Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 397
Filtrar
1.
Curr Atheroscler Rep ; 26(7): 289-304, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38805145

RESUMEN

PURPOSE OF REVIEW: In this review, we explore the intriguing and evolving connections between bacterial extracellular membrane nanovesicles (BEMNs) and atherosclerosis development, highlighting the evidence on molecular mechanisms by which BEMNs can promote the athero-inflammatory process that is central to the progression of atherosclerosis. RECENT FINDINGS: Atherosclerosis is a chronic inflammatory disease primarily driven by metabolic and lifestyle factors; however, some studies have suggested that bacterial infections may contribute to the development of both atherogenesis and inflammation in atherosclerotic lesions. In particular, the participation of BEMNs in atherosclerosis pathogenesis has attracted special attention. We provide some general insights into how the immune system responds to potential threats such as BEMNs during the development of atherosclerosis. A comprehensive understanding of contribution of BEMNs to atherosclerosis pathogenesis may lead to the development of targeted interventions for the prevention and treatment of the disease.


Asunto(s)
Aterosclerosis , Vesículas Extracelulares , Aterosclerosis/microbiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Humanos , Vesículas Extracelulares/metabolismo , Animales , Inflamación/metabolismo , Bacterias/metabolismo , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/metabolismo
2.
Cell Mol Biol (Noisy-le-grand) ; 70(8): 201-206, 2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39262241

RESUMEN

Oral infections can activate local and systemic inflammation. The inflammatory response plays a main role in atherosclerosis. several studies have reported a relation between oral pathogen infection and Atherosclerosis. Recently it was indicated that some oral microbiome has a significant role in triggering atherosclerosis. Denaturing Gradient Gel Electrophoresis (DGGE) is an acceptable assay for identification of uncultivable bacteria. Therefore, we compared the bacterial population diversity in the oral microbiota between atherosclerosis patients and healthy people. Oral microbiota profiling was performed for 139 individuals including 89 patients with CAD and 50 healthy individuals. After DNA extracted from saliva, PCR products were examined and evaluated using DGGE assay. We found that significant relationship between the increased risk of atherosclerosis and the presence of Actinomyces oris, Enterococcus faecalis, Bacterium strain sulresv, Bacterium Culaenoe, NC4, NC7, and NC5 in atherosclerosis patients and healthy individuals. There was also a significant relationship between reducing the risk of atherosclerosis in the presence of NC3 and Entreococcus munotii in atherosclerosis patients and healthy individuals.  In conclusion, presence of some oral microbiota increases the risk of atherosclerosis and the presence of some oral microbiota reduces the risk, so the oral microbiota should be further examined to determine its potential as a biomarker for atherosclerosis.


Asunto(s)
Aterosclerosis , Electroforesis en Gel de Gradiente Desnaturalizante , Microbiota , Boca , Humanos , Aterosclerosis/microbiología , Microbiota/genética , Femenino , Masculino , Persona de Mediana Edad , Boca/microbiología , Estudios de Casos y Controles , Saliva/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Anciano , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Adulto
3.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38892314

RESUMEN

GV1001, an anticancer vaccine, exhibits other biological functions, including anti-inflammatory and antioxidant activity. It also suppresses the development of ligature-induced periodontitis in mice. Porphyromonas gingivalis (Pg), a major human oral bacterium implicated in the development of periodontitis, is associated with various systemic disorders, such as atherosclerosis and Alzheimer's disease (AD). This study aimed to explore the protective effects of GV1001 against Pg-induced periodontal disease, atherosclerosis, and AD-like conditions in Apolipoprotein (ApoE)-deficient mice. GV1001 effectively mitigated the development of Pg-induced periodontal disease, atherosclerosis, and AD-like conditions by counteracting Pg-induced local and systemic inflammation, partly by inhibiting the accumulation of Pg DNA aggregates, Pg lipopolysaccharides (LPS), and gingipains in the gingival tissue, arterial wall, and brain. GV1001 attenuated the development of atherosclerosis by inhibiting vascular inflammation, lipid deposition in the arterial wall, endothelial to mesenchymal cell transition (EndMT), the expression of Cluster of Differentiation 47 (CD47) from arterial smooth muscle cells, and the formation of foam cells in mice with Pg-induced periodontal disease. GV1001 also suppressed the accumulation of AD biomarkers in the brains of mice with periodontal disease. Overall, these findings suggest that GV1001 holds promise as a preventive agent in the development of atherosclerosis and AD-like conditions associated with periodontal disease.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Enfermedades Periodontales , Porphyromonas gingivalis , Animales , Ratones , Apolipoproteínas E/deficiencia , Enfermedades Periodontales/microbiología , Enfermedades Periodontales/prevención & control , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Aterosclerosis/microbiología , Telomerasa/metabolismo , Fragmentos de Péptidos/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/microbiología , Periodontitis/microbiología , Periodontitis/prevención & control , Infecciones por Bacteroidaceae/microbiología , Infecciones por Bacteroidaceae/complicaciones , Infecciones por Bacteroidaceae/prevención & control , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Masculino , Humanos
4.
Int J Mol Sci ; 25(17)2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39273324

RESUMEN

Several lines of evidence have linked the intestinal bacterium Helicobacter cinaedi with the pathogenesis of atherosclerosis, identifying the Cinaedi Antigen Inflammatory Protein (CAIP) as a key virulence factor. Oxidative stress and inflammation are crucial in sustaining the atherosclerotic process and oxidized LDL (oxLDL) uptake. Primary human macrophages and endothelial cells were pre-incubated with 10 µM diphenyl iodonium salt (DPI) and stimulated with 20 µg/mL CAIP. Lectin-like oxLDL receptor (LOX-1) expression was evaluated by FACS analysis, reactive oxygen species (ROS) production was measured using the fluorescent probe H2DCF-DA, and cytokine release was quantified by ELISA assay. Foam cells formation was assessed by Oil Red-O staining, and phosphorylation of p38 and ERK1/2 MAP kinases and NF-κB pathway activation were determined by Western blot. This study demonstrated that CAIP triggered LOX-1 over-expression and increased ROS production in both macrophages and endothelial cells. Blocking ROS abrogated LOX-1 expression and reduced LDL uptake and foam cells formation. Additionally, CAIP-mediated pro-inflammatory cytokine release was significantly affected by ROS inhibition. The signaling pathway induced by CAIP-induced oxidative stress led to p38 MAP kinase phosphorylation and NF-κB activation. These findings elucidate the mechanism of action of CAIP, which heightens oxidative stress and contributes to the atherosclerotic process in H. cinaedi-infected patients.


Asunto(s)
Aterosclerosis , Infecciones por Helicobacter , Helicobacter , Lipoproteínas LDL , Macrófagos , Especies Reactivas de Oxígeno , Receptores Depuradores de Clase E , Humanos , Especies Reactivas de Oxígeno/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/microbiología , Aterosclerosis/patología , Macrófagos/metabolismo , Macrófagos/microbiología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Receptores Depuradores de Clase E/metabolismo , Lipoproteínas LDL/metabolismo , Helicobacter/patogenicidad , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , FN-kappa B/metabolismo , Células Espumosas/metabolismo , Citocinas/metabolismo , Estrés Oxidativo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Bacterianas/metabolismo , Sistema de Señalización de MAP Quinasas , Células Cultivadas , Transducción de Señal
5.
World J Microbiol Biotechnol ; 40(7): 198, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727952

RESUMEN

Atherosclerosis is viewed as not just as a problem of lipid build-up in blood vessels, but also as a chronic inflammatory disease involving both innate and acquired immunity. In atherosclerosis, the inflammation of the arterial walls is the key characteristic that significantly contributes to both the instability of plaque and the occlusion of arteries by blood clots. These events ultimately lead to stroke and acute coronary syndrome. Probiotics are living microorganisms that, when consumed in the right quantities, offer advantages for one's health. The primary objective of this study was to investigate the influence of Lactiplantibacillus plantarum ATCC 14917 (ATCC 14917) on the development of atherosclerotic plaques and its underlying mechanism in Apo lipoprotein E-knockout (Apoe-/- mice). In this study, Apoe-/- mice at approximately 8 weeks of age were randomly assigned to three groups: a Normal group that received a normal chow diet, a high fat diet group that received a gavage of PBS, and a Lactiplantibacillus plantarum ATCC 14917 group that received a high fat diet and a gavage of 0.2 ml ATCC 14917 (2 × 109 CFU/mL) per day for a duration of 12 weeks. Our strain effectively reduced the size of plaques in Apoe-/- mice by regulating the expression of inflammatory markers, immune cell markers, chemokines/chemokine receptors, and tight junction proteins (TJPs). Specifically, it decreased the levels of inflammatory markers (ICAM-1, CD-60 MCP-1, F4/80, ICAM-1, and VCAM-1) in the thoracic aorta, (Ccr7, cd11c, cd4, cd80, IL-1ß, TNF-α) in the colon, and increased the activity of ROS-scavenging enzymes (SOD-1 and SOD-2). It also influenced the expression of TJPs (occludin, ZO-1, claudin-3, and MUC-3). In addition, the treatment of ATCC 14917 significantly reduced the level of lipopolysaccharide in the mesenteric adipose tissue. The findings of our study demonstrated that our strain effectively decreased the size of atherosclerotic plaques by modulating inflammation, oxidative stress, intestinal integrity, and intestinal immunity.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Placa Aterosclerótica , Probióticos , Animales , Probióticos/administración & dosificación , Probióticos/farmacología , Ratones , Aterosclerosis/microbiología , Apolipoproteínas E/genética , Masculino , Modelos Animales de Enfermedad , Ratones Noqueados , Dieta Alta en Grasa , Lactobacillus plantarum , Citocinas/metabolismo , Ratones Endogámicos C57BL , Inflamación
6.
Circ Res ; 128(2): 287-303, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33476202

RESUMEN

Atherosclerotic cardiovascular disease (ASCVD) proceeds through a series of stages: initiation, progression (or regression), and complications. By integrating known biology regarding molecular signatures of each stage with recent advances in high-dimensional molecular data acquisition platforms (to assay the genome, epigenome, transcriptome, proteome, metabolome, and gut microbiome), snapshots of each phase of atherosclerotic cardiovascular disease development can be captured. In this review, we will summarize emerging approaches for assessment of atherosclerotic cardiovascular disease risk in humans using peripheral blood molecular signatures and molecular imaging approaches. We will then discuss the potential (and challenges) for these snapshots to be integrated into a personalized movie providing dynamic readouts of an individual's atherosclerotic cardiovascular disease risk status throughout the life course.


Asunto(s)
Aterosclerosis/metabolismo , Metaboloma , Proteoma , Transcriptoma , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/genética , Aterosclerosis/microbiología , Bacterias/metabolismo , Biomarcadores/metabolismo , Progresión de la Enfermedad , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Metabolómica , Imagen Molecular , Placa Aterosclerótica , Valor Predictivo de las Pruebas , Proteómica , Medición de Riesgo
7.
J Clin Periodontol ; 50(11): 1553-1567, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37621247

RESUMEN

AIM: The link between periodontitis and intestinal dysbiosis, two factors that contribute to atherosclerosis, has not been clearly defined. We investigated the integrative effects of oral infection with Porphyromonas gingivalis (PG), the major pathogen for periodontitis, on intestinal microbiota and atherosclerosis. MATERIALS AND METHODS: ApoE-/- mice were fed a normal chow diet (NC), a Western diet (WD) or a WD with oral PG infection (PG). The PG infection was investigated by placing a total of 109 CFUs of live PG into the oral cavity of each mouse using a feeding needle five times a week for 3 weeks. Atherosclerotic lesions of the aortae were measured, and blood lipoproteins and the expression of molecules related to lipid metabolism in the liver were analysed. We also performed 16S RNA sequencing and a microbiome analysis using faeces. RESULTS: En face bloc preparation of the aortae showed that the PG group had a 1.7-fold increase in atherosclerotic lesions compared with the WD group (p < .01). Serum analyses showed that oral PG infection induced a significant decrease in high-density lipoprotein (HDL) and triglyceride. Western blots of hepatic tissue lysates revealed that PG infection reduced the expression of scavenger receptor class B type 1 (SR-B1) in the liver by 50%. Faecal microbiota analysis revealed that species richness estimates (Chao1, ACE) decreased immediately after PG infection. PG infection also induced a significant decrease in Shannon diversity and an increase in Simpson's indices in the WD-fed mice. PG infection significantly increased the phyla Actinobacteria and Deferribacteres, along with the species Mucispirillum schaedleri and Lactobacillus gasseri, in the mice. The functional study showed that PG infection increased the expression of proteins that function in carbohydrate and glucose metabolism, including phosphotransferase system (PTS) proteins and the GntR family transcriptional regulator. CONCLUSIONS: Oral PG infection promotes atherosclerosis and induces significant metabolic changes, including reduced serum HDL and reduced hepatic SR-B1 and ABCA1 expression, as well as changes in intestinal microbiota. Our study suggests that intestinal dysbiosis accompanies periodontitis and could play a role in atherosclerosis.


Asunto(s)
Aterosclerosis , Microbioma Gastrointestinal , Periodontitis , Ratones , Animales , Porphyromonas gingivalis , Disbiosis , Aterosclerosis/microbiología
8.
Acta Microbiol Immunol Hung ; 70(4): 272-277, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-37975913

RESUMEN

Atherosclerosis is a progressive disease marked by the accumulation of lipids and fibrous components in the large arteries. It is one of the primary causes of heart disease and stroke. Periodontal diseases encompass conditions like gingivitis and periodontitis, which are multifactorial diseases associated with dysbiotic plaque biofilms that trigger an immune-inflammatory host response, eventually resulting in the destruction of periodontal tissues. Links between periodontal disease and atherosclerosis may be based on direct invasion of periodontal pathogens or inflammatory mechanisms triggered by bacteria related to periodontal lesions, locally or systemically, that may impact the initiation of the atherosclerotic lesion. The presence of periodontal pathogens within an atheromatous lesion implies hematogenous dissemination. The invasion of atheroma by periodontal pathogens results in changes in the proatherogenic and proinflammatory properties of endothelial cells, leading to endothelial dysfunction, which is a hallmark of atherosclerosis. Clinical and epidemiological studies have offered sufficient evidence of periodontitis having an adverse effect on systemic health, including atherosclerosis; however, a direct causal effect has not yet been proved. This review aims to analyse scientific results regarding the mechanism by which periodontal pathogens may cause atherosclerosis as well as to describe the role of Porphyromonas gingivalis in atherosclerotic plaque development and progression.


Asunto(s)
Aterosclerosis , Periodontitis , Placa Aterosclerótica , Humanos , Placa Aterosclerótica/complicaciones , Placa Aterosclerótica/microbiología , Placa Aterosclerótica/patología , Células Endoteliales/patología , Aterosclerosis/complicaciones , Aterosclerosis/microbiología , Aterosclerosis/patología , Periodontitis/complicaciones , Periodontitis/microbiología , Porphyromonas gingivalis
9.
Int J Mol Sci ; 24(4)2023 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-36834497

RESUMEN

Atherosclerosis is a major cause of death and disability. The beneficial effects of phytochemicals and probiotics on atherosclerosis have gained significant interest since these functional foods can improve inflammation, oxidative stress, and microbiome dysbiosis. The direct effect of the microbiome in atherosclerosis, however, needs further elucidation. The objective of this work was to investigate the effects of polyphenols, alkaloids, and probiotics on atherosclerosis using a meta-analysis of studies with mouse models of atherosclerosis. Identification of eligible studies was conducted through searches on PubMed, Embase, Web of Science, and Science Direct until November 2022. The results showed that phytochemicals reduced atherosclerosis, which was significant in male mice, but not in females. Probiotics, on the other hand, showed significant reductions in plaque in both sexes. Berries and phytochemicals modulated gut microbial composition by reducing the Firmicutes/Bacteroidetes (F/B) ratio and by upregulating health-promoting bacteria, including Akkermansia muciniphila. This analysis suggests that phytochemicals and probiotics can reduce atherosclerosis in animal models, with a potentially greater effect on male animals. Thus, consumption of functional foods rich in phytochemicals as well as probiotics are viable interventions to improve gut health and reduce plaque burden in patients suffering from cardiovascular disease (CVD).


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Microbioma Gastrointestinal , Placa Aterosclerótica , Probióticos , Femenino , Masculino , Animales , Ratones , Frutas , Aterosclerosis/microbiología , Fitoquímicos/farmacología , Verrucomicrobia , Disbiosis/microbiología
10.
Semin Cancer Biol ; 70: 85-97, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32610150

RESUMEN

Human gut microbiota is a dynamic and variable system that can change over time and in response to different diets and treatments. There is currently no doubt that gut microbiota can provide interesting therapeutic opportunities, since it can metabolize biologically active molecules, drugs, and their precursors, and control their bioavailability. Moreover, it can produce both beneficial and dangerous metabolites that influence host's health. In this review, we summarize the current knowledge on the involvement of gut microbiota in two chronic human pathologies that represent the greatest challenges of modern medicine: atherosclerosis and cancer. Interesting parallels are observed between the mechanisms and possible treatment approaches of these pathologies. Some of the common effects of therapeutic agents targeting both pathologies, such as anti-inflammatory activity, are partially mediated by the gut microbiota. We will discuss the effects of common drugs (metformin, statins and aspirin) and various nutraceuticals on gut microbiota and outline the pathways of microbial involvement in mediating the pleiotropic beneficial effects of these agents in atherosclerosis and cancer.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Microbioma Gastrointestinal , Neoplasias/tratamiento farmacológico , Prebióticos/administración & dosificación , Animales , Aterosclerosis/microbiología , Humanos , Neoplasias/microbiología
11.
FASEB J ; 35(11): e21942, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34670018

RESUMEN

Atherosclerosis is a chronic inflammatory disease. Pathophysiological similarities between chronic infections and atherosclerosis triggered interests between these conditions. The seroepidemiological study showed that Helicobacter pylori strains that express cytotoxin-associated gene A (CagA), an oncoprotein and a major virulence factor, was positively correlated with atherosclerosis and related clinical events. Nevertheless, the underlying mechanism is poorly understood. In this study, the seroprevalence of infection by H. pylori and by strains express CagA assessed by enzyme-linked immunosorbent assay (ELISA) showed that the prevalence of CagA strains rather than H. pylori in patients was positively correlated with atherogenesis. Correspondingly, we found that CagA augmented the growth of plaque of ApoE-/- mice in the early stage of atherosclerosis and promoted the expression of adhesion molecules and inflammatory cytokines in mouse aortic endothelial cells (MAECs). Mechanistically, both si-NLRP3 and si-IL-1ß mitigated the promoting effect of CagA on the inflammatory activation of HAECs. In vivo, the inhibition of NLRP3 by MCC950 significantly attenuated the promoting effect of CagA on plaque growth of ApoE-/- mice. We also propose NLRP3 as a potential therapeutic target for CagA-positive H. pylori infection-related atherosclerosis and emphasize the importance of inflammation in atherosclerosis pathology.


Asunto(s)
Antígenos Bacterianos/metabolismo , Aorta/patología , Aterosclerosis/sangre , Proteínas Bacterianas/metabolismo , Caspasa 1/metabolismo , Células Endoteliales/metabolismo , Infecciones por Helicobacter/sangre , Helicobacter pylori/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Placa Aterosclerótica/sangre , Anciano , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Aorta/metabolismo , Aterosclerosis/microbiología , Proteínas Bacterianas/inmunología , Modelos Animales de Enfermedad , Femenino , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Persona de Mediana Edad , Placa Aterosclerótica/microbiología , Estudios Seroepidemiológicos , Células THP-1
12.
Circ Res ; 126(6): e15-e29, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32078488

RESUMEN

RATIONALE: Atherosclerotic cardiovascular diseases are the leading cause of mortality worldwide. Atherosclerotic cardiovascular diseases are considered as chronic inflammation processes. In addition to risk factors associated with the cardiovascular system itself, pathogenic bacteria such as the periodontitis-associated Porphyromonas gingivalis (P gingivalis) are also closely correlated with the development of atherosclerosis, but the underlying mechanisms are still elusive. OBJECTIVE: To elucidate the mechanisms of P gingivalis-accelerated atherosclerosis and explore novel therapeutic strategies of atherosclerotic cardiovascular diseases. METHODS AND RESULTS: Bmal1-/- (brain and muscle Arnt-like protein 1) mice, ApoE-/- mice, Bmal1-/-ApoE-/- mice, conditional endothelial cell Bmal1 knockout mice (Bmal1fl/fl; Tek-Cre mice), and the corresponding jet-legged mouse model were used. Pgingivalis accelerates atherosclerosis progression by triggering arterial oxidative stress and inflammatory responses in ApoE-/- mice, accompanied by the perturbed circadian clock. Circadian clock disruption boosts P gingivalis-induced atherosclerosis progression. The mechanistic dissection shows that P gingivalis infection activates the TLRs-NF-κB signaling axis, which subsequently recruits DNMT-1 to methylate the BMAL1 promoter and thus suppresses BMAL1 transcription. The downregulation of BMAL1 releases CLOCK, which phosphorylates p65 and further enhances NF-κB signaling, elevating oxidative stress and inflammatory response in human aortic endothelial cells. Besides, the mouse model exhibits that joint administration of metronidazole and melatonin serves as an effective strategy for treating atherosclerotic cardiovascular diseases. CONCLUSIONS: P gingivalis accelerates atherosclerosis via the NF-κB-BMAL1-NF-κB signaling loop. Melatonin and metronidazole are promising auxiliary medications toward atherosclerotic cardiovascular diseases.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Aterosclerosis/metabolismo , Infecciones por Bacteroidaceae/complicaciones , Estrés Oxidativo , Factores de Transcripción ARNTL/genética , Animales , Antibacterianos/uso terapéutico , Antioxidantes/uso terapéutico , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/etiología , Aterosclerosis/microbiología , Proteínas CLOCK/metabolismo , Ritmo Circadiano , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Regulación hacia Abajo , Endotelio Vascular/metabolismo , Femenino , Masculino , Melatonina/uso terapéutico , Metronidazol/uso terapéutico , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Porphyromonas gingivalis/patogenicidad , Transducción de Señal , Receptores Toll-Like/metabolismo
13.
Helicobacter ; 27(2): e12877, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35099837

RESUMEN

BACKGROUND: The role of H. pylori infection has been reported in various extragastric diseases, particularly, the correlation between H. pylori and atherosclerosis (AS) have received lots of attention. Some scholars demonstrated that the presence of H. pylori-specific DNA in the sclerotic plaques of atheromatous patients provides biological evidences, with indicating that H. pylori infection is a potential factor of AS. However, the underlying mechanism of H. pylori or their products cross the epithelial barriers to enter the blood circulation remains unclear. Recent studies have shown that the extracellular vesicles (EVs) derived from H. pylori-infected gastric epithelial cells encapsulated H. pylori virulence factor cytotoxin-associated gene A (CagA) and existed in the blood samples of patients or mice, which indicating that they can carry CagA into the blood circulation. Based on these findings, some researchers proposed a hypothesis that H. pylori is involved in the pathogenesis of AS via EVs-based mechanisms. In addition, outer membrane vesicles (OMVs) serve as transport vehicles to deliver H. pylori virulence factors to epithelial cells. It is necessary to discuss the role of H. pylori OMVs in the development of AS. OBJECTIVES: This review will focus on the correlation between H. pylori infection and AS and tried to unveil the possible role of EVs from H. pylori-infected cells and H. pylori OMVs in the pathogenesis of AS, with a view to providing help in refining our knowledge in this aspect. METHODS: All of information included in this review was retrieved from published studies on H. pylori infection in AS. RESULTS: H. pylori infection may be an atherosclerotic risk factor and drives researchers to reevaluate the role of H. pylori in the pathogenesis of AS. Some findings proposed a new hypothesis that H. pylori may be involved in the pathogenesis of AS through EVs-based mechanisms. Besides EVs from H. pylori-infected cells, whether H. pylori OMVs may play some role in the pathogenesis of AS is still remain unclear. CONCLUSION: Existing epidemiological and clinical evidence had shown that there is a possible association between H. pylori and AS. However, except for the larger randomized controlled trials, more basic research about EVs from H. pylori-infected cells and H. pylori OMVs is the need of the hour to unveil the possible role of H. pylori infection in the pathogenesis of AS.


Asunto(s)
Aterosclerosis , Vesículas Extracelulares , Infecciones por Helicobacter , Helicobacter pylori , Animales , Aterosclerosis/complicaciones , Aterosclerosis/microbiología , Proteínas Bacterianas/metabolismo , Vesículas Extracelulares/microbiología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/patología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Humanos , Ratones , Factores de Virulencia/metabolismo
14.
Biochem Biophys Res Commun ; 556: 192-198, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33845309

RESUMEN

Helicobacter pylori (H. pylori) infection mainly causes gastroduodenal diseases, including chronic gastritis, peptic ulcer disease and gastric cancer. In recent years, several studies have demonstrated that infection with H. pylori, especially strains harboring the virulence factor CagA (cytotoxin-associated gene A), contribute to the development of non-gastric systemic diseases, including hypercholesterolemia and atherosclerotic cardiovascular diseases. However, mechanisms underlying this association has not been defined. In this study, we carried out a large-scale genetic screen using Drosophila and identified a novel CagA target low-density lipoprotein receptor (LDLR), which aids in the clearance of circulating LDL. We showed that CagA physically interacted with LDLR via its carboxy-terminal region and inhibited LDLR-mediated LDL uptake into cells. Since deficiency of LDLR-mediated LDL uptake has been known to increase plasma LDL and accelerate atherosclerosis, our findings may provide a novel mechanism for the association between infection with CagA-positive H. pylori and hypercholesterolemia leading to atherosclerotic cardiovascular diseases.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Helicobacter pylori/patogenicidad , Lipoproteínas LDL/metabolismo , Receptores de LDL/metabolismo , Factores de Virulencia/metabolismo , Animales , Animales Modificados Genéticamente , Aterosclerosis/microbiología , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ojo/metabolismo , Femenino , Humanos , Hipercolesterolemia/microbiología , Lipoproteínas LDL/sangre , Masculino , Unión Proteica
15.
Rheumatology (Oxford) ; 61(1): 258-269, 2021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33769467

RESUMEN

OBJECTIVES: An altered microbiota, which can be described quantitatively, has been identified as playing a pivotal role in host vascular physiology, and it may contribute to various diseases. The aim of this study was to better understand the role of the gut microbiota in vascular physiology in a subclinical elderly population, and to investigate how lifestyle affects the composition of host gut microbiota to further impact the pathogenesis of vascular diseases. METHODS: We performed a population-based faecal metagenomic study over 569 elderly asymptomatic subclinical individuals in rural China. An association network was built based on clinical measurements and detailed epidemiologic questionnaires, including blood chemistry, arterial stiffness, carotid ultrasonography, and metagenomic datasets. RESULTS: By analyzing the breadth, depth and impact of each node of the association network, we found carotid arterial atherosclerosis indices, including intima-media thickness (IMT), were essential in the network, and were significantly associated with living habits, socio-economic status, and diet. Using mediation analysis, we found that higher frequency of eating fresh fruits and vegetables, and more exercise significantly reduced carotid atherosclerosis in terms of IMT, peak systolic velocity and end-diastolic velocity values through the mediation of Alistepes, Oligella and Prevotella. Gut microbes explained 16.5% of the mediation effect of lifestyle on the pathogenesis of carotid atherosclerosis. After adjustment, Faecalicatena [odds ratio (OR) = 0.12 ∼0.65] was shown to be protective against the formation of carotid atherosclerosis, independently, while Libanicoccus (OR = 1.46 ∼4.20 ) was associated with increased carotid arterial IMT. KEGG/KO Kyoto Encyclopedia of Genes and Genomes/ KEGG Orthology (KEGG/KO) analyses revealed a loss of anti-inflammation function in IMT subjects. CONCLUSION: Our study revealed a Chinese population-wide phenotype-metagenomic association network and a mediation effect of gut microbiota on carotid artery atherosclerosis, hinting at potential therapeutic and preventive uses for microbiota in vascular diseases.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/microbiología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Femenino , Genómica , Estilo de Vida Saludable , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
16.
FASEB J ; 34(11): 14166-14181, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32939880

RESUMEN

The gut microbiota is associated with cardiovascular diseases, including atherosclerosis. However, the composition, functional capacity, and metabolites of the gut microbiome about atherosclerosis have not been comprehensively studied. Here, we reanalyzed 25 metagenomic stool samples from Sweden and 385 metagenomic stool samples from China using HUMAnN2, PanPhlAn, and MelonnPan to obtain more sufficient information. We found that the samples from atherosclerotic patients in both cohorts were depleted in Bacteroides xylanisolvens, Odoribacter splanchnicus, Eubacterium eligens, Roseburia inulinivorans, and Roseburia intestinalis. At the functional level, healthy metagenomes were both enriched in pathways of starch degradation V, glycolysis III (from glucose), CDP-diacylglycerol biosynthesis, and folate transformations. R inulinivorans and R intestinalis are major contributors to starch degradation V, while E eligens greatly contribute to the pathway CDP-diacylglycerol biosynthesis, and B xylanisolvens and B uniformis contribute to folate transformations II. The 11 marker species selected from the Chinese cohort distinguish patients from controls with an area under the receiver operating characteristics curve (AUC) of 0.86. Strain-level microbial analysis revealed a geographically associated adaptation of the strains from E eligens, B uniformis, and E coli. Two gut microbial metabolites, nicotinic acid and hydrocinnamic acid, had significantly higher predicted abundance in the control samples compared to the patients in the Chinese cohort, and interestinglynicotinic acid is already an effective lipid-lowering drug to reducing cardiovascular risk. Our results indicate intestinal bacteria such as B xylanisolvens, E eligens, and R inulinivorans could be promising probiotics and potential therapeutic target for atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/microbiología , Bacterias/genética , Heces/microbiología , Microbioma Gastrointestinal/genética , Metaboloma , Metagenoma , Anciano , Aterosclerosis/epidemiología , Aterosclerosis/metabolismo , Bacterias/clasificación , Bacterias/aislamiento & purificación , Estudios de Casos y Controles , China/epidemiología , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Suecia/epidemiología
17.
Circ Res ; 124(1): 94-100, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30582442

RESUMEN

RATIONALE: Several studies have suggested a role for the gut microbiota in inflammation and atherogenesis. A causal relation relationship between gut microbiota, inflammation, and atherosclerosis has not been explored previously. OBJECTIVE: Here, we investigated whether a proinflammatory microbiota from Caspase1-/- ( Casp1-/-) mice accelerates atherogenesis in Ldlr-/- mice. METHOD AND RESULTS: We treated female Ldlr-/- mice with antibiotics and subsequently transplanted them with fecal microbiota from Casp1-/- mice based on a cohousing approach. Autologous transplantation of fecal microbiota of Ldlr-/- mice served as control. Mice were cohoused for 8 or 13 weeks and fed chow or high-fat cholesterol-rich diet. Fecal samples were collected, and factors related to inflammation, metabolism, intestinal health, and atherosclerotic phenotypes were measured. Unweighted Unifrac distances of 16S rDNA (ribosomal DNA) sequences confirmed the introduction of the Casp1-/- and Ldlr-/- microbiota into Ldlr-/- mice (referred to as Ldlr-/-( Casp1-/-) or Ldlr-/-( Ldlr-/-) mice). Analysis of atherosclerotic lesion size in the aortic root demonstrated a significant 29% increase in plaque size in 13-week high-fat cholesterol-fed Ldlr-/-( Casp1-/-) mice compared with Ldlr-/-( Ldlr-/-) mice. We found increased numbers of circulating monocytes and neutrophils and elevated proinflammatory cytokine levels in plasma in high-fat cholesterol-fed Ldlr-/-( Casp1-/-) compared with Ldlr-/-( Ldlr-/-) mice. Neutrophil accumulation in the aortic root of Ldlr-/-( Casp1-/-) mice was enhanced compared with Ldlr-/-( Ldlr-/-) mice. 16S-rDNA-encoding sequence analysis in feces identified a significant reduction in the short-chain fatty acid-producing taxonomies Akkermansia, Christensenellaceae, Clostridium, and Odoribacter in Ldlr-/-( Casp1-/-) mice. Consistent with these findings, cumulative concentrations of the anti-inflammatory short-chain fatty acids propionate, acetate and butyrate in the cecum were significantly reduced in 13-week high-fat cholesterol-fed Ldlr-/-( Casp1-/-) compared with Ldlr-/-( Ldlr-/-) mice. CONCLUSIONS: Introduction of the proinflammatory Casp1-/- microbiota into Ldlr-/- mice enhances systemic inflammation and accelerates atherogenesis.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/microbiología , Aterosclerosis/microbiología , Bacterias/metabolismo , Citocinas/metabolismo , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Mediadores de Inflamación/metabolismo , Inflamación/microbiología , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Caspasa 1/genética , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Disbiosis , Ácidos Grasos/metabolismo , Femenino , Interacciones Huésped-Patógeno , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones Noqueados , Placa Aterosclerótica , Receptores de LDL/genética , Receptores de LDL/metabolismo , Factores de Tiempo
18.
Pharmacol Res ; 163: 105362, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33285231

RESUMEN

Gut microbiota (GM) has emerged as an essential and integral factor for maintaining human health and affecting pathological outcomes. Metagenomics and metabolomics characterization have furthered gut metagenome's understanding and unveiled that deviation of specific GM community members and GM-dependent metabolites imbalance orchestrate metabolic or cardiovascular diseases (CVDs). Restoring GM ecosystem with nutraceutical supplements keenly prebiotics and probiotics relatively decreases CVDs incidence and overall mortality. In Atherosclerosis, commensal and pathogenic gut microbes correlate with atherogenesis events. GM-dependent metabolites-trimethylamine N-oxide and short-chain fatty acids regulate atherosclerosis-related metabolic processes in opposite patterns to affect atherosclerosis outcomes. Therefore, GM might be a potential therapeutic target for atherosclerosis. In atherogenic animal models, natural products with cardioprotective properties could modulate the GM ecosystem by revitalizing healthier GM phylotypes and abrogating proatherogenic metabolites, paving future research paths for clinical therapeutics.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Productos Biológicos/uso terapéutico , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Aterosclerosis/microbiología , Humanos
19.
Cell Mol Life Sci ; 77(14): 2751-2769, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32002588

RESUMEN

Atherosclerotic vascular disease (ASVD) is a chronic process, with a progressive course over many years, but it can cause acute clinical events, including acute coronary syndromes (ACS), myocardial infarction (MI) and stroke. In addition to a series of typical risk factors for atherosclerosis, like hyperlipidemia, hypertension, smoking and obesity, emerging evidence suggests that atherosclerosis is a chronic inflammatory disease, suggesting that chronic infection plays an important role in the development of atherosclerosis. Toll-like receptors (TLRs) are the most characteristic members of pattern recognition receptors (PRRs), which play an important role in innate immune mechanism. TLRs play different roles in different stages of infection of atherosclerosis-related pathogens such as Chlamydia pneumoniae (C. pneumoniae), periodontal pathogens including Porphyromonas gingivalis (P. gingivalis), Helicobacter pylori (H. pylori) and human immunodeficiency virus (HIV). Overall, activation of TLR2 and 4 seems to have a profound impact on infection-related atherosclerosis. This article reviews the role of TLRs in the process of atherosclerosis after C. pneumoniae and other infections and the current status of treatment, with a view to providing a new direction and potential therapeutic targets for the study of ASVD.


Asunto(s)
Aterosclerosis/genética , Infecciones Bacterianas/genética , Infecciones por VIH/genética , Receptores Toll-Like/genética , Aterosclerosis/complicaciones , Aterosclerosis/microbiología , Aterosclerosis/virología , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/virología , Chlamydophila pneumoniae/patogenicidad , VIH/patogenicidad , Infecciones por VIH/complicaciones , Infecciones por VIH/microbiología , Infecciones por VIH/virología , Helicobacter pylori/patogenicidad , Humanos , Porphyromonas gingivalis/patogenicidad
20.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34360839

RESUMEN

Hypercholesterolemia plays a causal role in the development of atherosclerosis and is one of the main risk factors for cardiovascular disease (CVD), the leading cause of death worldwide especially in developed countries. Current data show that the role of microbiota extends beyond digestion by being implicated in several metabolic and inflammatory processes linked to several diseases including CVD. Studies have reported associations between bacterial metabolites and hypercholesterolemia. However, such associations remain poorly investigated and characterized. In this review, the mechanisms of microbial derived metabolites such as primary and secondary bile acids (BAs), trimethylamine N-oxide (TMAO), and short-chain fatty acids (SCFAs) will be explored in the context of cholesterol metabolism. These metabolites play critical roles in maintaining cardiovascular health and if dysregulated can potentially contribute to CVD. They can be modulated via nutritional and pharmacological interventions such as statins, prebiotics, and probiotics. However, the mechanisms behind these interactions also remain unclear, and mechanistic insights into their impact will be provided. Therefore, the objectives of this paper are to present current knowledge on potential mechanisms whereby microbial metabolites regulate cholesterol homeostasis and to discuss the feasibility of modulating intestinal microbes and metabolites as a novel therapeutic for hypercholesterolemia.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Microbioma Gastrointestinal/fisiología , Animales , Aterosclerosis/microbiología , Humanos , Hipercolesterolemia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA