Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Cell ; 182(2): 447-462.e14, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32758418

RESUMO

The precise mechanism by which oral infection contributes to the pathogenesis of extra-oral diseases remains unclear. Here, we report that periodontal inflammation exacerbates gut inflammation in vivo. Periodontitis leads to expansion of oral pathobionts, including Klebsiella and Enterobacter species, in the oral cavity. Amassed oral pathobionts are ingested and translocate to the gut, where they activate the inflammasome in colonic mononuclear phagocytes, triggering inflammation. In parallel, periodontitis results in generation of oral pathobiont-reactive Th17 cells in the oral cavity. Oral pathobiont-reactive Th17 cells are imprinted with gut tropism and migrate to the inflamed gut. When in the gut, Th17 cells of oral origin can be activated by translocated oral pathobionts and cause development of colitis, but they are not activated by gut-resident microbes. Thus, oral inflammation, such as periodontitis, exacerbates gut inflammation by supplying the gut with both colitogenic pathobionts and pathogenic T cells.


Assuntos
Colite/patologia , Enterobacter/fisiologia , Microbioma Gastrointestinal , Klebsiella/fisiologia , Boca/microbiologia , Animais , Colite/microbiologia , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Enterobacter/isolamento & purificação , Feminino , Inflamassomos/metabolismo , Interleucina-10/deficiência , Interleucina-10/genética , Interleucina-1beta/metabolismo , Klebsiella/isolamento & purificação , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Periodontite/microbiologia , Periodontite/patologia , Células Th17/citologia , Células Th17/imunologia , Células Th17/metabolismo
2.
Infect Immun ; 92(5): e0000624, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38629806

RESUMO

Enterococci are common commensal bacteria that colonize the gastrointestinal tracts of most mammals, including humans. Importantly, these bacteria are one of the leading causes of nosocomial infections. This study examined the role of colonic macrophages in facilitating Enterococcus faecalis infections in mice. We determined that depletion of colonic phagocytes resulted in the reduction of E. faecalis dissemination to the gut-draining mesenteric lymph nodes. Furthermore, we established that trafficking of monocyte-derived CX3CR1-expressing macrophages contributed to E. faecalis dissemination in a manner that was not reliant on CCR7, the conventional receptor involved in lymphatic migration. Finally, we showed that E. faecalis mutants with impaired intracellular survival exhibited reduced dissemination, suggesting that E. faecalis can exploit host immune cell migration to disseminate systemically and cause disease. Our findings indicate that modulation of macrophage trafficking in the context of antibiotic therapy could serve as a novel approach for preventing or treating opportunistic infections by disseminating enteric pathobionts like E. faecalis.


Assuntos
Receptor 1 de Quimiocina CX3C , Colo , Enterococcus faecalis , Macrófagos , Receptores CCR2 , Receptores de Quimiocinas , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Receptor 1 de Quimiocina CX3C/genética , Macrófagos/microbiologia , Macrófagos/imunologia , Camundongos , Colo/microbiologia , Colo/imunologia , Receptores CCR2/metabolismo , Receptores CCR2/genética , Receptores de Quimiocinas/metabolismo , Receptores de Quimiocinas/genética , Infecções por Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/microbiologia , Camundongos Endogâmicos C57BL , Linfonodos/microbiologia , Linfonodos/imunologia , Receptores CCR7/metabolismo , Receptores CCR7/genética
3.
Mol Med ; 30(1): 122, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39135000

RESUMO

BACKGROUND: Current therapy for patients suffering from inflammatory bowel diseases (IBD) is focused on inflammatory mechanisms exclusively and not the dysbiotic microbiota, despite growing evidence implicating a role for intestinal microbes in disease. MAIN BODY: Ongoing research into the intestinal microbiota of IBD patients, using new technologies and/or deeper application of existing ones, has identified a number of microorganisms whose properties and behaviors warrant consideration as causative factors in disease. Such studies have implicated both bacteria and fungi in the pathogenesis of disease. Some of these organisms manifest mechanisms that should be amenable to therapeutic intervention via either conventional or novel drug discovery platforms. Of particular note is a deeper characterization of microbial derived proteases and their destructive potential. CONCLUSION: Given the steady progress on the mechanistic role of the microbiota in inflammatory diseases, it is reasonable to anticipate a future in which therapeutics targeting microbial derived pathogenic factors play an important role in improving the lives of IBD patients.


Assuntos
Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , Humanos , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Animais , Bactérias/efeitos dos fármacos , Bactérias/metabolismo
4.
Microb Pathog ; 193: 106726, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38848931

RESUMO

Gut bacterial dysbiosis has been linked to several gastrointestinal diseases, including deadly colorectal cancer (CRC), a leading cause of mortality in cancer patients. However, perturbation in gut bacteriome during colon cancer (CC, devoid of colorectal malignancy) remains poorly explored. Here, 16S rRNA gene amplicon sequencing was carried out for fecal DNA samples targeted to hypervariable V3-V4 region by employing MiSeq platform to explore the gut bacterial community shift in CC patients. While alpha diversity indices predicted high species richness and diversity, beta diversity showed marked gut bacterial compositional dissimilarity in CC versus healthy controls (HC, n = 10 each). We observed a significant (p < 0.05, Wilcoxon Rank-Sum test) emergence of low-abundant anaerobic taxa, including Parvimonas and Peptostreptococcus, in addition to Subdoligranulum, Coprococcus, Holdemanella, Solobacterium, Bilophila, Blautia, Dorea, Moryella and several unidentified taxa, mainly affiliated to Firmicutes, in CC patients. In addition, we also traced the emergence of putative probiotic taxon Slackia, belonging to Actinomycetota, in CC patients. The emergence of anaerobic Firmicutes in CC is accompanied by a significant (p < 0.05) decline in the Klebsiella, as determined through linear discriminant analysis effect size (LEfSe) and heat tree analyses. Shifts in core microbiome and variation in network correlation were also witnessed. Taken together, this study highlighted a significant and consistent emergence of rare anaerobic Firmicutes suggesting possible anaerobiosis driving gut microbial community shift, which could be exploited in designing diagnostic and therapeutic tools targeted to CC.


Assuntos
Neoplasias do Colo , Disbiose , Fezes , Firmicutes , Microbioma Gastrointestinal , Klebsiella , RNA Ribossômico 16S , Humanos , Microbioma Gastrointestinal/genética , RNA Ribossômico 16S/genética , Neoplasias do Colo/microbiologia , Klebsiella/genética , Klebsiella/isolamento & purificação , Klebsiella/classificação , Fezes/microbiologia , Firmicutes/genética , Firmicutes/isolamento & purificação , Firmicutes/classificação , Disbiose/microbiologia , Masculino , Feminino , DNA Bacteriano/genética , Pessoa de Meia-Idade , Idoso , Filogenia , Anaerobiose
5.
Gastroenterology ; 162(5): 1409-1423.e1, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34998802

RESUMO

Current noninvasive methods for colorectal cancer (CRC) screening are not optimized for persons with inflammatory bowel diseases (IBDs), requiring patients to undergo frequent interval screening via colonoscopy. Although colonoscopy-based screening reduces CRC incidence in IBD patients, rates of interval CRC remain relatively high, highlighting the need for more targeted approaches. In recent years, the discovery of disease-specific microbiome signatures for both IBD and CRC has begun to emerge, suggesting that stool-based biomarker detection using metagenomics and other culture-independent technologies may be useful for personalized, early, noninvasive CRC screening in IBD patients. Here we discuss the utility of the stool microbiome as a noninvasive CRC screening tool. Comparing the performance of multiple microbiome-based CRC classifiers, including several multi-cohort meta-analyses, we find that noninvasive detection of colorectal adenomas and carcinomas from microbial biomarkers is an active area of study with promising early results.


Assuntos
Neoplasias Colorretais , Doenças Inflamatórias Intestinais , Microbiota , Colonoscopia , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/epidemiologia , Detecção Precoce de Câncer/métodos , Humanos , Doenças Inflamatórias Intestinais/diagnóstico
6.
Gastroenterology ; 163(4): 1038-1052, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35788347

RESUMO

BACKGROUND & AIMS: Medication is a major determinant of human gut microbiome structure, and its overuse increases the risks of morbidity and mortality. However, effects of certain commonly prescribed drugs and multiple medications on the gut microbiome are still underinvestigated. METHODS: We performed shotgun metagenomic analysis of fecal samples from 4198 individuals in the Japanese 4D (Disease, Drug, Diet, Daily life) microbiome project. A total of 759 drugs were profiled, and other metadata, such as anthropometrics, lifestyles, diets, physical activities, and diseases, were prospectively collected. Second fecal samples were collected from 243 individuals to assess the effects of drug initiation and discontinuation on the microbiome. RESULTS: We found that numerous drugs across different treatment categories influence the microbiome; more than 70% of the drugs we profiled had not been examined before. Individuals exposed to multiple drugs, polypharmacy, showed distinct gut microbiome structures harboring significantly more abundant upper gastrointestinal species and several nosocomial pathobionts due to additive drug effects. Polypharmacy was also associated with microbial functions, including the reduction of short-chain fatty acid metabolism and increased bacterial stress responses. Even nonantibiotic drugs were significantly correlated with an increased antimicrobial resistance potential through polypharmacy. Notably, a 2-time points dataset revealed the alteration and recovery of the microbiome in response to drug initiation and cessation, corroborating the observed drug-microbe associations in the cross-sectional cohort. CONCLUSION: Our large-scale metagenomics unravels extensive and disruptive impacts of individual and multiple drug exposures on the human gut microbiome, providing a drug-microbe catalog as a basis for a deeper understanding of the role of the microbiome in drug efficacy and toxicity.


Assuntos
Anti-Infecciosos , Microbioma Gastrointestinal , Microbiota , Estudos Transversais , Ácidos Graxos Voláteis/farmacologia , Fezes/microbiologia , Microbioma Gastrointestinal/fisiologia , Humanos , Metagenômica
7.
Mycopathologia ; 188(5): 821-823, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37589873

RESUMO

Kazachstania pintolopesii is an opportunistic mammalian pathobiont from the K. telluris species complex. No draft genomes of this species are currently available. Here, we report the first draft genome sequence of a primate isolate of K. pintolopesii (NCYC 4417).


Assuntos
Saccharomycetales , Animais , Saccharomycetales/genética , Primatas/genética , Genoma , Mamíferos/genética
8.
Periodontol 2000 ; 89(1): 166-180, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35244982

RESUMO

Accumulating evidence demonstrates that the oral pathobiont Fusobacterium nucleatum is involved in the progression of an increasing number of tumors types. Thus far, the mechanisms underlying tumor exacerbation by F. nucleatum include the enhancement of proliferation, establishment of a tumor-promoting immune environment, induction of chemoresistance, and the activation of immune checkpoints. This review focuses on the mechanisms that mediate tumor-specific colonization by fusobacteria. Elucidating the mechanisms mediating fusobacterial tumor tropism and promotion might provide new insights for the development of novel approaches for tumor detection and treatment.


Assuntos
Infecções por Fusobacterium , Neoplasias , Infecções por Fusobacterium/microbiologia , Fusobacterium nucleatum/fisiologia , Humanos
9.
J Invertebr Pathol ; 194: 107818, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35973510

RESUMO

Insect guts often harbor an abundance of bacteria. Many of these members are commensal, but some may emerge as opportunistic pathogens when the host is under stress. In this study, we evaluated how dietary nutritional concentration mediates a shift from commensal to pathogenic, and if host species influences those interactions. We used the lepidopterans (Noctuidae) fall armyworm (Spodoptera frugiperda), beet armyworm (Spodoptera exigua), and corn earworm (Helicoverpa zea) as hosts and a Serratia strain initially isolated from healthy fall armyworm. Diet concentration was altered by bulk reduction in nutritional content with dilution using cellulose. Our experiments revealed that low nutrient diet increased mortality from Serratia for beet armyworm and corn earworm. However, for fall armyworm, little mortality was observed in any of the diet combinations. Dietary nutrition and oral inoculation with Serratia did not change the expression of two antimicrobial peptides in fall and beet armyworm, suggesting that other mechanisms that mediate mortality were involved. Our results have implications for how pathogens may persist as commensals in the digestive tract of insects. These findings also suggest that diet plays a very important role in the switch from commensal to pathogen. Finally, our data indicate that the host response to changing conditions is critical in determining if a pathogen may overtake its host and that these three lepidopteran species have different responses to opportunistic enteric pathogens.


Assuntos
Mariposas , Serratia , Animais , Celulose , Dieta , Larva/fisiologia , Spodoptera/microbiologia , Zea mays
10.
Anaerobe ; 75: 102582, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35533828

RESUMO

Desulfovibrio spp. is a commensal sulfate reducing bacterium that is present in small numbers in the gastrointestinal tract. Increased concentrations of Desulfovibrio spp. (blooms) have been reported in patients with inflammatory bowel disease and irritable bowel syndrome. Since stress has been reported to exacerbate symptoms of these chronic diseases, this study examined whether the stress catecholamine norepinephrine (NE) promotes Desulfovibrio growth. Norepinephrine-stimulated growth has been reported in other bacterial taxa, and this effect may depend on the availability of the micronutrient iron. OBJECTIVES: This study tested whether norepinephrine exposure affects the in vitro growth of Desulfovibrio vulgaris in an iron dependent manner. METHODS: DSV was incubated in a growth medium with and without 1 µm of norepinephrine. An additional growth assay added the iron chelator deferoxamine in NE exposed DSV. Iron regulatory genes were assessed with and without the treatment of NE and Deferoxamine. RESULTS: We found that norepinephrine significantly increased growth of D. vulgaris. Norepinephrine also increased bacterial production of hydrogen sulfide. Additionally, norepinephrine significantly increased bacterial expression in three of the four tested iron regulatory genes. The iron chelator deferoxamine inhibited growth of D. vulgaris in a dose-dependent manner and reversed the effect of norepinephrine on proliferation of D. vulgaris and on bacterial expression of iron regulatory genes. CONCLUSION: The data presented in this work suggests that promotion of D. vulgaris growth by norepinephrine is iron dependent.


Assuntos
Desulfovibrio vulgaris , Desulfovibrio , Desferroxamina/metabolismo , Desferroxamina/farmacologia , Desulfovibrio/metabolismo , Desulfovibrio vulgaris/genética , Humanos , Ferro/metabolismo , Quelantes de Ferro/metabolismo , Quelantes de Ferro/farmacologia , Norepinefrina/metabolismo , Norepinefrina/farmacologia
11.
Int J Med Microbiol ; 311(2): 151472, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33461110

RESUMO

Numerous studies have associated alterations in the gut microbiota composition with almost every known inflammatory disease. However, proving the biological relevance of distinct microbial signatures and linking specific microorganisms to host phenotypes, remains a considerable challenge. Correspondingly, increased abundance of members of Prevotella genus within microbial communities colonizing distinct mucosal surfaces has been found in individuals diagnosed with rheumatoid arthritis, periodontitis, metabolic disorders, and intestinal and vaginal dysbiosis. Still, the role of Prevotella spp. in the incidence of these diseases continues to be debated. For many years, poor understanding of Prevotella biology could be in large part attributed to the lack of experimental tools. However, in the recent years significant advances have been made towards overcoming these limitations, including increased number of isolates and improved understanding of genetic diversity. Besides discussing the most relevant associations between Prevotella spp. and inflammatory disorders, in the present review we examine the recent efforts to expand the Prevotella experimental "toolbox" and we highlight remaining experimental challenges that should advance future research and our understanding of Prevotella-host interplay.


Assuntos
Disbiose , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos , Inflamação/microbiologia , Prevotella/metabolismo , Trato Gastrointestinal , Humanos , Intestinos
12.
Int Immunol ; 32(9): 597-603, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-31882997

RESUMO

Our bodies are constantly exposed to a wide variety of pathogenic micro-organisms through mucosal sites. Therefore, effective vaccines that can protect at the mucosa are vital; however, only a few clinically established mucosal vaccines are available. Although conventional injectable vaccines can induce antigen-specific serum immunoglobulin G (IgG) and prevent severe infection, it is difficult to efficiently inhibit the invasion of pathogens at mucosal surfaces because of the inadequate ability to induce antigen-specific IgA. Recently, we have developed a parenteral vaccine with emulsified curdlan and CpG oligodeoxynucleotides and reported its application. Unlike other conventional injectable vaccines, this immunization contributes to the induction of antigen-specific mucosal and systemic immune responses. Even if antigen-specific IgA at the mucosa disappears, this immunization can induce high-titer IgA after boosting with a small amount of antigen on the target mucosal surface. Indeed, vaccination with Streptococcus pneumoniae antigen effectively prevented lung infection induced by this bacterium. In addition, vaccination with Clostridium ramosum, which is a representative pathobiont associated with obesity and diabetes in humans, reduced obesity in mice colonized with this microorganism. This immunization approach might be an effective treatment for intestinal bacteria-mediated diseases that have been difficult to regulate so far, as well as common infectious diseases.


Assuntos
Mucosa/imunologia , Vacinas/imunologia , Animais , Humanos , Imunidade nas Mucosas/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia
13.
Trends Immunol ; 39(12): 992-1004, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30377046

RESUMO

The gut microbiota is important in health and disease. Whereas the intestinal immune system has evolved to protect the mucosal barrier against pathogens, there is much interest in understanding how it influences the composition and functions of resident microbial communities. Overall, host innate immunity exerts little influence on the microbiota at homeostasis, but increases upon immune activation and the onset of inflammation, as well as in the presence of certain members of the microbiota. However, many experiments have not adequately incorporated study design to detect such immune influences, including using proper control groups, precise sampling and timing, and measures beyond broad-scale descriptions of dysbiosis for microbial analysis. We discuss these and other challenges in the context of current understanding of chronic inflammatory disease.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunidade Inata/imunologia , Animais , Inflamação/imunologia , Doenças Inflamatórias Intestinais/imunologia , Camundongos , Modelos Animais
14.
Med Microbiol Immunol ; 210(4): 173-179, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34021796

RESUMO

Trillions of bacteria inhabit the mammalian gastrointestinal tract. In the majority of hosts, these symbionts contribute largely to beneficial functions promoting microbe-host homeostasis. However, an increasing number of human diseases is associated with altered microbiota composition and enrichment of certain bacterial species. A well-known example of this is Mucispirillum schaedleri, which has been associated with inflammatory conditions in the intestine. Mucispirillum spp. belong to the phylum Deferribacteres and are prevalent but low abundant members of the rodent, pig and human microbiota. Recently, M. schaedleri was causally linked to the development of Crohn's disease-like colitis in immunodeficient mice. While this study certifies a considerable pathogenic potential, the same organism can also promote health in the immunocompetent host: M. schaedleri protects from Salmonella enterica serovar Typhimurium (S. Tm)-induced colitis by interfering with the expression of the pathogen´s invasion machinery. In this review, we summarize the current knowledge on the mammalian gut symbiont M. schaedleri and its role in intestinal homeostasis and discuss open questions and perspectives for future research.


Assuntos
Bactérias , Doença de Crohn/microbiologia , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais/microbiologia , Infecções por Salmonella/prevenção & controle , Simbiose , Animais , Infecções Bacterianas/microbiologia , Homeostase , Interações entre Hospedeiro e Microrganismos , Humanos , Mucosa Intestinal/microbiologia , Camundongos , Interações Microbianas , Infecções por Salmonella/microbiologia , Salmonella typhimurium/crescimento & desenvolvimento
15.
Periodontol 2000 ; 86(1): 210-230, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33690950

RESUMO

In health, indigenous polymicrobial communities at mucosal surfaces maintain an ecological balance via both inter-microbial and host-microbial interactions that promote their own and the host's fitness, while preventing invasion by exogenous pathogens. However, genetic and acquired destabilizing factors (including immune deficiencies, immunoregulatory defects, smoking, diet, obesity, diabetes and other systemic diseases, and aging) may disrupt this homeostatic balance, leading to selective outgrowth of species with the potential for destructive inflammation. This process, known as dysbiosis, underlies the development of periodontitis in susceptible hosts. The pathogenic process is not linear but involves a positive-feedback loop between dysbiosis and the host inflammatory response. The dysbiotic community is essentially a quasi-organismal entity, where constituent organisms communicate via sophisticated physical and chemical signals and display functional specialization (eg, accessory pathogens, keystone pathogens, pathobionts), which enables polymicrobial synergy and dictates the community's pathogenic potential or nososymbiocity. In this review, we discuss early and recent studies in support of the polymicrobial synergy and dysbiosis model of periodontal disease pathogenesis. According to this concept, disease is not caused by individual "causative pathogens" but rather by reciprocally reinforced interactions between physically and metabolically integrated polymicrobial communities and a dysregulated host inflammatory response.


Assuntos
Doenças Periodontais , Periodontite , Disbiose , Interações Hospedeiro-Patógeno , Humanos , Inflamação
16.
Clin Infect Dis ; 70(6): 1236-1239, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-31309972

RESUMO

In Montevideo (2013-2018), 8 Campylobacter fetus extraintestinal infections were reported. The polyclonal nature of strains revealed by whole-genome sequencing and the apparent lack of epidemiological links was incompatible with a single contamination source, supporting alternative routes of transmission.


Assuntos
Infecções por Campylobacter , Campylobacter , Infecções por Campylobacter/epidemiologia , Campylobacter fetus/genética , Humanos , Uruguai/epidemiologia
17.
Ann Rheum Dis ; 78(7): 947-956, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30782585

RESUMO

BACKGROUND/PURPOSE: To search for a transmissible agent involved in lupus pathogenesis, we investigated the faecal microbiota of patients with systemic lupus erythematosus (SLE) for candidate pathobiont(s) and evaluated them for special relationships with host immunity. METHODS: In a cross-sectional discovery cohort, matched blood and faecal samples from 61 female patients with SLE were obtained. Faecal 16 S rRNA analyses were performed, and sera profiled for antibacterial and autoantibody responses, with findings validated in two independent lupus cohorts. RESULTS: Compared with controls, the microbiome in patients with SLE showed decreased species richness diversity, with reductions in taxonomic complexity most pronounced in those with high SLE disease activity index (SLEDAI). Notably, patients with SLE had an overall 5-fold greater representation of Ruminococcus gnavus (RG) of the Lachnospiraceae family, and individual communities also displayed reciprocal contractions of a species with putative protective properties. Gut RG abundance correlated with serum antibodies to only 1/8 RG strains tested. Anti-RG antibodies correlated directly with SLEDAI score and antinative DNA levels, but inversely with C3 and C4. These antibodies were primarily against antigen(s) in an RG strain-restricted pool of cell wall lipoglycans. Novel structural features of these purified lipoglycans were characterised by mass spectrometry and NMR. Highest levels of serum anti-RG strain-restricted antibodies were detected in those with active nephritis (including Class III and IV) in the discovery cohort, with findings validated in two independent cohorts. CONCLUSION: These findings suggest a novel paradigm in which specific strains of a gut commensal may contribute to the immune pathogenesis of lupus nephritis.


Assuntos
Anticorpos Antibacterianos/imunologia , Autoanticorpos/imunologia , Fezes/microbiologia , Lúpus Eritematoso Sistêmico/microbiologia , Nefrite Lúpica/imunologia , Adulto , Anticorpos Antibacterianos/sangue , Autoanticorpos/sangue , Estudos de Casos e Controles , Clostridiales/imunologia , Clostridiales/isolamento & purificação , Estudos Transversais , Feminino , Humanos , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Nefrite Lúpica/microbiologia , RNA Ribossômico 16S/análise , Índice de Gravidade de Doença
18.
Appl Environ Microbiol ; 84(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29079623

RESUMO

The critically endangered elkhorn coral (Acropora palmata) is affected by white pox disease (WPX) throughout the Florida Reef Tract and wider Caribbean. The bacterium Serratia marcescens was previously identified as one etiologic agent of WPX but is no longer consistently detected in contemporary outbreaks. It is now believed that multiple etiologic agents cause WPX; however, to date, no other potential pathogens have been thoroughly investigated. This study examined the association of Vibrio bacteria with WPX occurrence from August 2012 to 2014 at Looe Key Reef in the Florida Keys, USA. The concentration of cultivable Vibrio was consistently greater in WPX samples than in healthy samples. The abundance of Vibrio bacteria relative to total bacteria was four times higher in samples from WPX lesions than in adjacent apparently healthy regions of diseased corals based on quantitative PCR (qPCR). Multilocus sequence analysis (MLSA) was used to assess the diversity of 69 Vibrio isolates collected from diseased and apparently healthy A. palmata colonies and the surrounding seawater. Vibrio species with known pathogenicity to corals were detected in both apparently healthy and diseased samples. While the causative agent(s) of contemporary WPX outbreaks remains elusive, our results suggest that Vibrio spp. may be part of a nonspecific heterotrophic bacterial bloom rather than acting as primary pathogens. This study highlights the need for highly resolved temporal sampling in situ to further elucidate the role of Vibrio during WPX onset and progression.IMPORTANCE Coral diseases are increasing worldwide and are now considered a major contributor to coral reef decline. In particular, the Caribbean has been noted as a coral disease hot spot, owing to the dramatic loss of framework-building acroporid corals due to tissue loss diseases. The pathogenesis of contemporary white pox disease (WPX) outbreaks in Acropora palmata remains poorly understood. This study investigates the association of Vibrio bacteria with WPX.


Assuntos
Antozoários/microbiologia , Vibrioses/epidemiologia , Vibrio/genética , Animais , Recifes de Corais , Espécies em Perigo de Extinção , Florida/epidemiologia , Variação Genética , Tipagem de Sequências Multilocus , RNA Ribossômico 16S , Água do Mar/microbiologia , Vibrio/classificação , Vibrio/isolamento & purificação , Vibrio/patogenicidade
19.
J Physiol ; 595(2): 465-476, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27426277

RESUMO

The oral microbiome is established within a few minutes after birth and consists of stable multi-species communities that engage in a dynamic equilibrium with the host immune system. Dental caries, endodontic infections and periodontal diseases are bacterially driven diseases that are caused by dysbiotic microbiomes. Over a century ago, the focal infection theory implicated these infections in the aetiology of several systemic diseases, ranging from arthritis to neurodegenerative diseases. However, a lack of concrete evidence, combined with the urgency with which clinicians embraced this approach without regard for appropriate case selection, led to its demise within 30 years. In the last decade of the 20th century, the concept of periodontal medicine was introduced to explain the correlations that were being observed between periodontitis and cardiovascular disease, rheumatoid arthritis, Alzheimer's disease, pulmonary disease, pre-term delivery of low birth weight infants and metabolic disease. It was proposed that periodontal pathobionts played a causal role in the initiating or exacerbating certain diseases either by direct invasion or by stimulating a florid immune-inflammatory response that extended into the systemic circulation. This review will examine the strength of current evidence in establishing a causal link between oral pathobionts and systemic disease.


Assuntos
Microbiota , Boca/microbiologia , Doenças Periodontais/complicações , Sepse/complicações , Humanos , Doenças Periodontais/microbiologia , Periodontia , Sepse/microbiologia
20.
Trends Immunol ; 35(1): 3-11, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24269668

RESUMO

Recent studies have uncovered novel mechanisms underlying the breakdown of periodontal host-microbe homeostasis, which can precipitate dysbiosis and periodontitis in susceptible hosts. Dysbiotic microbial communities of keystone pathogens and pathobionts are thought to exhibit synergistic virulence whereby not only can they endure the host response but can also thrive by exploiting tissue-destructive inflammation, which fuels a self-feeding cycle of escalating dysbiosis and inflammatory bone loss, potentially leading to tooth loss and systemic complications. Here, I discuss new paradigms in our understanding of periodontitis, which may shed light into other polymicrobial inflammatory disorders. In addition, I highlight gaps in knowledge required for an integrated picture of the interplay between microbes and innate and adaptive immune elements that initiate and propagate chronic periodontal inflammation.


Assuntos
Periodontite/imunologia , Periodontite/microbiologia , Perda do Osso Alveolar , Comunicação Celular/imunologia , Suscetibilidade a Doenças/imunologia , Interações Hospedeiro-Patógeno , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Periodontite/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA