Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.251
Filtrar
1.
Commun Biol ; 7(1): 1145, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39277660

RESUMO

Pathogenic changes in gut microbial composition precede the onset of HIV-1 infection in men who have sex with men (MSM). This process is associated with increased levels of systemic inflammatory biomarkers and risk for AIDS development. Using mediation analysis framework, in this report we link the effects of unprotected receptive intercourse among MSM prior to primary HIV-1 infection to higher levels of proinflammatory cytokines sCD14 and sCD163 in plasma and a significant decrease in the abundance of A. muciniphila, B. caccae, B. fragilis, B. uniformis, Bacteroides spp., Butyricimonas spp., and Odoribacter spp., and a potential increase in the abundance of Dehalobacterium spp. and Methanobrevibacter spp. in stools of MSM with the highest number of sexual partners. These differences in microbiota, together with a reduction in the pairwise correlations among commensal and short-chain fatty acid-producing bacteria with a number of sexual partners, support an increase in gut dysbiosis with the number of sexual partners. These results demonstrate the interconnectedness of sexual behavior, immune response, and microbiota composition, notably among MSM participating in high-risk sexual behaviors.


Assuntos
Microbioma Gastrointestinal , Infecções por HIV , HIV-1 , Homossexualidade Masculina , Inflamação , Comportamento Sexual , Masculino , Humanos , Infecções por HIV/microbiologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Adulto , Inflamação/microbiologia , HIV-1/fisiologia , Disbiose/microbiologia , Pessoa de Meia-Idade
2.
Adv Exp Med Biol ; 1460: 357-372, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39287858

RESUMO

Gut microbiota is an indispensable commensal partner of human superorganism. The wealth of genetic repertoire provided by these microorganisms extends host's substrate processing capability. Energy and nutrient harvesting machinery primarily depends on the proper function of these organisms. However, the dynamic composition of microbiota changes with age, lifestyle, stress factors, infections, medications, and host pathophysiological conditions. Host immune system is primarily responsible for shaping up the microbial community and sustaining the symbiotic state. This involves controlling the delicate balance between agility toward pathobionts and tolerance toward symbionts. When things go wrong with this crosstalk, dysbiosis may arise.Metabolic syndrome is a multisystemic, low-grade chronic inflammatory disease that involves dyslipidemia, glucose intolerance, insulin resistance, and central obesity. Excess caloric intake with high-sugar and high-fat diet promote high energy harvesting and lipogenesis. The secretion of adipokines accompanies lipid spillover from fat cells, which contribute to insulin resistance and the expansion of adipose tissue in ectopic sites. Proinflammatory cytokines from adipose tissue macrophages increase the extent of adipose dysfunction.The inflammatory nature of obesity and metabolic syndrome recall the connection between dysbiosis and immune dysfunction. A remarkable association exits between obesity, inflammatory bowel disease, gluten-sensitive enteropathy, and dysbiosis. These conditions compromise the gut mucosa barrier and allow lipopolysaccharide to enter circulation. Unresolved chronic inflammation caused by one condition may overlap or trigger the other(s). Experimental studies and therapeutic trials of fecal microbiota transplantation promise limited improvement in some of these conditions.Typically, metabolic syndrome is considered as a consequence of overnutrition and the vicious cycle of lipogenesis, lipid accumulation, and chronic low-level inflammation. Because of the complex nature of this disorder, it remains inconclusive whether dysbiosis is a cause or consequence of obesity and metabolic syndrome.


Assuntos
Disbiose , Microbioma Gastrointestinal , Síndrome Metabólica , Obesidade , Humanos , Microbioma Gastrointestinal/fisiologia , Síndrome Metabólica/microbiologia , Síndrome Metabólica/metabolismo , Obesidade/microbiologia , Obesidade/metabolismo , Animais , Metabolismo dos Lipídeos , Inflamação/metabolismo , Inflamação/microbiologia , Tecido Adiposo/metabolismo , Tecido Adiposo/imunologia
3.
Nature ; 633(8031): 878-886, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39294375

RESUMO

Persistent colonization and outgrowth of potentially pathogenic organisms in the intestine can result from long-term antibiotic use or inflammatory conditions, and may perpetuate dysregulated immunity and tissue damage1,2. Gram-negative Enterobacteriaceae gut pathobionts are particularly recalcitrant to conventional antibiotic treatment3,4, although an emerging body of evidence suggests that manipulation of the commensal microbiota may be a practical alternative therapeutic strategy5-7. Here we isolated and down-selected commensal bacterial consortia from stool samples from healthy humans that could strongly and specifically suppress intestinal Enterobacteriaceae. One of the elaborated consortia, comprising 18 commensal strains, effectively controlled ecological niches by regulating gluconate availability, thereby re-establishing colonization resistance and alleviating Klebsiella- and Escherichia-driven intestinal inflammation in mice. Harnessing these activities in the form of live bacterial therapies may represent a promising solution to combat the growing threat of proinflammatory, antimicrobial-resistant Enterobacteriaceae infection.


Assuntos
Enterobacteriaceae , Fezes , Microbioma Gastrointestinal , Klebsiella , Simbiose , Animais , Camundongos , Humanos , Feminino , Klebsiella/isolamento & purificação , Fezes/microbiologia , Masculino , Infecções por Enterobacteriaceae/microbiologia , Inflamação/microbiologia , Inflamação/imunologia , Intestinos/microbiologia , Camundongos Endogâmicos C57BL
4.
FASEB J ; 38(18): e70065, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39305117

RESUMO

One in six people are projected to be 65 years or older by 2050. As the population ages, better treatments for injuries that disproportionately impact the aged population will be needed. Clinical studies show that people aged 65 and older experience higher rates of morbidity and mortality after burn injury, including a greater incidence of pulmonary complications when compared to younger burn injured adults, which we and others believe is mediated, in part, by inflammation originating in the intestines. Herein, we use our clinically relevant model of scald burn injury in young and aged mice to determine whether cohousing aged mice with young mice or giving aged mice oral gavage of fecal material from young mice is sufficient to alter the microbiome of the aged mice and protect them from inflammation in the ileum and the lungs. Aged burn injured mice have less DNA expression of Bacteroidetes in the feces and an unhealthy Firmicutes/Bacteroidetes ratio. Both Bacteroidetes and the ratio of these two phyla are restored in aged burn injured by prior cohousing for a month with younger mice but not fecal transfer from young mice. This shift in the microbiome coincides with heightened expression of danger-associated molecular patterns (DAMP), and pro-inflammatory cytokine interleukin-6 (il6) in the ileum and lung of aged, burn injured mice, and heightened antimicrobial peptide camp in the lung. Cohousing reverses DAMP expression in the ileum and lung, and cathelicidin-related antimicrobial peptide protein (camp) in the lung, while fecal transfer heightened DAMPs while reducing camp in the lung, and also increased IL-6 protein in the lungs. These results highlight the importance of the intestinal microbiome in mediating inflammation within the gut-lung axis, giving insights into potential future treatments in the clinic.


Assuntos
Queimaduras , Microbioma Gastrointestinal , Inflamação , Animais , Queimaduras/microbiologia , Camundongos , Inflamação/microbiologia , Camundongos Endogâmicos C57BL , Masculino , Envelhecimento , Fezes/microbiologia , Pulmão/microbiologia , Pulmão/metabolismo , Pulmão/patologia , Transplante de Microbiota Fecal , Bacteroidetes , Íleo/microbiologia , Íleo/metabolismo
5.
Gut Microbes ; 16(1): 2394249, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39224018

RESUMO

Inflammatory bowel diseases (IBD) etiology is multifactorial. Luminal microRNAs (miRNAs) have been suspected to play a role in the promotion of chronic inflammation, but the extent to which fecal miRNAs are interacting with the intestinal ecosystem in a way that contribute to diseases, including IBD, remains unknown. Here, fecal let-7b and miR-21 were found elevated, associated with inflammation, and correlating with multiple bacteria in IBD patients and IL-10-/- mice, model of spontaneous colitis. Using an in vitro microbiota modeling system, we revealed that these two miRNAs can directly modify the composition and function of complex human microbiota, increasing their proinflammatory potential. In vivo investigations revealed that luminal increase of let-7b drastically alters the intestinal microbiota and enhances macrophages' associated proinflammatory cytokines (TNF, IL-6, and IL-1ß). Such proinflammatory effects are resilient and dependent on the bacterial presence. Moreover, we identified that besides impairing the intestinal barrier function, miR-21 increases myeloperoxidase and antimicrobial peptides secretion, causing intestinal dysbiosis. More importantly, in vivo inhibition of let-7b and miR-21 with anti-miRNAs significantly improved the intestinal mucosal barrier function and promoted a healthier host-microbiota interaction in the intestinal lining, which altogether conferred protection against colitis. In summary, we provide evidence of the functional significance of fecal miRNAs in host-microbiota communication, highlighting their therapeutic potential in intestinal inflammation and dysbiosis-related conditions, such as IBD.


Assuntos
Colite , Fezes , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , MicroRNAs , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Humanos , Fezes/microbiologia , Camundongos , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/metabolismo , Colite/microbiologia , Colite/induzido quimicamente , Colite/genética , Inflamação/microbiologia , Inflamação/metabolismo , Disbiose/microbiologia , Camundongos Endogâmicos C57BL , Feminino , Camundongos Knockout , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Masculino , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Citocinas/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/metabolismo , Modelos Animais de Doenças , Interleucina-10/genética , Interleucina-10/metabolismo
6.
No Shinkei Geka ; 52(5): 931-937, 2024 Sep.
Artigo em Japonês | MEDLINE | ID: mdl-39285542

RESUMO

In recent years, the association of the microbiome with various diseases has been reported. The oral and gut microbiomes have been linked to cerebral aneurysms and are involved in the systemic inflammatory response, which is mediated mainly via the immune system. Chronic inflammation plays an important role in the pathogenesis and rupture of cerebral aneurysms, and the microbiome is potentially involved in this process. Moreover, the gut microbiome is involved in acute brain injury following subarachnoid hemorrhage. Thus, further studies on microbiome-targeted treatments for cerebral aneurysm are required.


Assuntos
Microbioma Gastrointestinal , Aneurisma Intracraniano , Aneurisma Intracraniano/microbiologia , Humanos , Boca/microbiologia , Microbiota , Inflamação/microbiologia
7.
Int J Mol Sci ; 25(17)2024 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-39273662

RESUMO

The gut microbiota is one of the most critical factors in human health. It involves numerous physiological processes impacting host health, mainly via immune system modulation. A balanced microbiome contributes to the gut's barrier function, preventing the invasion of pathogens and maintaining the integrity of the gut lining. Dysbiosis, or an imbalance in the gut microbiome's composition and function, disrupts essential processes and contributes to various diseases. This narrative review summarizes key findings related to the gut microbiota in modern multifactorial inflammatory conditions such as ulcerative colitis or Crohn's disease. It addresses the challenges posed by antibiotic-driven dysbiosis, particularly in the context of C. difficile infections, and the development of novel therapies like fecal microbiota transplantation and biotherapeutic drugs to combat these infections. An emphasis is given to restoration of the healthy gut microbiome through dietary interventions, probiotics, prebiotics, and novel approaches for managing gut-related diseases.


Assuntos
Disbiose , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Obesidade , Probióticos , Humanos , Disbiose/microbiologia , Disbiose/terapia , Obesidade/microbiologia , Probióticos/uso terapêutico , Animais , Inflamação/microbiologia , Prebióticos/administração & dosagem
8.
Int J Mol Sci ; 25(17)2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39273314

RESUMO

The human gastrointestinal tract hosts a complex and dynamic community of microorganisms known as the gut microbiota, which play a pivotal role in numerous physiological processes, including digestion, metabolism, and immune function. Recent research has highlighted the significant impact of diet on the gut microbiota composition and functionality, and the consequential effects on host health. Concurrently, there is growing evidence linking the gut microbiota to inflammation, a key factor in many chronic diseases such as inflammatory bowel disease (IBD), obesity, diabetes, and cardiovascular diseases (CVDs). This review explores how dietary components influence the gut microbiota composition, how these microbial changes affect inflammatory pathways, and the therapeutic implications of modulating this axis for chronic inflammatory disease prevention and management. Beneficial dietary patterns, such as the Mediterranean diet (MD) and plant-based diets, promote a diverse and balanced gut microbiota composition, supporting anti-inflammatory pathways. Conversely, the Western diet (WD), high in saturated fats and refined sugars, is associated with dysbiosis and increased inflammation. With all the links between the three variables considered, this review attempts to offer a thorough examination of the triangle formed by inflammation, the gut microbiota, and food.


Assuntos
Dieta , Microbioma Gastrointestinal , Inflamação , Humanos , Inflamação/microbiologia , Disbiose/microbiologia , Animais , Dieta Mediterrânea , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/etiologia , Dieta Ocidental/efeitos adversos
9.
Biochemistry ; 63(18): 2266-2279, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39189508

RESUMO

As part of its pathogenesis, Salmonella enterica serovar Typhimurium delivers effector proteins into host cells. One effector is SspH2, a member of the so-called novel E3 ubiquitin ligase family, that interacts with and enhances, NOD1 pro-inflammatory signaling, though the underlying mechanisms are unclear. Here, we report that SspH2 interacts with multiple members of the NLRC family to enhance pro-inflammatory signaling by targeted ubiquitination. We show that SspH2 modulates host innate immunity by interacting with both NOD1 and NOD2 in mammalian epithelial cell culture via the NF-κB pathway. Moreover, purified SspH2 and NOD1 directly interact, where NOD1 potentiates SspH2 E3 ubiquitin ligase activity. Mass spectrometry and mutational analyses identified four key lysine residues in NOD1 that are required for its enhanced activation by SspH2, but not its basal activity. These critical lysine residues are positioned in the same region of NOD1 and define a surface on the receptor that appears to be targeted by SspH2. Overall, this work provides evidence for post-translational modification of NOD1 by ubiquitin and uncovers a unique mechanism of spatially selective ubiquitination to enhance the activation of an archetypal NLR.


Assuntos
Proteína Adaptadora de Sinalização NOD1 , Salmonella typhimurium , Transdução de Sinais , Ubiquitinação , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD1/genética , Humanos , Salmonella typhimurium/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteína Adaptadora de Sinalização NOD2/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Células HEK293 , Imunidade Inata , Inflamação/metabolismo , Inflamação/microbiologia , NF-kappa B/metabolismo , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Infecções por Salmonella/imunologia
10.
J Int Soc Sports Nutr ; 21(1): 2388085, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39193818

RESUMO

BACKGROUND: Fatigue and gastrointestinal (GI) distress are common among athletes with an estimated 30-90% of athletes participating in marathons, triathlons, or similar events experiencing GI complaints. Intense exercise can lead to increased intestinal permeability, potentially allowing members of the gut microbiota to permeate into the bloodstream, resulting in an inflammatory response and cascade of performance-limiting outcomes. Probiotics, through their capacity to regulate the composition of the gut microbiota, may act as an adjunctive therapy by enhancing GI and immune function while mitigating inflammatory responses. This review investigates the effectiveness of probiotic supplementation on fatigue, inflammatory markers, and exercise performance based on randomized controlled trials (RCTs). METHODS: This review follows the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines and PICOS (Population, Intervention, Comparison, Outcome, Study design) framework. A comprehensive search was conducted in Sportdiscus, PubMed, and Scopus databases, and the screening of titles, abstracts, and full articles was performed based on pre-defined eligibility criteria. Of the 3505 records identified, 1884 were screened using titles and abstracts, of which 450 studies were selected for full-text screening. After final screening, 13 studies met the eligibility criteria and were included for review. The studies contained 513 participants, consisting of 351 males and 115 females, however, two studies failed to mention the sex of the participants. Among the participants, 246 were defined as athletes, while the remaining participants were classified as recreationally active (n = 267). All trials were fully described and employed a double- or triple-blind placebo-controlled intervention using either a single probiotic strain or a multi-strain synbiotic (containing both pro- and pre-biotics). RESULTS: This review assesses the effects of daily probiotic supplementation, ranging from 13 to 90 days, on physical performance and physiological markers in various exercise protocols. Ten studies reported improvements in various parameters, such as, enhanced endurance performance, improved anxiety and stress levels, decreased GI symptoms, and reduced upper respiratory tract infections (URTI). Moreover, despite no improvements in maximal oxygen uptake (VO2), several studies demonstrated that probiotic supplementation led to amelioration in lactate, creatine kinase (CK), and ammonia concentrations, suggesting beneficial effects on mitigating exercise-induced muscular stress and damage. CONCLUSION: Probiotic supplementation, specifically at a minimum dosage of 15 billion CFUs daily for a duration of at least 28 days, may contribute to the reduction of perceived or actual fatigue.


Assuntos
Desempenho Atlético , Fadiga , Gastroenteropatias , Inflamação , Probióticos , Humanos , Atletas , Desempenho Atlético/fisiologia , Fadiga/imunologia , Fadiga/prevenção & controle , Gastroenteropatias/complicações , Gastroenteropatias/dietoterapia , Gastroenteropatias/imunologia , Gastroenteropatias/microbiologia , Microbioma Gastrointestinal/imunologia , Inflamação/complicações , Inflamação/dietoterapia , Inflamação/imunologia , Inflamação/microbiologia , Probióticos/administração & dosagem , Ensaios Clínicos Controlados Aleatórios como Assunto , Fenômenos Fisiológicos da Nutrição Esportiva/imunologia
11.
Microbiol Res ; 288: 127838, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39153466

RESUMO

With the imbalance of intestinal microbiota, the body will then face an inflammatory response, which has serious implications for human health. Bodily allergies, injury or pathogens infections can trigger or promote inflammation and alter the intestinal environment. Meanwhile, excessive changes in the intestinal environment cause the imbalance of microbial homeostasis, which leads to the proliferation and colonization of opportunistic pathogens, invasion of the body's immune system, and the intensification of inflammation. Some natural compounds and gut microbiota and metabolites can reduce inflammation; however, the details of how they interact with the gut immune system and reduce the gut inflammatory response still need to be fully understood. The review focuses on inflammation and intestinal microbiota imbalance caused by pathogens. The body reacts differently to different types of pathogenic bacteria, and the ingestion of pathogens leads to inflamed gastrointestinal tract disorders or intestinal inflammation. In this paper, unraveling the interactions between the inflammation, pathogenic bacteria, and intestinal microbiota based on inflammation caused by several common pathogens. Finally, we summarize the effects of intestinal metabolites and natural anti-inflammatory substances on inflammation to provide help for related research of intestinal inflammation caused by pathogenic bacteria.


Assuntos
Bactérias , Microbioma Gastrointestinal , Inflamação , Humanos , Inflamação/microbiologia , Inflamação/metabolismo , Animais , Bactérias/metabolismo , Bactérias/classificação , Intestinos/microbiologia , Anti-Inflamatórios/metabolismo , Homeostase , Disbiose/microbiologia
12.
Lung ; 202(5): 711-722, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39096413

RESUMO

PURPOSE: Pseudomonas aeruginosa is the predominant bacterial pathogen colonizing the cystic fibrosis (CF) lung. Mixed populations of nonmucoid and mucoid variants of P. aeruginosa have been isolated from the CF airway. While the association between mucoid variants and pulmonary function decline is well-established, their impact on inflammation and tissue damage in advanced CF lung disease remains unclear. METHODS: This pilot study utilized 1 non-CF and 3 CF lung explants to examine lobar distribution, inflammation, and histopathology related to nonmucoid and mucoid P. aeruginosa infection. To study tissue damage, we developed a novel lung histopathology scoring system, the first applied to human CF lung biopsies, which is comprised of five indicators: bronchiolar epithelial infiltrate, luminal inflammation, peribronchial/bronchiolar infiltrate, peribronchiolar fibrosis, and alveolar involvement. RESULTS: Mucoid P. aeruginosa variants were distributed throughout the CF lung but associated with greater concentrations of proinflammatory cytokines, IL-1ß, TNF-α, IL-6, IL-8, and IFN-γ, and one anti-inflammatory cytokine, IL-10, compared to nonmucoid variants. CF lung explants exhibited higher histopathology scores compared to a non-CF lung control. In mixed-variant infection, nonmucoid constituents associated with increased bronchiolar epithelial infiltration, one indicator of histopathology. CONCLUSION: This pilot study suggests ongoing interplay between host and bacterial elements in late-stage CF pulmonary disease. Mucoid P. aeruginosa infection correlates with inflammation regardless of lung lobe, whereas nonmucoid P. aeruginosa is associated with increased inflammatory cell infiltration. The development of a novel lung histopathology scoring system lays the groundwork for future large-cohort investigations.


Assuntos
Fibrose Cística , Citocinas , Pulmão , Infecções por Pseudomonas , Pseudomonas aeruginosa , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Fibrose Cística/complicações , Humanos , Projetos Piloto , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/isolamento & purificação , Pulmão/patologia , Pulmão/microbiologia , Citocinas/metabolismo , Masculino , Feminino , Biópsia , Adulto , Estudos de Casos e Controles , Mediadores da Inflamação/metabolismo , Inflamação/patologia , Inflamação/microbiologia , Interleucina-8/metabolismo , Fator de Necrose Tumoral alfa
13.
Gut Microbes ; 16(1): 2392874, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39163515

RESUMO

Alterations in intestinal permeability and the gut microbiome caused by alcohol abuse are associated with alcoholic liver disease and with worsening of inflammatory bowel diseases (IBD) symptoms. To resolve the direct effects of chronic ethanol consumption on the colon and its microbiome in the absence of acute or chronic alcohol-induced liver disease, we developed a mouse model of chronic binge drinking that uncovers how alcohol may enhance susceptibility to colitis via the microbiota. Employing daily ethanol gavage, we recapitulate key features of binge ethanol consumption. We found that binge ethanol drinking worsens intestinal infection, colonic injury and inflammation, and this effect persists beyond the drinking period. Using gnotobiotics, we showed that alcohol-driven susceptibility to colitis is microbiota-dependent and transferable to ethanol-naïve mice by microbiome transplantation. Allobaculum spp. expanded in binge drinking mice, and administration of Allobaculum fili was sufficient to enhance colitis in non-drinking mice. Our study provides a model to study binge drinking-microbiota interactions and their effects on host disease and reinforces the pathogenic function of Allobaculum spp. as colitogenic bacteria. Our findings illustrate how chronic binge drinking-induced alterations of the microbiome may affect susceptibility to IBD onset or flares.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas , Colite , Colo , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Animais , Consumo Excessivo de Bebidas Alcoólicas/complicações , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Colite/microbiologia , Colite/induzido quimicamente , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Bactérias/classificação , Bactérias/isolamento & purificação , Bactérias/genética , Etanol/efeitos adversos , Suscetibilidade a Doenças , Masculino , Vida Livre de Germes , Inflamação/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia
14.
Exp Dermatol ; 33(8): e15150, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39113601

RESUMO

Acne is a chronic inflammatory skin condition that involves Cutibacterium acnes (C. acnes), which is classified into six main phylotypes (IA1, IA2, IB, IC, II and III). Acne development is associated with loss of C. acnes phylotype diversity, characterised by overgrowth of phylotype IA1 relative to other phylotypes. It was also shown that purified extracellular vesicles (EVs) secreted by C. acnes can induce an acne-like inflammatory response in skin models. We aimed to determine if the inflammatory profile of EVs secreted by C. acnes phylotype IA1 from an inflammatory acne lesion was different from C. acnes phylotype IA1 from normal skin, thus playing a direct role in the severity of inflammation. EVs were produced in vitro after culture of two clinical strains of C. acnes phylotype IA1, T5 from normal human skin and A47 from an inflammatory acne lesion, and then incubated with either human immortalised keratinocytes, HaCaT cells, or skin explants obtained from abdominoplasty. Subsequently, quantitative PCR (qPCR) was performed for human ß-defensin 2 (hBD2), cathelicidin (LL-37), interleukin (IL)-1ß, IL-6, IL-8, IL-17α and IL-36γ, and ELISA for IL-6, IL-8 and IL-17α. We found that EVs produced in vitro by C. acnes derived from inflammatory acne lesions significantly increased the pro-inflammatory cytokines and anti-microbial peptides at both transcriptional and protein levels compared with EVs derived from normal human skin. We show for the first time that C. acnes EVs from inflammatory acne play a crucial role in acne-associated inflammation in vitro and that C. acnes phylotype IA1 collected from inflammatory acne lesion and normal skin produce different EVs and inflammatory profiles in vitro.


Assuntos
Acne Vulgar , Vesículas Extracelulares , Queratinócitos , Propionibacterium acnes , Humanos , Vesículas Extracelulares/metabolismo , Acne Vulgar/microbiologia , Queratinócitos/microbiologia , Pele/microbiologia , Inflamação/microbiologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Células HaCaT , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Citocinas/metabolismo , Interleucina-17/metabolismo , Propionibacteriaceae
15.
Front Immunol ; 15: 1413485, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39144142

RESUMO

Millions of microorganisms make up the complex microbial ecosystem found in the human gut. The immune system's interaction with the gut microbiota is essential for preventing inflammation and maintaining intestinal homeostasis. Numerous metabolic products that can cross-talk between immune cells and the gut epithelium are metabolized by the gut microbiota. Traumatic injury elicits a great and multifaceted immune response in the minutes after the initial offense, containing simultaneous pro- and anti-inflammatory responses. The development of innovative therapies that improve patient outcomes depends on the gut microbiota and immunological responses to trauma. The altered makeup of gut microbes, or gut dysbiosis, can also dysregulate immunological responses, resulting in inflammation. Major human diseases may become more common as a result of chronic dysbiosis and the translocation of bacteria and the products of their metabolism beyond the mucosal barrier. In this review, we briefly summarize the interactions between the gut microbiota and the immune system and human disease and their therapeutic probiotic formulations. We also discuss the immune response to traumatic injury.


Assuntos
Disbiose , Microbioma Gastrointestinal , Ferimentos e Lesões , Humanos , Microbioma Gastrointestinal/imunologia , Disbiose/imunologia , Animais , Ferimentos e Lesões/imunologia , Ferimentos e Lesões/microbiologia , Probióticos/uso terapêutico , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Inflamação/imunologia , Inflamação/microbiologia
16.
Gut Microbes ; 16(1): 2387857, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39171684

RESUMO

Imbalances in proteolytic activity have been linked to the development of inflammatory bowel diseases (IBD) and experimental colitis. Proteases in the intestine play important roles in maintaining homeostasis, but exposure of mucosal tissues to excess proteolytic activity can promote pathology through protease-activated receptors (PARs). Previous research implicates microbial proteases in IBD, but the underlying pathways and specific interactions between microbes and PARs remain unclear. In this study, we investigated the role of microbial proteolytic activation of the external domain of PAR2 in intestinal injury using mice expressing PAR2 with a mutated N-terminal external domain that is resistant to canonical activation by proteolytic cleavage. Our findings demonstrate the key role of proteolytic cleavage of the PAR2 external domain in promoting intestinal permeability and inflammation during colitis. In wild-type mice expressing protease-sensitive PAR2, excessive inflammation leads to the expansion of bacterial taxa that cleave the external domain of PAR2, exacerbating colitis severity. In contrast, mice expressing mutated protease-resistant PAR2 exhibit attenuated colitis severity and do not experience the same proteolytic bacterial expansion. Colonization of wild-type mice with proteolytic PAR2-activating Enterococcus and Staphylococcus worsens colitis severity. Our study identifies a previously unknown interaction between proteolytic bacterial communities, which are shaped by inflammation, and the external domain of PAR2 in colitis. The findings should encourage new therapeutic developments for IBD by targeting excessive PAR2 cleavage by bacterial proteases.


Assuntos
Colite , Proteólise , Receptor PAR-2 , Animais , Receptor PAR-2/metabolismo , Receptor PAR-2/genética , Colite/microbiologia , Colite/patologia , Colite/metabolismo , Camundongos , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Inflamação/metabolismo , Inflamação/microbiologia , Enterococcus/genética , Enterococcus/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Bactérias/genética , Bactérias/metabolismo , Bactérias/classificação , Bactérias/enzimologia , Modelos Animais de Doenças , Humanos , Domínios Proteicos , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia
17.
Vet Microbiol ; 297: 110203, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39089141

RESUMO

Many cattle infected with Mycoplasma bovis remain healthy while others develop severe chronic respiratory disease. We hypothesized that inflammatory stimuli such as co-pathogens worsen disease outcomes in M. bovis-infected calves. Calves (n=24) were intrabronchially inoculated with M. bovis and either killed bacterial lysate, transient M. haemolytica infection, or saline. Caseonecrotic lesions developed in 7/7 animals given M. haemolytica and M. bovis compared to 2/8 given M. bovis with no inflammatory stimulus, and 6/9 animals given bacterial lysate and M. bovis (P=0.01). Animals receiving M. haemolytica and M. bovis had more caseonecrotic foci in lungs than those receiving M. bovis with no inflammatory stimulus (median = 21 vs 0; P = 0.01), with an intermediate response (median = 5) in animals given bacterial lysate. In addition to caseonecrotic foci, infected animals developed neutrophilic bronchiolitis that appeared to develop into caseonecrotic foci, peribronchiolar lymphocytic cuffs that were not associated with the other lesions, and 4 animals with bronchiolitis obliterans. The data showed that transient lung inflammation at the time of M. bovis infection provoked the development of caseonecrotic bronchopneumonia, and the severity of inflammation influenced the number of caseonecrotic foci that developed. In contrast, caseonecrotic lesions were few or absent in M. bovis-infected calves without a concurrent inflammatory stimulus. These studies provide insight into how caseonecrotic lesions develop within the lung of M. bovis-infected calves. This and other studies suggest that controlling co-pathogens and harmful inflammatory responses in animals infected with M. bovis could potentially minimize development of M. bovis caseonecrotic bronchopneumonia.


Assuntos
Doenças dos Bovinos , Pulmão , Mycoplasma bovis , Pneumonia por Mycoplasma , Animais , Bovinos , Pneumonia por Mycoplasma/veterinária , Pneumonia por Mycoplasma/microbiologia , Doenças dos Bovinos/microbiologia , Doenças dos Bovinos/imunologia , Pulmão/microbiologia , Pulmão/patologia , Inflamação/veterinária , Inflamação/microbiologia , Mannheimia haemolytica/patogenicidade , Coinfecção/veterinária , Coinfecção/microbiologia
18.
CNS Neurosci Ther ; 30(8): e70003, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39161161

RESUMO

AIMS: We evaluated the potential of Parkinson's disease (PD) fecal microbiota transplantation to initiate or exacerbate PD pathologies and investigated the underlying mechanisms. METHODS: We transplanted the fecal microbiota from PD patients into mice by oral gavage and assessed the motor and intestinal functions, as well as the inflammatory and pathological changes in the colon and brain. Furthermore, 16S rRNA gene sequencing combined with metabolomics analysis was conducted to assess the impacts of fecal delivery on the fecal microbiota and metabolism in recipient mice. RESULTS: The fecal microbiota from PD patients increased intestinal inflammation, deteriorated intestinal barrier function, intensified microglia and astrocyte activation, abnormal deposition of α-Synuclein, and dopaminergic neuronal loss in the brains of A53T mice. A mechanistic study revealed that the fecal microbiota of PD patients stimulated the TLR4/NF-κB/NLRP3 pathway in both the brain and colon. Additionally, multiomics analysis found that transplantation of fecal microbiota from PD patients not only altered the composition of the gut microbiota but also influenced the fecal metabolic profile of the recipient mice. CONCLUSION: The fecal microbiota from PD patients intensifies inflammation and neurodegeneration in A53T mice. Our findings demonstrate that imbalance and dysfunction in the gut microbiome play significant roles in the development and advancement of PD.


Assuntos
Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Doença de Parkinson , Animais , Camundongos , Doença de Parkinson/microbiologia , Doença de Parkinson/metabolismo , Humanos , Microbioma Gastrointestinal/fisiologia , Masculino , Inflamação/metabolismo , Inflamação/microbiologia , Fezes/microbiologia , Camundongos Transgênicos , Camundongos Endogâmicos C57BL , Feminino , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia
19.
BMJ Open Gastroenterol ; 11(1)2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39209769

RESUMO

OBJECTIVE: Gut microbes and microbe-dependent metabolites (eg, tryptophan-kynurenine-serotonin pathway metabolites) have been linked to systemic inflammation, but the microbiota-metabolite-inflammation axis remains uncharacterised in children. Here we investigated whether gut microbiota features and circulating metabolites (both microbe-dependent and non-microbe-dependent metabolites) associated with circulating inflammation markers in children. METHODS: We studied children from the prospective Gen3G birth cohort who had data on untargeted plasma metabolome (n=321 children; Metabolon platform), gut microbiota (n=147; 16S rRNA sequencing), and inflammation markers (plasminogen activator inhibitor-1 (PAI-1), monocyte chemoattractant protein-1, and tumour necrosis factor-α) measured at 5-7 years. We examined associations of microbial taxa and metabolites-examining microbe-dependent and non-microbe-dependent metabolites separately-with each inflammatory marker and with an overall inflammation score (InfSc), adjusting for key confounders and correcting for multiple comparisons. We also compared the proportion of significantly associated microbe-dependent versus non-microbe-dependent metabolites, identified a priori (Human Microbial Metabolome Database), with each inflammation marker. RESULTS: Of 335 taxa tested, 149 were associated (qFDR<0.05) with at least one inflammatory marker; 10 of these were robust to pseudocount choice. Several bacterial taxa involved in tryptophan metabolism were associated with inflammation, including kynurenine-degrading Ruminococcus, which was inversely associated with all inflammation markers. Of 1037 metabolites tested, 315 were previously identified as microbe dependent and were more frequently associated with PAI-1 and the InfSc than non-microbe dependent metabolites. In total, 87 metabolites were associated (qFDR<0.05) with at least one inflammation marker, including kynurenine (positively), serotonin (positively), and tryptophan (inversely). CONCLUSION: A distinct set of gut microbes and microbe-dependent metabolites, including those involved in the tryptophan-kynurenine-serotonin pathway, may be implicated in inflammatory pathways in childhood.


Assuntos
Biomarcadores , Microbioma Gastrointestinal , Inflamação , Metaboloma , Inibidor 1 de Ativador de Plasminogênio , Humanos , Microbioma Gastrointestinal/fisiologia , Criança , Feminino , Masculino , Inflamação/microbiologia , Inflamação/sangue , Biomarcadores/sangue , Estudos Prospectivos , Pré-Escolar , Inibidor 1 de Ativador de Plasminogênio/sangue , Metaboloma/fisiologia , Triptofano/sangue , Triptofano/metabolismo , Cinurenina/sangue , Cinurenina/metabolismo , Fator de Necrose Tumoral alfa/sangue , RNA Ribossômico 16S/genética , Quimiocina CCL2/sangue
20.
Nat Commun ; 15(1): 7604, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39217183

RESUMO

Interrupting transmission events is critical to tuberculosis control. Cough-generated aerosol cultures predict tuberculosis transmission better than microbiological or clinical markers. We hypothesize that highly infectious individuals with pulmonary tuberculosis (positive for cough aerosol cultures) have elevated inflammatory markers and unique transcriptional profiles compared to less infectious individuals. We performed a prospective, longitudinal study using cough aerosol sampling system. We enrolled 142 participants with treatment-naïve pulmonary tuberculosis in Kenya and assessed the association of clinical, microbiologic, and immunologic characteristics with Mycobacterium tuberculosis aerosolization and transmission in 129 household members. Contacts of the forty-three aerosol culture-positive participants (30%) are more likely to have a positive interferon-gamma release assay (85% vs 53%, P = 0.006) and higher median IFNγ level (P < 0.001, 4.28 IU/ml (1.77-5.91) vs. 0.71 (0.01-3.56)) compared to aerosol culture-negative individuals. We find that higher bacillary burden, younger age, larger mean upper arm circumference, and host inflammatory profiles, including elevated serum C-reactive protein and lower plasma TNF levels, associate with positive cough aerosol cultures. Notably, we find pre-treatment whole blood transcriptional profiles associate with aerosol culture status, independent of bacillary load. These findings suggest that tuberculosis infectiousness is associated with epidemiologic characteristics and inflammatory signatures and that these features may identify highly infectious persons.


Assuntos
Aerossóis , Tosse , Mycobacterium tuberculosis , Tuberculose Pulmonar , Humanos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/fisiologia , Tosse/microbiologia , Masculino , Feminino , Adulto , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/imunologia , Estudos Prospectivos , Estudos Longitudinais , Quênia/epidemiologia , Pessoa de Meia-Idade , Adulto Jovem , Interferon gama/sangue , Interferon gama/genética , Proteína C-Reativa/análise , Proteína C-Reativa/metabolismo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/genética , Inflamação/microbiologia , Adolescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...