Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Neuroinflammation ; 20(1): 228, 2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37803465

ABSTRACT

INTRODUCTION: Periodontitis-related oral microbial dysbiosis is thought to contribute to Alzheimer's disease (AD) neuroinflammation and brain amyloid production. Since probiotics can modulate periodontitis/oral dysbiosis, this study examined the effects of a probiotic/lantibiotic, nisin, in modulating brain pathology triggered by periodontitis. METHODS: A polymicrobial mouse model of periodontal disease was used to evaluate the effects of this disease on brain microbiome dysbiosis, neuroinflammation, Alzheimer's-related changes, and nisin's therapeutic potential in this context. RESULTS: 16S sequencing and real-time PCR data revealed that Nisin treatment mitigated the changes in the brain microbiome composition, diversity, and community structure, and reduced the levels of periodontal pathogen DNA in the brain induced by periodontal disease. Nisin treatment significantly decreased the mRNA expression of pro-inflammatory cytokines (Interleukin-1ß/IL-1 ß, Interleukin 6/IL-6, and Tumor Necrosis Factor α/TNF-α) in the brain that were elevated by periodontal infection. In addition, the concentrations of amyloid-ß 42 (Aß42), total Tau, and Tau (pS199) (445.69 ± 120.03, 1420.85 ± 331.40, 137.20 ± 36.01) were significantly higher in the infection group compared to the control group (193.01 ± 31.82, 384.27 ± 363.93, 6.09 ± 10.85), respectively. Nisin treatment markedly reduced the Aß42 (261.80 ± 52.50), total Tau (865.37 ± 304.93), and phosphorylated Tau (82.53 ± 15.77) deposition in the brain of the infection group. DISCUSSION: Nisin abrogation of brain microbiome dysbiosis induces beneficial effects on AD-like pathogenic changes and neuroinflammation, and thereby may serve as a potential therapeutic for periodontal-dysbiosis-related AD.


Subject(s)
Alzheimer Disease , Bacteriocins , Microbiota , Nisin , Periodontitis , Probiotics , Mice , Animals , Alzheimer Disease/pathology , Nisin/metabolism , Bacteriocins/metabolism , Neuroinflammatory Diseases , Dysbiosis/drug therapy , Dysbiosis/metabolism , Periodontitis/metabolism , Brain/metabolism , Amyloid beta-Peptides/metabolism , Interleukin-6/metabolism , Probiotics/therapeutic use
2.
PLoS Pathog ; 17(7): e1009311, 2021 07.
Article in English | MEDLINE | ID: mdl-34255809

ABSTRACT

Periodontal disease is driven by dysbiosis in the oral microbiome, resulting in over-representation of species that induce the release of pro-inflammatory cytokines, chemokines, and tissue-remodeling matrix metalloproteinases (MMPs) in the periodontium. These chronic tissue-destructive inflammatory responses result in gradual loss of tooth-supporting alveolar bone. The oral spirochete Treponema denticola, is consistently found at significantly elevated levels in periodontal lesions. Host-expressed Toll-Like Receptor 2 (TLR2) senses a variety of bacterial ligands, including acylated lipopolysaccharides and lipoproteins. T. denticola dentilisin, a surface-expressed protease complex comprised of three lipoproteins has been implicated as a virulence factor in periodontal disease, primarily due to its proteolytic activity. While the role of acylated bacterial components in induction of inflammation is well-studied, little attention has been given to the potential role of the acylated nature of dentilisin. The purpose of this study was to test the hypothesis that T. denticola dentilisin activates a TLR2-dependent mechanism, leading to upregulation of tissue-destructive genes in periodontal tissue. RNA-sequencing of periodontal ligament cells challenged with T. denticola bacteria revealed significant upregulation of genes associated with extracellular matrix organization and degradation including potentially tissue-specific inducible MMPs that may play novel roles in modulating host immune responses that have yet to be characterized within the context of oral disease. The Gram-negative oral commensal, Veillonella parvula, failed to upregulate these same MMPs. Dentilisin-induced upregulation of MMPs was mediated via TLR2 and MyD88 activation, since knockdown of expression of either abrogated these effects. Challenge with purified dentilisin upregulated the same MMPs while a dentilisin-deficient T. denticola mutant had no effect. Finally, T. denticola-mediated activation of TLR2/MyD88 lead to the nuclear translocation of the transcription factor Sp1, which was shown to be a critical regulator of all T. denticola-dependent MMP expression. Taken together, these data suggest that T. denticola dentilisin stimulates tissue-destructive cellular processes in a TLR2/MyD88/Sp1-dependent fashion.


Subject(s)
Bacterial Proteins/metabolism , Peptide Hydrolases/metabolism , Periodontal Diseases , Treponemal Infections/metabolism , Virulence Factors/metabolism , Cells, Cultured , Humans , Matrix Metalloproteinases/metabolism , Myeloid Differentiation Factor 88/metabolism , Periodontal Diseases/metabolism , Periodontal Diseases/microbiology , Periodontal Diseases/pathology , Periodontal Ligament , Sp1 Transcription Factor/metabolism , Toll-Like Receptor 2/metabolism , Treponema denticola , Treponemal Infections/pathology , Up-Regulation
3.
Periodontol 2000 ; 2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38073011

ABSTRACT

The oral squamous cell carcinoma (OSCC) 5 year survival rate of 41% has marginally improved in the last few years, with less than a 1% improvement per year from 2005 to 2017, with higher survival rates when detected at early stages. Based on histopathological grading of oral dysplasia, it is estimated that severe dysplasia has a malignant transformation rate of 7%-50%. Despite these numbers, oral dysplasia grading does not reliably predict its clinical behavior. Thus, more accurate markers predicting oral dysplasia progression to cancer would enable better targeting of these lesions for closer follow-up, especially in the early stages of the disease. In this context, molecular biomarkers derived from genetics, proteins, and metabolites play key roles in clinical oncology. These molecular signatures can help predict the likelihood of OSCC development and/or progression and have the potential to detect the disease at an early stage and, support treatment decision-making and predict treatment responsiveness. Also, identifying reliable biomarkers for OSCC detection that can be obtained non-invasively would enhance management of OSCC. This review will discuss biomarkers for OSCC that have emerged from different biological areas, including genomics, transcriptomics, proteomics, metabolomics, immunomics, and microbiomics.

4.
PLoS Pathog ; 16(10): e1008881, 2020 10.
Article in English | MEDLINE | ID: mdl-33002094

ABSTRACT

Epidemiological studies reveal significant associations between periodontitis and oral cancer. However, knowledge about the contribution of periodontal pathogens to oral cancer and potential regulatory mechanisms involved is limited. Previously, we showed that nisin, a bacteriocin and commonly used food preservative, reduced oral cancer tumorigenesis and extended the life expectancy in tumor-bearing mice. In addition, nisin has antimicrobial effects on key periodontal pathogens. Thus, the purpose of this study was to test the hypothesis that key periodontal pathogens (Porphyromonas gingivalis, Treponema denticola, and Fusobacterium nucleatum) promote oral cancer via specific host-bacterial interactions, and that bacteriocin/nisin therapy may modulate these responses. All three periodontal pathogens enhanced oral squamous cell carcinoma (OSCC) cell migration, invasion, tumorsphere formation, and tumorigenesis in vivo, without significantly affecting cell proliferation or apoptosis. In contrast, oral commensal bacteria did not affect OSCC cell migration. Pathogen-enhanced OSCC cell migration was mediated via integrin alpha V and FAK activation, since stably blocking alpha V or FAK expression abrogated these effects. Nisin inhibited these pathogen-mediated processes. Further, Treponema denticola induced TLR2 and 4 and MyD88 expression. Stable suppression of MyD88 significantly inhibited Treponema denticola-induced FAK activation and abrogated pathogen-induced migration. Together, these data demonstrate that periodontal pathogens contribute to a highly aggressive cancer phenotype via crosstalk between TLR/MyD88 and integrin/FAK signaling. Nisin can modulate these pathogen-mediated effects, and thus has therapeutic potential as an antimicrobial and anti-tumorigenic agent.


Subject(s)
Bacteroidaceae Infections/drug therapy , Carcinoma, Squamous Cell/drug therapy , Mouth Neoplasms/drug therapy , Periodontitis/drug therapy , Porphyromonas gingivalis/drug effects , Probiotics/pharmacology , Animals , Apoptosis , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Cell Movement , Cell Proliferation , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Periodontitis/metabolism , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/pathogenicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Periodontol 2000 ; 87(1): 76-93, 2021 10.
Article in English | MEDLINE | ID: mdl-34463982

ABSTRACT

The oral microbiome is a community of microorganisms, comprised of bacteria, fungi, viruses, archaea, and protozoa, that form a complex ecosystem within the oral cavity. Although minor perturbations in the environment are frequent and compensable, major shifts in the oral microbiome can promote an unbalanced state, known as dysbiosis. Dysbiosis can promote oral diseases, including periodontitis. In addition, oral dysbiosis has been associated with other systemic diseases, including cancer. The objective of this review is to evaluate the epidemiologic evidence linking periodontitis to oral, gastrointestinal, lung, breast, prostate, and uterine cancers, as well as describe new evidence and insights into the role of oral dysbiosis in the etiology and pathogenesis of the cancer types discussed. Finally, we discuss how antimicrobials, antimicrobial peptides, and probiotics may be promising tools to prevent and treat these cancers, targeting both the microbes and associated carcinogenesis processes. These findings represent a novel paradigm in the pathogenesis and treatment of cancer focused on the oral microbiome and antimicrobial-based therapies.


Subject(s)
Anti-Infective Agents , Microbiota , Mouth Neoplasms , Anti-Infective Agents/therapeutic use , Dysbiosis , Humans , Male , Mouth Neoplasms/drug therapy
6.
Periodontol 2000 ; 87(1): 50-75, 2021 10.
Article in English | MEDLINE | ID: mdl-34463996

ABSTRACT

Periodontitis has been associated with many systemic diseases and conditions, including metabolic syndrome. Metabolic syndrome is a cluster of conditions that occur concomitantly and together they increase the risk of cardiovascular disease and double the risk of type 2 diabetes. In this review, we focus on the association between metabolic syndrome and periodontitis; however, we also include information on diabetes mellitus and cardiovascular disease, since these two conditions are significantly intertwined with metabolic syndrome. With regard to periodontitis and metabolic syndrome, to date, the vast majority of studies point to an association between these two conditions and also demonstrate that periodontitis can contribute to the development of, or can worsen, metabolic syndrome. Evaluating the effect of metabolic syndrome on the salivary microbiome, data presented herein support the hypothesis that the salivary bacterial profile is altered in metabolic syndrome patients compared with healthy patients. Considering periodontitis and these three conditions, the vast majority of human and animal studies point to an association between periodontitis and metabolic syndrome, diabetes, and cardiovascular disease. Moreover, there is evidence to suggest that metabolic syndrome and diabetes can alter the oral microbiome. However, more studies are needed to fully understand the influence these conditions have on each other.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Syndrome , Microbiota , Periodontitis , Animals , Cytokines , Diabetes Mellitus, Type 2/complications , Humans , Lipids , Metabolic Syndrome/complications , Periodontitis/complications
7.
J Periodontal Res ; 56(2): 314-329, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33314132

ABSTRACT

BACKGROUND AND OBJECTIVE: There is a close relationship between inflammation and bone remodeling in the periodontium. However, previous studies have not delineated the alterations in calcium (Ca) metabolism during periodontitis progression. The aim of this current investigation was to examine Ca dynamics in alveolar bone of rats during progression of ligature-induced periodontal inflammation by using 45 Ca, which is an index of hard tissue neogenesis. MATERIAL AND METHODS: To induce periodontitis, the maxillary right first molar (M1) of 8-week-old male rats was ligated with a silk suture for 1, 3, 7, and 28 days. The left M1 was not ligated as a control. To evaluate resultant changes in bone neogenesis, 45 CaCl2 was injected intraperitoneally 24 hours before euthanasia. The left-and-right palatal mucosa, molar teeth (M1 and M2), and alveolar bone were harvested for evaluation of 45 Ca radioactivity using a liquid scintillation counter. The distribution of 45 Ca in maxillary tissues was evaluated using autoradiography (ARG). In addition, we analyzed the bone volume fraction (BV/TV) and bone mineral density (BMD) of the alveolar bone by micro-computed tomography. To investigate the number of osteoclasts and osteoblasts, tartrate-resistant acid phosphatase (TRAP) and bone-specific alkaline phosphatase (BAP) were measured by an enzymatic assay and immunohistochemistry, respectively. RESULTS: 45 Ca radioactivity in the alveolar bone of the ligature side decreased by 8% compared to the unligated control-side on day 1, whereas on day 7, it markedly increased by 33%. The 45 Ca levels in the gingival tissue and molar teeth were slightly but significantly lower than the control-side on day 1 and higher from day 3 to 28. The variation in 45 Ca levels for the alveolar bone was greater and specific compared with other tissues. Furthermore, on day 7, ARG data revealed that 45 Ca on the control side was primarily localized to the periodontal ligament (PDL) space and alveolar bone crest and barely detected in the gingival tissues and deeper parts of the alveolar bone. On the ligature side, 45 Ca disappeared from the PDL and alveolar crest, but instead was broadly and significantly increased within the deeper zones of the alveolar bone and furcation areas and distant from the site of ligature placement and periodontal inflammation. In the shallow zone of the alveolar bone, these changes in 45 Ca levels on day 7 were consistent with decreases in the bone structural parameters (BV/TV and BMD), enhanced osteoclast presence, and suppressed levels of BAP expression in osteoblasts. In contrast, the deep zone and furcation area showed that TRAP-positive cells increased, but BAP expression was maintained in the resorption lacunae of the alveolar bone. CONCLUSION: During periodontitis progression in rats, 45 Ca levels in the alveolar bone exhibited biphasic alterations, namely decreases and increases. These data indicate that periodontitis induces a wide range of site-specific Ca metabolism alterations within the alveolar bone.


Subject(s)
Alveolar Bone Loss , Alveolar Bone Loss/diagnostic imaging , Animals , Calcium , Disease Models, Animal , Inflammation , Male , Osteoclasts , Radius , Rats , Rats, Wistar , X-Ray Microtomography
8.
Periodontol 2000 ; 82(1): 115-128, 2020 02.
Article in English | MEDLINE | ID: mdl-31850641

ABSTRACT

Periodontal studies using transcriptomics, proteomics, and metabolomics encompass the collection of mRNA transcripts, proteins, and small-molecule chemicals in the context of periodontal health and disease. The number of studies using these approaches has significantly increased in the last decade and they have provided new insight into the pathogenesis and host-microbe interactions that define periodontal diseases. This review provides an overview of current molecular findings using -omic approaches that underlie periodontal disease, including modulation of the host immune response, tissue homeostasis, and complex metabolic processes of the host and the oral microbiome. Integration of these -omic approaches will broaden our perspective of the molecular mechanisms involved in periodontal disease, advancing and improving the diagnosis and treatment of various stages and forms of periodontal disease.


Subject(s)
Metabolome , Transcriptome , Humans , Metabolomics , Proteome , Proteomics
9.
Periodontol 2000 ; 82(1): 173-185, 2020 02.
Article in English | MEDLINE | ID: mdl-31850634

ABSTRACT

The effects of probiotic supplementation on systemic health and gastrointestinal diseases have been investigated in numerous studies. The aim of this review is to provide an overview of probiotics and their effects on periodontal health. Probiotics show beneficial effects as adjunctive therapeutics and as stand-alone agents in the treatment and prevention of gingivitis as well as specific clinical parameters of periodontitis. This review focuses on the clinical and microbiological aspects of probiotics in the context of health, gingivitis, and periodontitis. In addition, a special focus on nisin-producing probiotics and nisin itself showcase their significant potential for oral and systemic use.


Subject(s)
Gingivitis , Nisin , Periodontitis , Probiotics , Humans
10.
Cell Microbiol ; 20(4)2018 04.
Article in English | MEDLINE | ID: mdl-29205773

ABSTRACT

Host-derived matrix metalloproteinases (MMPs) and bacterial proteases mediate destruction of extracellular matrices and supporting alveolar bone in periodontitis. The Treponema denticola dentilisin protease induces MMP-2 expression and activation in periodontal ligament (PDL) cells, and dentilisin-mediated activation of pro-MMP-2 is required for cellular fibronectin degradation. Here, we report that T. denticola regulates MMP-2 expression through epigenetic modifications in the periodontium. PDL cells were treated with epigenetic enzyme inhibitors before or after T. denticola challenge. Fibronectin fragmentation, MMP-2 expression, and activation were assessed by immunoblot, zymography, and qRT-PCR, respectively. Chromatin modification enzyme expression in T. denticola-challenged PDL cells and periodontal tissues were evaluated using gene arrays. Several classes of epigenetic enzymes showed significant alterations in transcription in diseased tissue and T. denticola-challenged PDL cells. T. denticola-mediated MMP-2 expression and activation were significantly reduced in PDL cells treated with inhibitors of aurora kinases and histone deacetylases. In contrast, DNA methyltransferase inhibitors had little effect, and inhibitors of histone acetyltransferases, methyltransferases, and demethylases exacerbated T. denticola-mediated MMP-2 expression and activation. Chronic epigenetic changes in periodontal tissues mediated by T. denticola or other oral microbes may contribute to the limited success of conventional treatment of chronic periodontitis and may be amenable to therapeutic reversal.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Periodontal Ligament/enzymology , Periodontal Ligament/microbiology , Treponema denticola , Cells, Cultured , Epigenesis, Genetic , Histone Code , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Treponema denticola/physiology
11.
J Proteome Res ; 16(3): 1315-1326, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28168879

ABSTRACT

Cancer cells use alternate energetic pathways; however, cancer stem cell (CSC) metabolic energetic pathways are unknown. The purpose of this study was to define the metabolic characteristics of head and neck cancer at different points of its pathogenesis with a focus on its CSC compartment. UPLC-MS/MS-profiling and GC-MS-validation studies of human head and neck cancer tissue, saliva, and plasma were used in conjunction with in vitro and in vivo models to carry out this investigation. We identified metabolite biomarker panels that distinguish head and neck cancer from healthy controls, and confirmed involvement of glutamate and glutaminolysis. Glutaminase, which catalyzes glutamate formation from glutamine, and aldehyde dehydrogenase (ALDH), a stemness marker, were highly expressed in primary and metastatic head and neck cancer tissues, tumorspheres, and CSC versus controls. Exogenous glutamine induced stemness via glutaminase, whereas inhibiting glutaminase suppressed stemness in vitro and tumorigenesis in vivo. Head and neck CSC (CD44hi/ALDHhi) exhibited higher glutaminase, glutamate, and sphere levels than CD44lo/ALDHlo cells. Glutaminase drove transcriptional and translational ALDH expression, and glutamine directed even CD44lo/ALDHlo cells toward stemness. Glutaminolysis regulates tumorigenesis and CSC metabolism via ALDH. These findings indicate that glutamate is an important marker of cancer metabolism whose regulation via glutaminase works in concert with ALDH to mediate cancer stemness. Future analyses of glutaminolytic-ALDH driven mechanisms underlying tumorigenic transitions may help in the development of targeted therapies for head and neck cancer and its CSC compartment.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Carcinoma, Squamous Cell/metabolism , Glutamine/metabolism , Head and Neck Neoplasms/metabolism , Neoplastic Stem Cells/pathology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Gas Chromatography-Mass Spectrometry , Glutamic Acid , Glutaminase , Humans , Male , Middle Aged , Neoplastic Stem Cells/metabolism , Tandem Mass Spectrometry
12.
Bio Protoc ; 14(7): e4970, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38618176

ABSTRACT

Periodontal disease is characterized by the destruction of the hard and soft tissues comprising the periodontium. This destruction translates to a degradation of the extracellular matrices (ECM), mediated by bacterial proteases, host-derived matrix metalloproteinases (MMPs), and other proteases released by host tissues and immune cells. Bacterial pathogens interact with host tissue, triggering adverse cellular functions, including a heightened immune response, tissue destruction, and tissue migration. The oral spirochete Treponema denticola is highly associated with periodontal disease. Dentilisin, a T. denticola outer membrane protein complex, contributes to the chronic activation of pro-MMP-2 in periodontal ligament (PDL) cells and triggers increased expression levels of activators and effectors of active MMP-2 in PDL cells. Despite these advances, no mechanism for dentilisin-induced MMP-2 activation or PDL cytopathic behaviors leading to disease is known. Here, we describe a method for purification of large amounts of the dentilisin protease complex from T. denticola and demonstrate its ability to activate MMP-2, a key regulator of periodontal tissue homeostasis. The T. denticola dentilisin and MMP-2 activation model presented here may provide new insights into the dentilisin protein and identify potential therapeutic targets for further research. Key features • This protocol builds upon a method described by Cunningham et al. [1] for selective release of Treponema outer membrane proteins. • We adapted the protocol for the purification of biologically active, detergent-stable outer membrane protein complexes from large batch cultures of T. denticola. • The protocol involves large-scale preparative electrophoresis using a Model 491 Prep Cell. • We then use gelatin zymography to demonstrate the activity of the purified dentilisin complex by its ability to activate matrix metalloproteinase 2 (MMP-2).

13.
NPJ Biofilms Microbiomes ; 10(1): 3, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38233485

ABSTRACT

Oral microbiome dysbiosis mediates chronic periodontal disease, gut microbial dysbiosis, and mucosal barrier disfunction that leads to steatohepatitis via the enterohepatic circulation. Improving this dysbiosis towards health may improve liver disease. Treatment with antibiotics and probiotics have been used to modulate the microbial, immunological, and clinical landscape of periodontal disease with some success. The aim of the present investigation was to evaluate the potential for nisin, an antimicrobial peptide produced by Lactococcus lactis, to counteract the periodontitis-associated gut dysbiosis and to modulate the glycolipid-metabolism and inflammation in the liver. Periodontal pathogens, namely Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia and Fusobacterium nucleatum, were administrated topically onto the oral cavity to establish polymicrobial periodontal disease in mice. In the context of disease, nisin treatment significantly shifted the microbiome towards a new composition, commensurate with health while preventing the harmful inflammation in the small intestine concomitant with decreased villi structural integrity, and heightened hepatic exposure to bacteria and lipid and malondialdehyde accumulation in the liver. Validation with RNA Seq analyses, confirmed the significant infection-related alteration of several genes involved in mitochondrial dysregulation, oxidative phosphorylation, and metal/iron binding and their restitution following nisin treatment. In support of these in vivo findings indicating that periodontopathogens induce gastrointestinal and liver distant organ lesions, human autopsy specimens demonstrated a correlation between tooth loss and severity of liver disease. Nisin's ability to shift the gut and liver microbiome towards a new state commensurate with health while mitigating enteritis, represents a novel approach to treating NAFLD-steatohepatitis-associated periodontal disease.


Subject(s)
Bacteriocins , Nisin , Non-alcoholic Fatty Liver Disease , Periodontal Diseases , Mice , Humans , Animals , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Nisin/pharmacology , Nisin/metabolism , Dysbiosis , Periodontal Diseases/microbiology , Porphyromonas gingivalis/metabolism , Inflammation/complications , Oxidative Stress
14.
Microorganisms ; 11(9)2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37764094

ABSTRACT

Oral potentially malignant disorders (OPMDs) are a group of conditions that carry a risk of oral squamous cell carcinoma (OSCC) development. Recent studies indicate that periodontal disease-associated pathogenic bacteria may play a role in the transition from healthy mucosa to dysplasia and to OSCC. Yet, the microbial signatures associated with the transition from healthy mucosa to dysplasia have not been established. To characterize oral microbial signatures at these different sites, we performed a 16S sequencing analysis of both oral swab and formalin-fixed, paraffin-embedded tissue (FFPE) samples. We collected oral swabs from healthy mucosa (from healthy patients), histologically normal mucosa adjacent to dysplasia, and low-grade oral dysplasia. Additionally, FFPE samples from histologically normal mucosa adjacent to OSCC, plus low grade and high-grade oral dysplasia samples were also collected. The collected data demonstrate significant differences in the alpha and beta microbial diversities of different sites in oral mucosa, dysplasia, and OSCC, as well as increased dissimilarities within these sites. We found that the Proteobacteria phyla abundance increased, concurrent with a progressive decrease in the Firmicutes phyla abundance, as well as altered levels of Enterococcus cecorum, Fusobacterium periodonticum, Prevotella melaninogenica, and Fusobacterium canifelinum when moving from healthy to diseased sites. Moreover, the swab sample analysis indicates that the oral microbiome may be altered in areas that are histologically normal, including in mucosa adjacent to dysplasia. Furthermore, trends in specific microbiome changes in oral swab samples preceded those in the tissues, signifying early detection opportunities for clinical diagnosis. In addition, we evaluated the gene expression profile of OSCC cells (HSC-3) infected with either P. gingivalis, T. denticola, F. nucelatum, or S. sanguinis and found that the three periodontopathogens enrich genetic processes related to cancer progression, including skin keratinization/cornification, while the commensal enriched processes related to RNA processing and adhesion. Finally, we reviewed the dysplasia microbiome literature and found a significant decrease in commensal bacteria, such as the Streptococci genus, and a simultaneous increase in pathogenic bacteria, mainly Bacteroidetes phyla and Fusobacterium genus. These findings suggest that features of the oral microbiome can serve as novel biomarkers for dysplasia and OSCC disease progression.

15.
Biochim Biophys Acta ; 1816(1): 80-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21586315

ABSTRACT

Sirtuins (SIRT1-7), the mammalian homologues of the Sir2 gene in yeast, have emerging roles in age-related diseases, such as cardiac hypertrophy, diabetes, obesity, and cancer. However, the role of several sirtuin family members, including SIRT1 and SIRT3, in cancer has been controversial. The aim of this review is to explore and discuss the seemingly dichotomous role of SIRT3 in cancer biology with particular emphasis on its potential role as a tumor promoter and tumor suppressor. This review will also discuss the potential role of SIRT3 as a novel therapeutic target to treat cancer.


Subject(s)
Neoplasms/etiology , Sirtuin 3/physiology , Animals , Apoptosis , Cell Survival , Energy Metabolism , Humans , Neoplasms/prevention & control , Sirtuin 3/analysis , Sirtuin 3/antagonists & inhibitors
16.
Cancer ; 118(23): 5800-10, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22674009

ABSTRACT

BACKGROUND: Regulating cross-talk between anoikis and survival signaling pathways is crucial to regulating tissue processes and mitigating diseases like cancer. Previously, the authors demonstrated that anoikis activates a signaling pathway involving the CD95/Fas-mediated signaling pathway that is regulated by receptor-interacting protein (RIP), a kinase that shuttles between Fas-mediated cell death and integrin/focal adhesion kinase (FAK)-mediated survival pathways. Because it is known that sirtuin-3 (SIRT3), a nicotinamide adenine dinucleotide-dependent deacetylase, regulates cell survival, metabolism, and tumorigenesis, the authors hypothesized that SIRT3 may engage in cross-talk with Fas/RIP/integrin/FAK survival-death pathways in cancer cell systems. METHODS: Using immunohistochemical staining, immunoblotting, human tissue microarrays, and overexpression and suppression approaches in vitro and in vivo, the roles of RIP and SIRT3 were examined in oral squamous cell carcinoma (OSCC) anoikis resistance and tumorigenesis. RESULTS: RIP and SIRT3 had opposite expression profiles in OSCC cells and tissues. Stable suppression of RIP enhanced SIRT3 levels, whereas stable suppression of SIRT3 did not impact RIP levels in OSCC cells. The authors observed that, as OSCC cells became anoikis-resistant, they formed multicellular aggregates or oraspheres in suspension conditions, and their expression of SIRT3 increased as their RIP expression decreased. Also, anoikis-resistant OSCC cells with higher SIRT3 and low RIP expression induced an increased tumor burden and incidence in mice, unlike their adherent OSCC cell counterparts. Furthermore, stable suppression of SIRT3 inhibited anoikis resistance and reduced tumor incidence. CONCLUSIONS: The current results indicted that RIP is a likely upstream, negative regulator of SIRT3 in anoikis resistance, and an anoikis-resistant orasphere phenotype defined by higher SIRT3 and low RIP expression contributes to a more aggressive phenotype in OSCC development.


Subject(s)
Anoikis , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Sirtuin 3/physiology , Adult , Aged , Carcinoma, Squamous Cell/etiology , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Mouth Neoplasms/etiology , Receptor-Interacting Protein Serine-Threonine Kinases/analysis , Sirtuin 3/analysis
17.
J Biomed Nanotechnol ; 18(4): 1227-1235, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35854440

ABSTRACT

Bacteriocins are peptides produced by bacteria to inhibit the growth of other prokaryotes. Nisin is a bacteriocin widely used in the food industry and for biomedical applications. However, bacteriocins have some limitations, as they experience mechanisms of resistance, degradation by proteases, and suboptimal intracellular delivery. Combining bacteriocins with nanoscale drug delivery systems (nano-DDS) is an approach that can help overcome these limitations. Among the nano-DDS, solid lipid nanoparticles (SLN) have been described as promising candidates, because of their potential for industrial scale-up and lower toxicity. The objective of this proof-of-concept study was to investigate the use of nisin-loaded SLN (SLN-Nisin) as an antimicrobial and anticancer therapeutic. We show that SLN-Nisin can significantly inhibit the growth of the oral pathogen, Treponema denticola, disrupt oral biofilms, and decrease oral squamous cell carcinoma cell (OSCC) viability compared to free nisin. Further, analysis with scanning electron microscopy (SEM) revealed significant morphological changes in OSCC cells challenged with SLN-Nisin, compared to the empty-nanoparticle or free nisin, indicating that SLN-Nisin likely decreases cell viability by increasing pore formation. This data reveals that nano-DDS are robust tools that can enhance bacteriocin properties.


Subject(s)
Antineoplastic Agents , Bacteriocins , Carcinoma, Squamous Cell , Mouth Neoplasms , Nanoparticles , Nisin , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/pharmacology , Bacteriocins/chemistry , Bacteriocins/metabolism , Bacteriocins/pharmacology , Biofilms , Humans , Liposomes , Nisin/chemistry , Nisin/metabolism , Nisin/pharmacology
18.
Microorganisms ; 10(7)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35889055

ABSTRACT

Peri-implantitis is characterized by chronic inflammation of the peri-implant supporting tissues that progressively and irreversibly leads to bone loss and, consequently, implant loss. Similar to periodontal disease, oral dysbiosis is thought to be a driver of peri-implantitis. However, managing peri-implantitis with traditional treatment methods, such as nonsurgical debridement or surgery, is not always successful. Thus, novel strategies have been proposed to address these shortcomings. One strategy is the use of probiotics as antimicrobial agents since they are considered safe for humans and the environment. Specifically, the probiotic Lactococcus lactis produces nisin, which has been used worldwide for food preservation. The objective of this study was to determine whether nisin and the wild-type (WT) nisin-producing L. lactis probiotic can disrupt oral pathogenic biofilms and promote a healthier oral microbiome within these oral biofilms on titanium discs. Using confocal imaging and 16S rRNA sequencing, this study revealed that nisin and WT L. lactis probiotic disrupt oral pathogenic biofilms in a peri-implantitis setting in vitro. More specifically, nisin decreased the viability of the pathogen-spiked biofilms dose-dependently from 62.53 ± 3.69% to 54.26 ± 3.35% and 44.88 ± 2.98%, respectively. Similarly, 105 CFU/mL of WT L. lactis significantly decreased biofilm viability to 52.45 ± 3.41%. Further, both treatments shift the composition, relative abundance, and diversity levels of these biofilms towards healthy control levels. A total of 1 µg/mL of nisin and 103 CFU/mL of WT L. lactis were able to revert the pathogen-mediated changes in the Proteobacteria (from 80.5 ± 2.9% to 75.6 ± 2.0%, 78.0 ± 2.8%, and 75.1 ± 5.3%, respectively) and Firmicutes (from 11.6 ± 1.6% to 15.4 ± 1.3%, 13.8 ± 1.8%, and 13.7 ± 2.6%, respectively) phyla back towards control levels. Thus, nisin and its nisin-producing L. lactis probiotic may be useful in treating peri-implantitis by promoting healthier oral biofilms, which may be useful for improving patient oral health.

19.
NPJ Biofilms Microbiomes ; 8(1): 45, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672331

ABSTRACT

Dysbiosis of the oral microbiome mediates chronic periodontal disease. Realignment of microbial dysbiosis towards health may prevent disease. Treatment with antibiotics and probiotics can modulate the microbial, immunological, and clinical landscape of periodontal disease with some success. Antibacterial peptides or bacteriocins, such as nisin, and a nisin-producing probiotic, Lactococcus lactis, have not been examined in this context, yet warrant examination because of their biomedical benefits in eradicating biofilms and pathogenic bacteria, modulating immune mechanisms, and their safety profile in humans. This study's goal was to examine the potential for nisin and a nisin-producing probiotic to abrogate periodontal bone loss, the host inflammatory response, and changes in oral microbiome composition in a polymicrobial mouse model of periodontal disease. Nisin and a nisin-producing Lactococcus lactis probiotic significantly decreased the levels of several periodontal pathogens, alveolar bone loss, and the oral and systemic inflammatory host response. Surprisingly, nisin and/or the nisin-producing L. lactis probiotic enhanced the population of fibroblasts and osteoblasts despite the polymicrobial infection. Nisin mediated human periodontal ligament cell proliferation dose-dependently by increasing the proliferation marker, Ki-67. Nisin and probiotic treatment significantly shifted the oral microbiome towards the healthy control state; health was associated with Proteobacteria, whereas 3 retroviruses were associated with disease. Disease-associated microbial species were correlated with IL-6 levels. Nisin or nisin-producing probiotic's ability to shift the oral microbiome towards health, mitigate periodontal destruction and the host immune response, and promote a novel proliferative phenotype in reparative connective tissue cells, addresses key aspects of the pathogenesis of periodontal disease and reveals a new biomedical application for nisin in treatment of periodontitis and reparative medicine.


Subject(s)
Alveolar Bone Loss , Lactococcus lactis , Microbiota , Nisin , Periodontal Diseases , Probiotics , Alveolar Bone Loss/prevention & control , Animals , Anti-Bacterial Agents , Cell Proliferation , Dysbiosis , Lactococcus lactis/genetics , Mice , Periodontal Diseases/microbiology
20.
J Proteome Res ; 10(11): 5232-41, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21961579

ABSTRACT

High-resolution magic-angle spinning (HR-MAS) proton NMR spectroscopy is used to explore the metabolic signatures of head and neck squamous cell carcinoma (HNSCC) which included matched normal adjacent tissue (NAT) and tumor originating from tongue, lip, larynx and oral cavity, and associated lymph-node metastatic (LN-Met) tissues. A total of 43 tissues (18 NAT, 18 Tumor and 7 LN-Met) from 22 HNSCC patients were analyzed. Principal Component Analysis of NMR data showed a clear classification between NAT and tumor tissues, however, LN-Met tissues were classified among tumor. A partial least-squares discriminant analysis model generated from NMR metabolic profiles was used to differentiate normal from tumor samples (Q(2) > 0.80, Receiver Operator Characteristic area under the curve >0.86, using 7-fold cross validation). HNSCC and LN-Met tissues showed elevated levels of lactate, amino acids including leucine, isoleucine, valine, alanine, glutamine, glutamate, aspartate, glycine, phenylalanine and tyrosine, choline containing compounds, creatine, taurine, glutathione, and decreased levels of triglycerides. These elevated metabolites were associated with highly active glycolysis, increased amino acids influx (anaplerosis) into the TCA cycle, altered energy metabolism, membrane choline phospholipid metabolism, and oxidative and osmotic defense mechanisms. Moreover, decreased levels of triglycerides may indicate lipolysis followed by ß-oxidation of fatty acids that may exist to deliver bioenergy for rapid tumor cell proliferation and growth.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Amino Acids/metabolism , Area Under Curve , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology , Female , Glycolysis , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/pathology , Humans , Lipid Metabolism , Magnetic Resonance Spectroscopy , Male , Middle Aged , Multivariate Analysis , Prognosis , ROC Curve
SELECTION OF CITATIONS
SEARCH DETAIL