Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Mol Sci ; 25(7)2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38612423

RESUMEN

Periodontitis, characterized by persistent inflammation in the periodontium, is intricately connected to systemic diseases, including oral cancer. Bacteria, such as Porphyromonas gingivalis and Fusobacterium nucleatum, play a pivotal role in periodontitis development because they contribute to dysbiosis and tissue destruction. Thus, comprehending the interplay between these bacteria and their impacts on inflammation holds significant relevance in clinical understanding and treatment advancement. In the present work, we explored, for the first time, their impacts on the expressions of pro-inflammatory mediators after infecting oral keratinocytes (OKs) with a co-culture of pre-incubated P. gingivalis and F. nucleatum. Our results show that the co-culture increases IL-1ß, IL-8, and TNF-α expressions, synergistically augments IL-6, and translocates NF-kB to the cell nucleus. These changes in pro-inflammatory mediators-associated with chronic inflammation and cancer-correlate with an increase in cell migration following infection with the co-cultured bacteria or P. gingivalis alone. This effect depends on TLR4 because TLR4 knockdown notably impacts IL-6 expression and cell migration. Our study unveils, for the first time, crucial insights into the outcomes of their co-culture on virulence, unraveling the role of bacterial interactions in polymicrobial diseases and potential links to oral cancer.


Asunto(s)
Neoplasias de la Boca , Periodontitis , Humanos , Técnicas de Cocultivo , Interleucina-6 , Receptor Toll-Like 4 , Inflamación , Mediadores de Inflamación , Queratinocitos
2.
Clin Oral Investig ; 27(9): 5509-5518, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37505241

RESUMEN

OBJECTIVES: To evaluate the relationship between obesity and periodontitis staging compared with periodontal healthy or gingivitis in pregnant women. MATERIALS AND METHODS: An analytical cross-sectional study was conducted on pregnant women between 11 and 14 weeks of pregnancy. Sociodemographic, clinical, obstetric, and periodontal variables were studied. The exposure variable was obesity (body mass index [BMI] ≥ 30), and the primary outcome was periodontitis staging versus periodontal healthy/gingivitis. Data were analysed and estimated by multinomial logistic regression models. RESULTS: The present study screened 1086 pregnancies and analysed 972 women with a median age of 29 years; 36.8% were diagnosed as obese. 26.9% of patients were diagnosed as periodontal healthy or gingivitis, 5.5% with stage I periodontitis, 38.6% with stage II periodontitis, 24% with stage III periodontitis, and 5.1% with stage IV periodontitis. After identifying and adjusting for confounding variables (educational level and plaque index), obesity had a relative risk ratio (RRR) of 1.66 (95% CI: 1.05-2.64; p = 0.03) and 1.57 (95% CI: 1.09-2.27; p = 0.015) for stage III periodontitis compared to periodontal healthy/gingivitis and stage II periodontitis, respectively. CONCLUSION: Besides the already known risk indicators for periodontitis (age, smoking, and educational level), our study suggests a relationship between obesity and periodontitis staging in pregnancy. CLINICAL RELEVANCE: Obesity can alter host immune responses, leading to increased susceptibility to infections and overactive host immunity, which could influence the prevalence and severity of maternal periodontitis in pregnancy.


Asunto(s)
Gingivitis , Periodontitis , Femenino , Humanos , Embarazo , Adulto , Estudios Transversales , Periodontitis/complicaciones , Periodontitis/epidemiología , Obesidad/complicaciones , Obesidad/epidemiología , Factores de Riesgo , Gingivitis/epidemiología
3.
Int Endod J ; 56(10): 1270-1283, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37461231

RESUMEN

AIM: Angiogenesis contributes to the development of apical periodontitis, periodontitis, and other oral pathologies; however, it remains unclear how this process is triggered. The aim was to evaluate whether lipopolysaccharide (LPS) from Porphyromonas endodontalis and Porphyromonas gingivalis induced angiogenesis-related effects in vitro via TLR2 and TLR4. METHODOLOGY: Porphyromonas endodontalis LPS (ATCC 35406 and clinical isolate) was purified with TRIzol, whereas P. gingivalis LPS was obtained commercially. The effects of the different LPS (24 h) in endothelial cell migration were analysed by Transwell assays, following quantification in an optical microscope (40×). The effects of LPS on FAK Y397 phosphorylation were assessed by Western blotting. Angiogenesis in vitro was determined in an endothelial tube formation assay (14 h) in Matrigel in the absence or presence of either LPS. IL-6 and VEGF-A levels were determined in cell supernatants, following 24 h treatment with LPS, and measured in multiplex bead immunoassay. The involvement of TLR2 and TLR4 was assessed with blocking antibodies. The statistical analysis was performed using STATA 12® (StataCorp LP). RESULTS: The results revealed that P. endodontalis LPS, but not P. gingivalis LPS, stimulated endothelial cell migration. Pre-treatment with anti-TLR2 and anti-TLR4 antibodies prevented P. endodontalis LPS-induced cell migration. P. endodontalis LPS promoted FAK phosphorylation on Y397, as observed by an increased p-FAK/FAK ratio. Both P. gingivalis and P. endodontalis LPS (ATCC 35406) induced endothelial tube formation in a TLR-2 and -4-dependent manner, as shown by using blocking antibodies, however, only TLR2 blocking decreased tube formation induced by P. endodontalis (clinical isolate). Moreover, all LPS induced IL-6 and VEGF-A synthesis in endothelial cells. TLR2 and TLR4 were required for IL-6 induction by P. endodontalis LPS (ATCC 35406), while only TLR4 was involved in IL-6 secretion by the other LPS. Finally, VEGF-A synthesis did not require TLR signalling. CONCLUSION: Porphyromonas endodontalis and P. gingivalis LPS induced angiogenesis via TLR2 and TLR4. Collectively, these data contribute to understanding the role of LPS from Porphyromonas spp. in angiogenesis and TLR involvement.


Asunto(s)
Lipopolisacáridos , Receptor Toll-Like 2 , Lipopolisacáridos/farmacología , Receptor Toll-Like 2/metabolismo , Porphyromonas gingivalis/metabolismo , Porphyromonas endodontalis/metabolismo , Factor A de Crecimiento Endotelial Vascular , Células Endoteliales/metabolismo , Anticuerpos Bloqueadores , Interleucina-6 , Receptor Toll-Like 4/metabolismo
4.
Int J Mol Sci ; 23(17)2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36077408

RESUMEN

Apical Lesions of Endodontic Origin (ALEO) are initiated by polymicrobial endodontic canal infection. Porphyromonas gingivalis (Pg) and Porphyromonas endodontalis (Pe) lipopolysaccharides (LPS) can induce a pro-inflammatory macrophage response through their recognition by TLR2 and TLR4. However, polarization responses induced by Pg and/or Pe LPS in macrophages are not fully understood. We aimed to characterize the polarization profiles of macrophages differentiated from THP-1 cells following Pg and/or Pe LPS stimulation from reference strain and clinical isolates. A modified LPS purification protocol was implemented and the electrophoretic LPS profiles were characterized. THP-1 human monocytes differentiated to macrophages were stimulated with Pg and Pe LPS. Polarization profiles were characterized through cell surface markers and secreted cytokines levels after 24 h of stimulation. TLR2 and TLR4 cell surfaces and transcriptional levels were determined after 24 or 2 h of LPS stimulation, respectively. LPS from Pg induced a predominant M1 profile in macrophages evidenced by changes in the expression of the surface marker CD64 and pro-inflammatory cytokine profiles, TNF-α, IL-1ß, IL-6, and IL-12. Pe LPS was unable to induce a significant response. TLR2 and TLR4 expressions were neither modified by Pg or Pe LPS. Pg LPS, but not Pe LPS, induced a macrophage M1 Profile.


Asunto(s)
Lipopolisacáridos , Porphyromonas gingivalis , Humanos , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Porphyromonas endodontalis/metabolismo , Porphyromonas gingivalis/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
5.
Clin Oral Investig ; 26(1): 993-1001, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34313848

RESUMEN

OBJECTIVE: Porphyromonas (P.) species (spp.) are a major etiological agent of apical periodontitis (AP), which in turn represents a risk factor for cardiovascular diseases. This study explored the associations between endodontic infection with Porphyromonas species, the systemic bacterial burden, and cardiovascular risk, based on high-sensitivity C-reactive protein (hsCRP), in young adults with AP. MATERIALS AND METHODS: Cross-sectional study. Otherwise, healthy individuals with AP and controls (n = 80, ≤ 40 years) were recruited at the University Dental Clinic. Oral parameters and classic cardiovascular risk factors were registered. Endodontic Porphyromonas endodontalis and Porphyromonas gingivalis were identified using conventional PCR. Serum concentrations of anti-P. endodontalis and anti-P. gingivalis antibodies, and endotoxins were determined through ELISA and Limulus-amebocyte assays. Serum hsCRP was determined for cardiovascular risk stratification. RESULTS: Intracanal detection of P. endodontalis and P. gingivalis in AP were 33.3% and 22.9%, respectively. Serum anti-P. endodontalis and anti-P. gingivalis IgG was higher in AP than controls (p < 0.05 and p = 0.057, respectively). Intracanal P. endodontalis associated with higher endotoxemia (p < 0.05). Among endodontic factors, the presence (OR 4.2-5.5, p < 0.05) and the number of apical lesions (OR 2.3, p < 0.05) associated with moderate-severe cardiovascular risk, whereas anti-P. endodontalis IgG were protective (OR 0.3, p > 0.05). CONCLUSIONS: AP and infection with P. endodontalis positively associated with cardiovascular risk based on hsCRP levels and endotoxemia, respectively, whereas anti-P. endodontalis IgG response seems to be protective against low-grade systemic inflammation. CLINICAL RELEVANCE: Apical periodontitis and endodontic P. endodontalis can influence the systemic burden with impact on the surrogate cardiovascular risk marker hsCRP, providing mechanistic links.


Asunto(s)
Enfermedades Cardiovasculares , Periodontitis Periapical , Estudios Transversales , ADN Bacteriano , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Porphyromonas/genética , Factores de Riesgo , Adulto Joven
6.
Int J Mol Sci ; 22(21)2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34769262

RESUMEN

Periodontitis is a chronic inflammatory immune disease associated with a dysbiotic state, influenced by keystone bacterial species responsible for disrupting the periodontal tissue homeostasis. Furthermore, the severity of periodontitis is determined by the interaction between the immune cell response in front of periodontitis-associated species, which leads to the destruction of supporting periodontal tissues and tooth loss in a susceptible host. The persistent bacterial challenge induces modifications in the permeability and ulceration of the sulcular epithelium, which facilitates the systemic translocation of periodontitis-associated bacteria into distant tissues and organs. This stimulates the secretion of pro-inflammatory molecules and a chronic activation of immune cells, contributing to a systemic pro-inflammatory status that has been linked with a higher risk of several systemic diseases, such as type 2 diabetes mellitus (T2DM) and gestational diabetes mellitus (GDM). Although periodontitis and GDM share the common feature of systemic inflammation, the molecular mechanistic link of this association has not been completely clarified. This review aims to examine the potential biological mechanisms involved in the association between periodontitis and GDM, highlighting the contribution of both diseases to systemic inflammation and the role of new molecular participants, such as extracellular vesicles and non-coding RNAs, which could act as novel molecular intercellular linkers between periodontal and placental tissues.


Asunto(s)
Diabetes Gestacional , Periodontitis , Periodoncio , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/microbiología , Diabetes Gestacional/metabolismo , Diabetes Gestacional/microbiología , Femenino , Humanos , Periodontitis/etiología , Periodontitis/metabolismo , Periodontitis/microbiología , Periodoncio/metabolismo , Periodoncio/microbiología , Embarazo
7.
Front Cell Infect Microbiol ; 11: 649925, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33816354

RESUMEN

Apical periodontitis is an inflammatory disease of microbial etiology. It has been suggested that endodontic bacterial DNA might translocate to distant organs via blood vessels, but no studies have been conducted. We aimed first to explore overall extraradicular infection, as well as specifically by Porphyromonas spp; and their potential to translocate from infected root canals to blood through peripheral blood mononuclear cells. In this cross-sectional study, healthy individuals with and without a diagnosis of apical periodontitis with an associated apical lesion of endodontic origin (both, symptomatic and asymptomatic) were included. Apical lesions (N=64) were collected from volunteers with an indication of tooth extraction. Intracanal samples (N=39) and respective peripheral blood mononuclear cells from apical periodontitis (n=14) individuals with an indication of endodontic treatment, as well as from healthy individuals (n=14) were collected. The detection frequencies and loads (DNA copies/mg or DNA copies/µL) of total bacteria, Porphyromonas endodontalis and Porphyromonas gingivalis were measured by qPCR. In apical lesions, the detection frequencies (%) and median bacterial loads (DNA copies/mg) respectively were 70.8% and 4521.6 for total bacteria; 21.5% and 1789.7 for Porphyromonas endodontalis; and 18.4% and 1493.9 for Porphyromonas gingivalis. In intracanal exudates, the detection frequencies and median bacterial loads respectively were 100% and 21089.2 (DNA copies/µL) for total bacteria, 41% and 8263.9 for Porphyromonas endodontalis; and 20.5%, median 12538.9 for Porphyromonas gingivalis. Finally, bacteria were detected in all samples of peripheral blood mononuclear cells including apical periodontitis and healthy groups, though total bacterial loads (median DNA copies/µL) were significantly higher in apical periodontitis (953.6) compared to controls (300.7), p<0.05. Porphyromonas endodontalis was equally detected in both groups (50%), but its bacterial load tended to be higher in apical periodontitis (262.3) than controls (158.8), p>0.05; Porphyromonas gingivalis was not detected. Bacteria and specifically Porphyromonas spp. were frequently detected in endodontic canals and apical lesions. Also, total bacteria and Porphyromonas endodontalis DNA were detected in peripheral blood mononuclear cells, supporting their plausible role in bacterial systemic translocation.


Asunto(s)
Traslocación Bacteriana , Periodontitis Periapical , Estudios Transversales , ADN Bacteriano , Humanos , Leucocitos Mononucleares , Porphyromonas endodontalis
8.
Periodontol 2000 ; 86(1): 57-78, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33690899

RESUMEN

The subgingival crevice harbors diverse microbial communities. Shifts in the composition of these communities occur with the development of gingivitis and periodontitis, which are considered as successive stages of periodontal health deterioration. It is not clear, however, to what extent health- and gingivitis-associated microbiota are protective, or whether these communities facilitate the successive growth of periodontitis-associated taxa. To further our understanding of the dynamics of the microbial stimuli that trigger disruptions in periodontal homeostasis, we reviewed the available literature with the aim of defining specific microbial signatures associated with different stages of periodontal dysbiosis. Although several studies have evaluated the subgingival communities present in different periodontal conditions, we found limited evidence for the direct comparison of communities in health, gingivitis, and periodontitis. Therefore, we aimed to better define subgingival microbiome shifts by merging and reanalyzing, using unified bioinformatic processing strategies, publicly available 16S ribosomal RNA gene amplicon datasets of periodontal health, gingivitis, and periodontitis. Despite inherent methodological differences across studies, distinct community structures were found for health, gingivitis, and periodontitis, demonstrating the specific associations between gingival tissue status and the subgingival microbiome. Consistent with the concept that periodontal dysbiosis is the result of a process of microbial succession without replacement, more species were detected in disease than in health. However, gingivitis-associated communities were more diverse than those from subjects with periodontitis, suggesting that certain species ultimately become dominant as dysbiosis progresses. We identified the bacterial species associated with each periodontal condition and prevalent species that do not change in abundance from one state to another (core species), and we also outlined species co-occurrence patterns via network analysis. Most periodontitis-associated species were rarely detected in health but were frequently detected, albeit in low abundance, in gingivitis, which suggests that gingivitis and periodontitis are a continuum. Overall, we provide a framework of subgingival microbiome shifts, which can be used to generate hypotheses with respect to community assembly processes and the emergence of periodontal dysbiosis.


Asunto(s)
Gingivitis , Microbiota , Periodontitis , Disbiosis , Humanos , ARN Ribosómico 16S/genética
9.
ISME J ; 15(5): 1490-1504, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33372193

RESUMEN

Recent studies describe in detail the shifts in composition of human-associated polymicrobial communities from health to disease. However, the specific processes that drive the colonization and overgrowth of pathogens within these communities remain incompletely understood. We used in vitro culture systems and a disease-relevant mouse model to show that population size, which determines the availability of an endogenous diffusible small molecule, limits the growth, colonization, and in vivo virulence of the human oral pathogen Porphyromonas gingivalis. This bacterial pathogen overcomes the requirement for an endogenous cue by utilizing a cell-density dependent, growth-promoting, soluble molecule provided by the symbiotic early colonizer Veillonella parvula, but not produced by other commensals tested. Our work shows that exchange of cell-density-dependent diffusible cues between specific early and late colonizing species in a polymicrobial community drives microbial successions, pathogen colonization and disease development, representing a target process for manipulation of the microbiome towards the healthy state.


Asunto(s)
Biopelículas , Veillonella , Animales , Ratones , Porphyromonas gingivalis , Virulencia
10.
Front Immunol ; 11: 591240, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193431

RESUMEN

Aggregatibacter actinomycetemcomitans is a Gram-negative oral bacterium with high immunostimulatory and pathogenic potential involved in the onset and progression of periodontitis, a chronic disease characterized by aberrant immune responses followed by tooth-supporting bone resorption, which eventually leads to tooth loss. While several studies have provided evidence related to the virulence factors of A. actinomycetemcomitans involved in the host cell death and immune evasion, such as its most studied primate-specific virulence factor, leukotoxin, the role of specific lipopolysaccharide (LPS) domains remain poorly understood. Here, we analyzed the role of the immunodominant domain of the LPS of A. actinomycetemcomitans termed O-polysaccharide (O-PS), which differentiates the distinct bacterial serotypes based on its antigenicity. To determine the role of the O-PS in the immunogenicity and virulence of A. actinomycetemcomitans during periodontitis, we analyzed the in vivo and in vitro effect of an O-PS-defective transposon mutant serotype b strain, characterized by the deletion of the rmlC gene encoding the α-L-rhamnose sugar biosynthetic enzyme. Induction of experimental periodontitis using the O-PS-defective rmlC mutant strain resulted in lower tooth-supporting bone resorption, infiltration of Th1, Th17, and Th22 lymphocytes, and expression of Ahr, Il1b, Il17, Il23, Tlr4, and RANKL (Tnfsf11) in the periodontal lesions as compared with the wild-type A. actinomycetemcomitans strain. In addition, the O-PS-defective rmlC mutant strain led to impaired activation of antigen-presenting cells, with less expression of the co-stimulatory molecules CD40 and CD80 in B lymphocytes and dendritic cells, and downregulated expression of Tnfa and Il1b in splenocytes. In conclusion, these data demonstrate that the O-PS from the serotype b of A. actinomycetemcomitans plays a key role in the capacity of the bacterium to prime oral innate and adaptive immune responses, by triggering the Th1 and Th17-driven tooth-supporting bone resorption during periodontitis.


Asunto(s)
Aggregatibacter actinomycetemcomitans/inmunología , Aggregatibacter actinomycetemcomitans/patogenicidad , Periodontitis/inmunología , Periodontitis/microbiología , Polisacáridos Bacterianos/inmunología , Virulencia/inmunología , Aggregatibacter actinomycetemcomitans/genética , Pérdida de Hueso Alveolar/diagnóstico por imagen , Pérdida de Hueso Alveolar/etiología , Pérdida de Hueso Alveolar/patología , Animales , Biología Computacional/métodos , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno/inmunología , Lipopolisacáridos/inmunología , Ratones , Mutación , Periodontitis/complicaciones , Periodontitis/diagnóstico , Serogrupo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Virulencia , Microtomografía por Rayos X
11.
Artículo en Inglés | MEDLINE | ID: mdl-31355151

RESUMEN

Porphyromonas gingivalis has been extensively associated with both the onset and progression of periodontitis. We previously isolated and characterized two P. gingivalis strains, one from a patient exhibiting severe chronic periodontitis (CP3) and another from a periodontally healthy individual (H3). We previously showed that CP3 and H3 exhibit differences in virulence since H3 showed a lower resistance to cationic peptides compared with CP3, and a lower ability to induce proliferation in gingival epithelial cells. Here, we aimed to determine whether differences in virulence between these two strains are associated with the presence or absence of specific genes encoding virulence factors. We sequenced the whole genomes of both P. gingivalis CP3 and H3 and conducted a comparative analysis regarding P. gingivalis virulence genetic determinants. To do so, we performed a homology search of predicted protein sequences in CP3 and H3 genomes against the most characterized virulence genes for P. gingivalis available in the literature. In addition, we performed a genomic comparison of CP3 and H3 with all the 62 genomes of P. gingivalis found in NCBI's RefSeq database. This approach allowed us to determine the evolutionary relationships of CP3 and H3 with other virulent and avirulent strains; and additionally, to detect variability in presence/absence of virulence genes among P. gingivalis genomes. Our results show genetic variability in the hemagglutinin genes. While CP3 possesses one copy of hagA and two of hagC, H3 has no hagA and only one copy of hagC. Experimentally, this finding is related to lower in vitro hemmaglutination ability of H3 compared to CP3. Moreover, while CP3 encodes a gene for a major fimbrium subunit FimA type 4 (CP3_00160), H3 possess a FimA type 1 (H3_01400). Such genetic differences are in agreement with both lower biofilm formation ability and less intracellular invasion to oral epithelial cells exhibited by H3, compared with the virulent strain CP3. Therefore, here we provide new results on the genome sequences, comparative genomics analyses, and phenotypic analyses of two P. gingivalis strains. The genomics comparison of these two strains with the other 62 genomes included in the analysis provided relevant results regarding genetic determinants and their association with P. gingivalis virulence.


Asunto(s)
Periodontitis Crónica/patología , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidad , Factores de Virulencia/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Estudios de Casos y Controles , Línea Celular , Periodontitis Crónica/microbiología , Células Epiteliales/microbiología , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Ontología de Genes , Variación Genética , Genómica , Encía/microbiología , Humanos , Lectinas/genética , Lectinas/metabolismo , Anotación de Secuencia Molecular , Fenotipo , Filogenia , Porphyromonas gingivalis/clasificación , Porphyromonas gingivalis/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Análisis de Secuencia de ADN , Virulencia , Factores de Virulencia/metabolismo
12.
Mediators Inflamm ; 2019: 1029857, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31049022

RESUMEN

Periodontitis is characterized by a chronic inflammation produced in response to a disease-associated multispecies bacterial community in the subgingival region. Although the inflammatory processes occur locally in the oral cavity, several studies have determined that inflammatory mediators produced during periodontitis, as well as subgingival species and bacterial components, can disseminate from the oral cavity, contributing therefore, to various extraoral diseases like cancer. Interestingly, carcinogenesis associated with periodontal species has been observed in both the oral cavity and in extra oral sites. In this review, several studies were summarized showing a strong association between orodigestive cancers and poor oral health, presence of periodontitis-associated bacteria, tooth loss, and clinical signs of periodontitis. Proinflammatory pathways were also summarized. Such pathways are activated either by mono- or polymicrobial infections, resulting in an increase in the expression of proinflammatory molecules such as IL-6, IL-8, IL-1ß, and TNF-α. In addition, it has been shown that several periodontitis-associated species induce the expression of genes related to cell proliferation, cell cycle, apoptosis, transport, and immune and inflammatory responses. Intriguingly, many of these pathways are linked to carcinogenesis. Among them, the activation of Toll-like receptors (TLRs) and antiapoptotic pathways (such as the PI3K/Akt, JAK/STAT, and MAPK pathways), the reduction of proapoptotic protein expression, the increase in cell migration and invasion, and the enhancement in metastasis are addressed. Considering that periodontitis is a polymicrobial disease, it is likely that mixed species promote carcinogenesis both in the oral cavity and in extra oral tissues and probably-as observed in periodontitis-synergistic and/or antagonistic interactions occur between microbes in the community. To date, a good amount of studies has allowed us to understand how monospecies infections activate pathways involved in tumorigenesis; however, more studies are needed to determine the combined effect of oral species in carcinogenesis.


Asunto(s)
Carcinogénesis/inmunología , Carcinogénesis/metabolismo , Periodontitis Crónica/inmunología , Periodontitis Crónica/metabolismo , Inflamación/metabolismo , Animales , Citocinas/metabolismo , Humanos
13.
World J Gastroenterol ; 24(28): 3071-3089, 2018 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-30065554

RESUMEN

Helicobacter pylori (H. pylori) is present in roughly 50% of the human population worldwide and infection levels reach over 70% in developing countries. The infection has classically been associated with different gastro-intestinal diseases, but also with extra gastric diseases. Despite such associations, the bacterium frequently persists in the human host without inducing disease, and it has been suggested that H. pylori may also play a beneficial role in health. To understand how H. pylori can produce such diverse effects in the human host, several studies have focused on understanding the local and systemic effects triggered by this bacterium. One of the main mechanisms by which H. pylori is thought to damage the host is by inducing local and systemic inflammation. However, more recently, studies are beginning to focus on the effects of H. pylori and its metabolism on the gastric and intestinal microbiome. The objective of this review is to discuss how H. pylori has co-evolved with humans, how H. pylori presence is associated with positive and negative effects in human health and how inflammation and/or changes in the microbiome are associated with the observed outcomes.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Infecciones por Helicobacter/fisiopatología , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno/fisiología , Inflamación/fisiopatología , Coevolución Biológica/fisiología , Mucosa Gástrica/microbiología , Mucosa Gástrica/fisiopatología , Infecciones por Helicobacter/epidemiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Humanos , Inflamación/microbiología
14.
J Appl Oral Sci ; 26: e20170075, 2018 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-29364340

RESUMEN

The aim of this double-blind, placebo-controlled and parallel- arm randomized clinical trial was to evaluate the effects of Lactobacillus rhamnosus SP1-containing probiotic sachet and azithromycin tablets as an adjunct to nonsurgical therapy in clinical parameters and in presence and levels of Tannerella forsythia, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans. Forty-seven systemically healthy volunteers with chronic periodontitis were recruited and monitored clinically and microbiologically at baseline for 3, 6 and 9 months after therapy. Subgingival plaque samples were collected from four periodontal sites with clinical attachment level ≥1 mm, probing pocket depth ≥4 mm and bleeding on probing, one site in each quadrant. Samples were cultivated and processed using the PCR technique. Patients received nonsurgical therapy including scaling and root planing (SRP) and were randomly assigned to a probiotic (n=16), antibiotic (n = 16) or placebo (n = 15) group. L. rhamnosus SP1 was taken once a day for 3 months. Azithromycin 500mg was taken once a day for 5 days. All groups showed improvements in clinical and microbiological parameters at all time points evaluated. Probiotic and antibiotic groups showed greater reductions in cultivable microbiota compared with baseline. The placebo group showed greater reduction in number of subjects with P. gingivalis compared with baseline. However, there were no significant differences between groups. The adjunctive use of L. rhamnosus SP1 sachets and azithromycin during initial therapy resulted in similar clinical and microbiological improvements compared with the placebo group.


Asunto(s)
Antibacterianos/uso terapéutico , Azitromicina/uso terapéutico , Periodontitis Crónica/tratamiento farmacológico , Lacticaseibacillus rhamnosus/química , Probióticos/uso terapéutico , Adulto , Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Aggregatibacter actinomycetemcomitans/aislamiento & purificación , Análisis de Varianza , Antibacterianos/farmacología , Azitromicina/farmacología , Recuento de Colonia Microbiana , Placa Dental/tratamiento farmacológico , Placa Dental/microbiología , Raspado Dental/métodos , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Índice Periodontal , Efecto Placebo , Reacción en Cadena de la Polimerasa , Porphyromonas gingivalis/efectos de los fármacos , Porphyromonas gingivalis/aislamiento & purificación , Probióticos/farmacología , Estadísticas no Paramétricas , Tannerella forsythia/efectos de los fármacos , Tannerella forsythia/aislamiento & purificación , Factores de Tiempo , Resultado del Tratamiento
15.
J. appl. oral sci ; 26: e20170075, 2018. tab, graf
Artículo en Inglés | LILACS, BBO - Odontología | ID: biblio-893719

RESUMEN

ABSTRACT Objective: The aim of this double-blind, placebo-controlled and parallel- arm randomized clinical trial was to evaluate the effects of Lactobacillus rhamnosus SP1-containing probiotic sachet and azithromycin tablets as an adjunct to nonsurgical therapy in clinical parameters and in presence and levels of Tannerella forsythia, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans. Material and Methods: Forty-seven systemically healthy volunteers with chronic periodontitis were recruited and monitored clinically and microbiologically at baseline for 3, 6 and 9 months after therapy. Subgingival plaque samples were collected from four periodontal sites with clinical attachment level ≥1 mm, probing pocket depth ≥4 mm and bleeding on probing, one site in each quadrant. Samples were cultivated and processed using the PCR technique. Patients received nonsurgical therapy including scaling and root planing (SRP) and were randomly assigned to a probiotic (n=16), antibiotic (n = 16) or placebo (n = 15) group. L. rhamnosus SP1 was taken once a day for 3 months. Azithromycin 500mg was taken once a day for 5 days. Results: All groups showed improvements in clinical and microbiological parameters at all time points evaluated. Probiotic and antibiotic groups showed greater reductions in cultivable microbiota compared with baseline. The placebo group showed greater reduction in number of subjects with P. gingivalis compared with baseline. However, there were no significant differences between groups. Conclusions: The adjunctive use of L. rhamnosus SP1 sachets and azithromycin during initial therapy resulted in similar clinical and microbiological improvements compared with the placebo group.


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Azitromicina/uso terapéutico , Probióticos/uso terapéutico , Lacticaseibacillus rhamnosus/química , Periodontitis Crónica/tratamiento farmacológico , Antibacterianos/uso terapéutico , Factores de Tiempo , Recuento de Colonia Microbiana , Efecto Placebo , Índice Periodontal , Reacción en Cadena de la Polimerasa , Método Doble Ciego , Análisis de Varianza , Raspado Dental/métodos , Resultado del Tratamiento , Aggregatibacter actinomycetemcomitans/aislamiento & purificación , Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Azitromicina/farmacología , Porphyromonas gingivalis/aislamiento & purificación , Porphyromonas gingivalis/efectos de los fármacos , Estadísticas no Paramétricas , Probióticos/farmacología , Placa Dental/microbiología , Placa Dental/tratamiento farmacológico , Tannerella forsythia/aislamiento & purificación , Tannerella forsythia/efectos de los fármacos , Persona de Mediana Edad , Antibacterianos/farmacología
16.
Microbiol Spectr ; 5(4)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28840820

RESUMEN

The three main oral diseases of humans, that is, caries, periodontal diseases, and oral candidiasis, are associated with microbiome shifts initiated by changes in the oral environment and/or decreased effectiveness of mucosal immune surveillance. In this review, we discuss the role that microbial-based therapies may have in the control of these conditions. Most investigations on the use of microorganisms for management of oral disease have been conducted with probiotic strains with some positive but very discrete clinical outcomes. Other strategies such as whole oral microbiome transplantation or modification of community function by enrichment with health-promoting indigenous oral strains may offer more promise, but research in this field is still in its infancy. Any microbial-based therapeutics for oral conditions, however, are likely to be only one component within a holistic preventive strategy that should also aim at modification of the environmental influences responsible for the initiation and perpetuation of microbiome shifts associated with oral dysbiosis.


Asunto(s)
Agentes de Control Biológico/uso terapéutico , Candidiasis Bucal/terapia , Caries Dental/terapia , Microbiota/fisiología , Boca/microbiología , Enfermedades Periodontales/terapia , Probióticos/uso terapéutico , Candidiasis Bucal/microbiología , Caries Dental/microbiología , Disbiosis/terapia , Humanos , Enfermedades Periodontales/microbiología , Terapia de Fagos/métodos
17.
Bio Protoc ; 7(24)2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29333479

RESUMEN

The oral microbiome has been implicated as a trigger for immune responsiveness in the oral cavity, particularly in the setting of the inflammatory disease periodontitis. The protocol presented here is aimed at characterizing the oral microbiome in murine models at steady state and during perturbations of immunity or physiology. Herein, we describe murine oral microbiome sampling procedures, processing of low biomass samples and subsequent microbiome characterization based on 16S rRNA gene sequencing.

18.
J Calif Dent Assoc ; 44(7): 421-35, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27514154

RESUMEN

High-throughput 16S rRNA gene sequencing has allowed the characterization of subgingival microbiome shifts from health to periodontitis identifying health-associated, periodontitis-associated and core species, which preserve their proportions from health to disease. The development of gingivitis is also characterized by distinct shifts. Microbiome shifts resemble microbial successions and result from interspecies interactions and community adaptation to the changing environment as inflammation ensues. Gingivitis-associated and core species are proposed as likely mediators of microbiome transitions.


Asunto(s)
Encía/microbiología , Microbiota/fisiología , Enfermedades Periodontales/microbiología , Genes Bacterianos/genética , Gingivitis/microbiología , Humanos , Interacciones Microbianas/fisiología , ARN Bacteriano/análisis , ARN Ribosómico 16S/análisis , Transcriptoma/genética
19.
J Periodontol ; 87(8): 944-52, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26944407

RESUMEN

BACKGROUND: Probiotics are living microorganisms that provide beneficial effects for the host when administered in proper quantities. The aim of this double-masked placebo-controlled parallel-arm randomized clinical trial is to evaluate the clinical effects of a Lactobacillus rhamnosus SP1-containing probiotic sachet as an adjunct to non-surgical therapy. METHODS: Twenty-eight systemically healthy volunteers with chronic periodontitis were recruited and monitored clinically at baseline and 3, 6, 9, and 12 months after therapy. Clinical parameters measured included plaque accumulation, bleeding on probing, probing depths (PDs), and clinical attachment loss. Patients received non-surgical therapy, including scaling and root planing (SRP), and were assigned randomly to a test (SRP + probiotic, n = 14) or control (SRP + placebo, n = 14) group. The intake, once a day for 3 months, of an L. rhamnosus SP1 probiotic sachet commenced after the last session of SRP. RESULTS: Both test and control groups showed improvements in clinical parameters at all time points evaluated. However, the test group showed greater reductions in PD than the control. Also, at initial visits and after 1-year follow-up, the test group showed a statistically significant reduction in the number of participants with PD ≥6 mm, indicating a reduced need for surgery, in contrast to the placebo group. CONCLUSION: The results of this trial indicate that oral administration of L. rhamnosus SP1 resulted in similar clinical improvements compared with SRP alone.


Asunto(s)
Periodontitis Crónica/terapia , Lacticaseibacillus rhamnosus , Probióticos/uso terapéutico , Raspado Dental , Estudios de Seguimiento , Humanos , Pérdida de la Inserción Periodontal , Índice Periodontal , Aplanamiento de la Raíz
20.
Anaerobe ; 33: 25-32, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25638398

RESUMEN

Virulence factors on the surface of Porphyromonas gingivalis constitute the first line of interaction with host cells and contribute to immune modulation and periodontitis progression. In order to characterize surface virulence factors present on P. gingivalis, we obtained clinical isolates from healthy and periodontitis subjects and compared them with reference strains. Colony morphology, aggregation in liquid medium, surface charge, membrane permeability to bactericidal compounds, novobiocin and polymyxin B resistance, capsule presence and lipopolysaccharide (LPS) profiles were evaluated. By comparing isolates from healthy and periodontitis subjects, differences in colony morphology and aggregation in liquid culture were found; the latter being similar to two reference strains. These differences were not a consequence of variations in bacterial surface charge. Furthermore, isolates also presented differences in polymyxin B and novobiocin resistance; isolates from healthy subjects were susceptible to polymyxin B and resistant to novobiocin and, in contrast, isolates from periodontitis subjects were resistant to polymyxin B and susceptible to novobiocin. These changes in antimicrobial resistance levels correlate with variations in LPS profiles, since -unlike periodontitis isolates-isolates from healthy samples synthesize LPS molecules lacking both O-antigen moieties and anionic polysaccharide. Additionally, this phenotype correlated with the absence of O-antigen ligase activity. Altogether, our results reveal novel variations on surface components of P. gingivalis isolates obtained from healthy and periodontitis subjects that could be associated with differences in bacterial virulence and periodontitis progression.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Lipopolisacáridos/metabolismo , Periodontitis/microbiología , Polimixina B/farmacología , Porphyromonas gingivalis/efectos de los fármacos , Porphyromonas gingivalis/fisiología , Adulto , Infecciones por Bacteroidaceae/microbiología , Estudios de Casos y Controles , Permeabilidad de la Membrana Celular , Femenino , Genes Bacterianos , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Factores de Virulencia , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...