of the mice. CFU count, scoring of lesions and microscopic observations were carried out to determine the severity of infection. LFKO(-/-) I mice showed a 2 log (P = 0·001) higher CFUs of C. albicans in the oral cavity compared to the WT mice infected with C. albicans (WTI). LFKO(-/-) I mice given hLF had a 3 log (P = 0·001) reduction in CFUs in the oral cavity compared to untreated LFKO(-/-) I mice. The severity of infection, observed by light microscopy, revealed that the tongue of the LFKO(-/-) I mice showed more white patches compared to WTI and LFKO(-/-) I + hLF mice. Scanning electron microscopic observations revealed that more filiform papillae were destroyed in LFKO(-/-) I mice when compared to WTI or LFKO(-/-) I + hLF mice. CONCLUSIONS: Human LF is important in protecting mice from oral C. albicans infection. Administered hLF may be used to prevent C. albicans infection. SIGNIFICANCE AND IMPACT OF THE STUDY: Human LF, a multifunctional iron-binding glycoprotein can be used as a therapeutic active ingredient in oral healthcare products against C. albicans.


Subject(s)
Candidiasis/prevention & control , Lactoferrin/therapeutic use , Mouth Diseases/prevention & control , Administration, Oral , Animals , Anti-Bacterial Agents/therapeutic use , Candida albicans/drug effects , Candida albicans/growth & development , Candidiasis/microbiology , Candidiasis/pathology , Humans , Lactoferrin/administration & dosage , Lactoferrin/genetics , Male , Mice , Mice, Knockout , Mouth Diseases/microbiology , Mouth Diseases/pathology , Tongue/pathology
9.
Antimicrob Agents Chemother ; 58(1): 397-404, 2014.
Article in English | MEDLINE | ID: mdl-24189260

ABSTRACT

Aggregatibacter actinomycetemcomitans, a periodontopathogen, has been associated with several systemic diseases. Herein, we report the protective effect of human lactoferrin (hLF) during A. actinomycetemcomitans bacteremia in lactoferrin knockout (LFKO(-/-)) mice. The prophylactic, concurrent, and therapeutic intravenous (i.v.) administrations of hLF significantly cleared the bacteria from blood and organs. Nevertheless, all modes of hLF administration significantly decreased the concentrations of serum proinflammatory cytokines, such as interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6, IL-10, and IL-12p70. Additionally, hLF administration significantly decreased hepatic and splenic proinflammatory cytokine expression levels compared to those in the non-hLF-treated group. Furthermore, administration of hLF decreased the serum C-reactive protein level, inducible nitric oxide synthase (iNOS) and myeloperoxidase (MPO) gene expression levels in liver and spleen. hLF treatment has also resulted in a 6-fold decrease in spleen weight with the migration of typical inflammatory cells in infected mice as a result of decreased inflammatory response. These results reveal that hLF protects against A. actinomycetemcomitans bacteremia, as indicated by rapid bacterial clearance and decreased host proinflammatory mediators.


Subject(s)
Aggregatibacter actinomycetemcomitans/pathogenicity , Bacteremia/drug therapy , Bacteremia/microbiology , Lactoferrin/therapeutic use , Aged, 80 and over , Animals , C-Reactive Protein/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lactoferrin/deficiency , Lactoferrin/genetics , Lactoferrin/metabolism , Male , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
J Periodontol ; 84(11): 1690-701, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23327622

ABSTRACT

BACKGROUND: Among the innate defense mechanisms in the oral cavity, lactoferrin (LF) is a vital antimicrobial that can modify the host response against periodontopathogens. Aggregatibacter actinomycetemcomitans is the main periodontopathogen of localized aggressive periodontitis. The aim of this study is to evaluate the role of LF during A. actinomycetemcomitans-induced periodontitis. METHODS: Differences in the expression levels of cytokines, chemokines, chemokine receptors, and bone loss markers between wild-type (WT) and LF knockout mice (LFKO(-/-)) were evaluated by real time-PCR. Serum IgG and LF levels were quantified by ELISA. Alveolar bone loss among the groups was estimated by measuring the distance from cemento-enamel junction (CEJ) to the alveolar bone crest (ABC) at 20 molar sites. RESULTS: Oral infection with A. actinomycetemcomitans increased LF levels in periodontal tissue (P = 0.01) and saliva (P = 0.0004) of wild-type infected (WTI) mice compared to wild-type control mice. Pro-inflammatory cytokines such as interferon-γ, tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, and IL-12 were increased in the infected LF knockout (LFKO(-/-)I) mice compared to the WTI mice, whereas the anti-inflammatory cytokines IL-4 and IL-10 were decreased. Chemokines and chemokine receptors showed different expression patterns between WTI and LFKO(-/-)I mice. The LFKO(-/-)I mice developed increased bone loss (P = 0.002), in conjunction with increased expression of receptor activator of nuclear factor-κB ligand and decrease in osteoprotegerin, compared to WTI mice. CONCLUSIONS: These results demonstrate that the infected LFKO(-/-) mice were more susceptible to A. actinomycetemcomitans-induced alveolar bone loss, with different patterns of immune responses compared to those of WTI mice.


Subject(s)
Aggregatibacter actinomycetemcomitans/immunology , Aggressive Periodontitis/microbiology , Disease Susceptibility/immunology , Lactoferrin/immunology , Aggressive Periodontitis/immunology , Alveolar Bone Loss/immunology , Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , Animals , Chemokines/immunology , Cytokines/immunology , Immunoglobulin G/blood , Interferon-gamma/analysis , Interleukin-10/analysis , Interleukin-12/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , Interleukins/analysis , Lactoferrin/blood , Lactoferrin/genetics , Mice , Mice, Knockout , Osteoprotegerin/analysis , Pasteurellaceae Infections/immunology , Pasteurellaceae Infections/microbiology , Periodontium/immunology , Periodontium/microbiology , RANK Ligand/analysis , Receptors, Chemokine/immunology , Saliva/chemistry , Tooth Cervix/pathology , Tumor Necrosis Factor-alpha/analysis , Vesicular Transport Proteins
11.
J Periodontal Res ; 47(5): 661-73, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22582873

ABSTRACT

BACKGROUND AND OBJECTIVE: Healthy subjects who do not have Aggregatibacter actinomycetemcomitans in their oral cavity may possess factors in saliva that might demonstrate antibacterial activity against the bacterium. The aim of this study was to identify and purify proteins from saliva of healthy subjects that might demonstrate antibacterial activity against A. actinomycetemcomitans and test the same against the bacteria. MATERIAL AND METHODS: Saliva from 10 healthy volunteers was tested individually for its anti-A. actinomycetemcomitans activity. Among the 10 subjects, eight demonstrated anti-A. actinomycetemcomitans activity. Saliva was collected from one healthy volunteer who demonstrated the highest antimicrobial activity against A. actinomycetemcomitans. After clarifying the saliva, it was subjected to an affinity chromatography column with A. actinomycetemcomitans. The proteins bound to A. actinomycetemcomitans were eluted from the column and identified using mass spectrometry (MALDI-TOF/TOF MS). Among other proteins that bound to A. actinomycetemcomitans, which included lactoferrin, immunoglobulin A and kallikrein, cystatin SA was observed in significantly higher concentrations, and this was purified from the eluate. The purified cystatin SA was tested at different concentrations for its ability to kill A. actinomycetemcomitans in a 2 h cell killing assay. The bacteria were also treated with a proteinase inhibitor, leupeptin, to clarify whether the antimicrobial effect of cystatin SA was related to its protease inhibitory function. Cystatin SA was also tested for its ability to prevent binding of A. actinomycetemcomitans to buccal epithelial cells (BECs) in an A. actinomycetemcomitans-BEC binding assay. RESULTS: Cystatin SA (0.1 mg/mL) demonstrated a statistically significant antimicrobial activity against A. actinomycetemcomitans. The effect of cystatin SA decreased with lower concentrations, with 0.01 mg/mL showing no effect. The addition of monoclonal cystatin SA antibodies to the purified sample completely negated the antimicrobial effect. Treatment of A. actinomycetemcomitans with leupeptin resulted in no antimicrobial effect, suggesting that the antimicrobial activity of cystatin SA is independent of its protease inhibitory function. A. actinomycetemcomitans pretreated with cystatin SA showed reduced binding to BECs, suggesting a potential role for cystatin SA in decreasing the colonization of A. actinomycetemcomitans. CONCLUSION: The present study shows that cystatin SA demonstrates antimicrobial activity against the periodontopathogen A. actinomycetemcomitans, and future studies determining the mechanism of action are necessary. The study also shows the ability of cystatin SA to reduce significantly the binding of A. actinomycetemcomitans to BECs.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Salivary Cystatins/pharmacology , Bacterial Adhesion/drug effects , Cathepsins/antagonists & inhibitors , Chromatography, Affinity , Cysteine Proteinase Inhibitors/isolation & purification , Electrophoresis, Polyacrylamide Gel , Endopeptidase K/pharmacology , Epithelial Cells/microbiology , Fusobacterium nucleatum/drug effects , Humans , Immunoglobulin A, Secretory/isolation & purification , Kallikreins/isolation & purification , Lactoferrin/isolation & purification , Leupeptins/pharmacology , Microscopy, Confocal , Mouth Mucosa/cytology , Mouth Mucosa/microbiology , Porphyromonas gingivalis/drug effects , Saliva/drug effects , Salivary Cystatins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors
12.
J Microbiol Methods ; 89(1): 71-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22326236

ABSTRACT

Aggregatibacter actinomycetemcomitans is a Gram negative oral bacterium associated with localized aggressive periodontitis (LAP). Detection of A. actinomycetemcomitans in clinical samples is routinely done by PCR. Our aim was to develop a rapid and reliable PCR method that can be used as a chair-side tool to detect A. actinomycetemcomitans in clinical samples. Sensitivity and specificity assessment was performed on buccal and plaque samples obtained from 40 adolescents enrolled in an ongoing LAP study by comparing 20 A. actinomycetemcomitans-positive subjects and 20 who were negative. In a second study, A. actinomycetemcomitans presence was tested in oral samples from eighty-six primates that included rhesus monkeys, chimpanzees, marmosets, tamarins and baboons. All samples were processed for detection of A. actinomycetemcomitans by means of culture, conventional PCR (cPCR) and rapid PCR (rPCR) using a Super Convection based AmpXpress thermal cycler (AlphaHelix, Sweden). For human samples, culture, cPCR and rPCR showed perfect agreement. Using this method A. actinomycetemcomitans was detected in 27 of 32 rhesus monkeys, 4 of 8 chimpanzees and 1 of 34 marmosets. Rapidity of AmpXpress thermal cycler, combined with Ready-To-Go PCR beads (GE Life sciences), a quick DNA extraction kit (Epicentre Biotechnologies, Madison, Wisconsin, USA) and a bufferless fast agarose gel system, made it possible to obtain results on A. actinomycetemcomitans detection within 35 min. We conclude that AmpXpress fast PCR can be conveniently used as a chair-side tool for rapid detection of A. actinomycetemcomitans in clinical samples.


Subject(s)
Bacteriological Techniques/methods , Mouth/microbiology , Pasteurellaceae Infections/microbiology , Pasteurellaceae Infections/veterinary , Pasteurellaceae/isolation & purification , Polymerase Chain Reaction/methods , Adolescent , Animals , Female , Human Experimentation , Humans , Male , Point-of-Care Systems , Primates , Sensitivity and Specificity , Time Factors
13.
Mol Oral Microbiol ; 25(4): 275-92, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20618701

ABSTRACT

There is mounting evidence that innate and adaptive immunity are critical for periodontal disease-mediated bone resorption. These studies examined the role of B and CD4 T cells in adaptive immunity of rats infected with Aggregatibacter actinomycetemcomitans (Aa). Sprague-Dawley male rats were fed Aa-containing mash or control-mash for 2 weeks. B and CD4 T cells were obtained from draining lymph nodes at 2, 4 and 12 weeks, postinoculation. Quantitative polymerase chain reaction-based messenger RNA expression was conducted for 89 cytokine family genes. Disease-relevance of the differentially expressed genes was assessed using a biological interaction pathway analysis software. B and CD4 T cells of Aa-infected rats increased and were activated, resulting in enhanced isotype-switched serum immunoglobulin G by 2 weeks postinoculation. Bone resorption was evident 12 weeks after Aa-feeding. In B cells, interleukin-2 (IL-2), macrophage-inhibiting factor, IL-19, IL-21, tumor necrosis factor (TNF), CD40 ligand (CD40L), CD70, bone morphogenetic protein 2 (BMP2), BMP3, and BMP10 were upregulated early; while IL-7, Fas ligand (FasL), small inducible cytokine subfamily E1, and growth differentiation factor 11 (GDF11; BMP11) were upregulated late (12 weeks). BMP10 was sustained throughout. In CD4 T cells, IL-10, IL-16, TNF, lymphotoxin-beta (LTbeta), APRIL, CD40L, FasL, RANKL and osteoprotegerin were upregulated early, whereas IL-1beta, IL-1RN, IL-1F8, IL-24, interferon-alpha1, GDF11 (BMP11), and GDF15 were upregulated late (12 weeks). Adaptive immunity appears crucial for bone resorption. Several of the deregulated genes are, for the first time, shown to be associated with bone resorption, and the results indicate that activated B cells produce BMP10. The study provides a rationale for a link between periodontal disease and other systemic diseases.


Subject(s)
Adaptive Immunity/genetics , Aggregatibacter actinomycetemcomitans/physiology , Alveolar Bone Loss/immunology , Alveolar Bone Loss/microbiology , CD4-Positive T-Lymphocytes/metabolism , Alveolar Bone Loss/genetics , Animals , Antibodies, Bacterial/biosynthesis , B-Lymphocytes/metabolism , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Gene Expression Profiling , Growth Differentiation Factors/biosynthesis , Growth Differentiation Factors/genetics , Lymphocyte Activation , Male , Osteoclasts/immunology , Rats , Rats, Sprague-Dawley
14.
Oral Microbiol Immunol ; 23(2): 127-30, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18279180

ABSTRACT

BACKGROUND/AIMS: Intergeneric bacterial coaggregation may play an important role in plaque development. METHODS: In this study we investigated the coaggregation reaction between two periodontal pathogens, Aggregatibacter actinomycetemcomitans and Fusobacterium nucleatum. RESULTS: Previous studies showed that A. actinomycetemcomitans serotype b strains coaggregate with F. nucleatum strain PK1594, and that A. actinomycetemcomitans serotype b O-polysaccharide (O-PS) is the receptor responsible for coaggregation between A. actinomycetemcomitans and F. nucleatum. A. actinomycetemcomitans serotype f O-PS has been shown to be structurally and antigenically related to serotype b O-PS. In the present study we show that A. actinomycetemcomitans strain CU1060N, a serotype f strain, also coaggregated with F. nucleatum PK1594. Like coaggregation between serotype b strains and F. nucleatum, coaggregation between CU1060N and F. nucleatum was inhibited by galactose. An O-PS mutant of CU1060N failed to coaggregate with F. nucleatum. CONCLUSION: We concluded that A. actinomycetemcomitans serotype f O-PS, like serotype b O-PS, mediates coaggregation between A. actinomycetemcomitans and fusobacteria.


Subject(s)
Aggregatibacter actinomycetemcomitans/physiology , Bacterial Adhesion/physiology , Dental Plaque/microbiology , Fusobacterium nucleatum/physiology , Polysaccharides, Bacterial/physiology , Biofilms/growth & development , Quorum Sensing/physiology , Serotyping , Species Specificity
15.
J Bacteriol ; 188(17): 6361-75, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16923904

ABSTRACT

The tad locus of Actinobacillus actinomycetemcomitans encodes a molecular transport system required for tenacious, nonspecific adherence to surfaces and formation of extremely strong biofilms. This locus is dedicated to the biogenesis of Flp pili, which are required for colonization and virulence. We have previously shown that 11 of the 14 tad locus genes are required for adherence and Flp pilus production. Here, we present genetic and phylogenetic analyses of flp-2, tadV, and rcpB genes in biofilm formation. We show that tadV, predicted to encode prepilin peptidase, is required for adherence. In contrast, targeted insertional inactivation of flp-2, a gene closely related to the prepillin gene flp-1, did not abrogate biofilm formation. Expression studies did not detect Flp2-T7 protein under standard laboratory conditions. We present phylogenetic data showing that there is no significant evidence for natural selection in the available flp-2 sequences from A. actinomycetemcomitans, suggesting that flp-2 does not play a significant role in the biology of this organism. Mutants with insertions at the 3' end of rcpB formed biofilms equivalent to wild-type A. actinomycetemcomitans. Surprisingly, 5' end chromosomal insertion mutants in rcpB were obtained only when a wild-type copy of the rcpB gene was provided in trans or when the Tad secretion system was inactivated. Together, our results strongly suggest that A. actinomycetemcomitans rcpB is essential in the context of a functional tad locus. These data show three different phenotypes for the three genes.


Subject(s)
Aggregatibacter actinomycetemcomitans/physiology , Bacterial Outer Membrane Proteins/physiology , Bacterial Proteins/physiology , Biofilms/growth & development , Endopeptidases/physiology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Endopeptidases/genetics , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Genes, Bacterial , Molecular Sequence Data , Mutagenesis , Phenotype
16.
Genes Immun ; 6(7): 632-5, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16208406

ABSTRACT

Lactoferrin is an antimicrobial protein which plays an important role in regulating bacteria that are associated with aggressive periodontitis. Lactoferrin kills directly (via its strongly cationic N-terminal region) and indirectly, through sequestering the iron that bacteria require for growth. As aggressive periodontitis has a strong heritable component, we hypothesized that genetic variation within the lactoferrin gene may play a role in susceptibility to this condition. We have identified and examined a novel, functional, single-point A/G nucleotide mutation causing a threonine/alanine substitution at position 11 (T11A) of the secreted lactoferrin protein. In a pilot case-controlled study of aggressive periodontitis, analysis of 46 African-American patients and 78 controls showed that patients were twice as likely to express the G nucleotide (alanine) allele over controls (60.3 vs 30.4%; P=0.0007, odds ratio=2.564, 95% CI=1.475-4.459). A Caucasian population of 77 patients and 131 controls showed no such association (P=0.5201, odds ratio=0.862, 95% CI=0.548-1.356). The data presented provide a new insight into the genetic susceptibility to aggressive periodontitis.


Subject(s)
Black or African American/genetics , Lactoferrin/genetics , Periodontitis/genetics , Polymorphism, Single Nucleotide , Alanine/genetics , Amino Acid Substitution , Case-Control Studies , Conserved Sequence , Female , Humans , Male , Periodontitis/ethnology , Point Mutation , Threonine/genetics , White People
17.
J Clin Periodontol ; 32(4): 335-40, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15811048

ABSTRACT

OBJECTIVES: Two studies were conducted to determine the antimicrobial effect of rinsing with an essential oil-containing mouth rinse 12 h after a single rinse and 12 h after 2 weeks of twice daily rinsing, during the daytime and overnight. MATERIALS AND METHODS: These studies utilized a randomized, double-blind, controlled crossover design. Following baseline sampling of bacteria from supragingival plaque and the dorsum of the tongue, subjects began twice-daily rinsing with either an essential oil mouth rinse containing 0.09% zinc chloride (Tartar Control Listerine Antiseptic) or a negative control rinse. Bacterial sampling was repeated 12 h after the first rinse, and again 12 h after the final rinse 14 days later. The sampling schedule was adjusted according to whether the study was investigating daytime or overnight activity. Samples were plated on Schaedlers medium (total anaerobes), Schaedlers Nalidixic/Vancomycin medium (Gram-negative anaerobes), and OOPS medium (volatile sulphur compound (VSC)-producing organisms). Inter-group log10 transformed colony-forming units/ml counts from samples of supragingival plaque and tongue swabs on each of the three media were compared by analysis of covariance. RESULTS: The mean bacterial counts in subjects using the essential oil mouth rinse were significantly lower (p< or =0.005) than mean counts in subjects using the control rinse in all the comparisons, i.e., tongue and supragingival plaque samples on each of three media at two sampling periods in the daytime and overnight study, respectively. Mean bacterial count percent reductions for plaque samples ranged from 56.3 to 95.3; percent reductions for tongue samples ranged from 61.1 to 96.1. There was a trend to higher reductions after 14 days' rinsing than after the initial rinse. CONCLUSION: Rinsing with the essential oil mouth rinse can have long-lasting effects in reducing anaerobic bacteria overall as well as Gram-negative anaerobes and VSC-producing bacteria. The significant reductions in numbers of these bacteria produced by the essential oil mouth rinse, both in plaque and on the dorsum of the tongue, can play a key role in explaining the essential oil mouth rinse's effectiveness in reducing supragingival plaque and gingivitis as well as its effectiveness in controlling intrinsic oral malodor over prolonged periods.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Bacteria, Anaerobic/drug effects , Mouthwashes/pharmacology , Oils, Volatile/pharmacology , Adult , Bacteria/drug effects , Chlorides/pharmacology , Colony Count, Microbial , Cross-Over Studies , Dental Plaque/drug therapy , Dental Plaque/microbiology , Double-Blind Method , Female , Halitosis/drug therapy , Halitosis/microbiology , Humans , Male , Middle Aged , Mouthwashes/administration & dosage , Mouthwashes/chemistry , Organic Chemicals , Sulfur Compounds/metabolism , Time Factors , Tongue/microbiology , Zinc Compounds/pharmacology
18.
Clin Oral Investig ; 9(1): 38-45, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15449114

ABSTRACT

The control of oral malodor is well-recognized in efforts to improve oral health. Antimicrobial formulations can mitigate oral malodor, however, procedures to assess effects on oral bacteria including those implicated in halitosis are unavailable. This investigation examined the antimicrobial effects of a new liquid triclosan/copolymer dentifrice (test) formulation that demonstrated significant inhibition of oral malodor in previous organoleptic clinical studies. Procedures compared antimicrobial effects of the test and control formulations on a range of oral micro-organisms including members implicated in halitosis, substantive antimicrobial effects of formulations with hydroxyapatite as a surrogate for human teeth and ex vivo effects on oral bacteria from human volunteers. With Actinomyces viscosus, as a model system, the test formulation demonstrated a dose-dependent effect. At these concentrations the test formulation provided significant antimicrobial effects on 13 strains of oral bacteria including those implicated in bad breath at selected posttreatment time points. Treatment of hydroxyapatite by the test dentifrice resulted in a significant and substantive antimicrobial effect vs. controls. Oral bacteria from subjects treated ex vivo with the test dentifrice resulted in significant reductions in cultivable oral bacteria and odorigenic bacteria producing hydrogen sulfide. In summary, microbiological methods adapted to study odorigenic bacteria demonstrate the significant antimicrobial effects of the test (triclosan/copolymer) dentifrice with laboratory and clinical strains of oral bacteria implicated in bad breath.


Subject(s)
Anti-Infective Agents, Local/therapeutic use , Dentifrices/therapeutic use , Halitosis/drug therapy , Triclosan/therapeutic use , Actinomyces viscosus/drug effects , Complex Mixtures , Dose-Response Relationship, Drug , Female , Fluorides , Halitosis/microbiology , Humans , Male , Saliva/drug effects , Silicic Acid , Toothpastes
19.
Oral Microbiol Immunol ; 17(6): 354-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12485326

ABSTRACT

Cells of the periodontal pathogen Actinobacillus actinomycetemcomitans exhibit tight adherence to surfaces such as glass, plastic and hydroxyapatite, a property that probably plays an important role in the ability of this bacterium to colonize teeth and other surfaces. Tight adherence is mediated by long fibrils of bundled pili (fimbriae) that form on the surface of the cell. The flp-1 gene encodes the major pilin protein component of A. actinomycetemcomitans fimbriae. In this study we compared flp-1 DNA sequences from 43 strains of A. actinomycetemcomitans isolated in Europe, Japan and the United States and identified seven distinct flp-1 allelic classes. DNA and predicted protein sequences were almost completely conserved within each flp-1 class but were highly divergent between classes. Most amino acid substitutions occurred in the C-terminus of the pilin protein, a region that has been shown to be important for the bundling and adhesive properties of the pili. flp-1 classes correlated with serotypes and 16S rRNA genotypes in most strains. At least five strains showed evidence of horizontal transfer of flp-1 between strains of different serotypes and 16S rRNA genotypes. Four of the seven flp-1 classes were present in geographically diverse isolates. Strains representing all seven flp-1 classes, but not a strain carrying a transposon insertion in flp-1, bound avidly to polystyrene in an in vitro adherence assay. Strains representing six of the seven flp-1 classes were isolated from localized juvenile periodontitis patients, suggesting that phylogenetically diverse strains carry pathogenic potential. Our findings provide a framework for future biochemical, immunological and genetic studies of A. actinomycetemcomitans fimbriae.


Subject(s)
Aggregatibacter actinomycetemcomitans/genetics , Bacterial Proteins/genetics , Fimbriae, Bacterial/genetics , Genetic Variation/genetics , Actinobacillus Infections/physiopathology , Adult , Aggregatibacter actinomycetemcomitans/pathogenicity , Aggressive Periodontitis/microbiology , Alleles , Bacterial Adhesion/genetics , Conserved Sequence/genetics , DNA Transposable Elements/genetics , DNA, Bacterial/genetics , Female , Fimbriae Proteins/genetics , Gene Transfer, Horizontal/genetics , Genotype , Humans , Male , Periodontitis/microbiology , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Sequence Analysis, Protein , Serotyping , Virulence/genetics
20.
J Periodontol ; 73(6): 624-30, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12083535

ABSTRACT

BACKGROUND: Actinobacillus actinomycetemcomitans (Aa) is associated with localized aggressive periodontal disease in juveniles (LAgP). Lactoferrin (LF) is an iron-binding salivary protein that has been shown to kill Aa in its iron-free form (apo) and reduce binding to host cells in its iron-saturated form (halo). However, recent in vitro studies show that LF does not kill clinical isolates of Aa, and LF with reduced levels of bound iron does not interfere with its attachment. These findings suggest that colonization of Aa may occur more readily in an environment containing LF with low iron levels. The purpose of this study was to examine the relationship of LF iron levels in saliva of LAgP patients as compared to their age-, gender-, and race-matched controls. METHODS: Whole and parotid saliva was collected from LAgP patients and matched controls. Micrograms of LF/mg of protein as well as nanograms of iron/micrograms of LF were determined. Iron binding was determined in parotid saliva by addition of nonlabeled and 59Fe labeled iron. RESULTS: LAgP patients' whole saliva had higher LF levels than controls, but their LF contained less iron (P < or =0.005). No iron was found in LF from parotid saliva in either group. When iron was added to parotid saliva, the LAgP saliva bound 20 to 30 times less iron than controls (P< or =0.001). Finally, LF was identified as the major iron-binding protein in parotid saliva by 59Fe autoradiography and Western blotting. CONCLUSIONS: This study shows that the level of bound iron in LF is significantly reduced in LAgP patients compared to controls. These data suggest that LF from LAgP patients has a reduced capacity to bind iron and that LF iron levels may play an important role in Aa-induced LAgP.


Subject(s)
Aggressive Periodontitis/metabolism , Iron/metabolism , Lactoferrin/metabolism , Salivary Proteins and Peptides/metabolism , Adolescent , Adult , Autoradiography , Blotting, Western , Case-Control Studies , Child , Female , Humans , Immunoelectrophoresis/methods , Iron/analysis , Iron Radioisotopes/metabolism , Lactoferrin/analysis , Lactoferrin/chemistry , Male , Saliva/chemistry , Salivary Proteins and Peptides/analysis , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...