Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 736
Filtrar
1.
Alcohol ; 107: 136-143, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36150609

RESUMO

Alcohol use among older adults is on the rise. This increase is clinically relevant as older adults are at risk for increased morbidity and mortality from many alcohol-related chronic diseases compared to younger patients. However, little is known regarding the synergistic effects of alcohol and age. There are intriguing data suggesting that aging may lead to impaired intestinal barrier integrity and dysbiosis of the intestinal microbiome, which could increase susceptibility to alcohol's negative effects. To study the effects of alcohol in age we exposed aged and young mice to 3 days of moderate ethanol and evaluated changes in gut parameters. We found that these levels of drinking do not have obvious effects in young mice but cause significant alcohol-induced gut barrier dysfunction and expression of the pro-inflammatory cytokine TNFα in aged mice. Ethanol-induced downregulation of expression of the gut-protective antimicrobial peptides Defa-rs1, Reg3b, and Reg3g was observed in aged, but not young mice. Analysis of the fecal microbiome revealed age-associated shifts in microbial taxa, which correlated with intestinal and hepatic inflammatory gene expression. Taken together, these data demonstrate that age drives microbiome dysbiosis, while ethanol exposure in aged mice induces changes in the expression of antimicrobial genes important for separating these potentially damaging microbes from the intestinal lumen. These changes highlight potential mechanistic targets for prevention of the age-related exacerbation of effects of ethanol on the gut.


Assuntos
Disbiose , Etanol , Microbioma Gastrointestinal , Inflamação , Intestinos , Animais , Camundongos , Peptídeos Antimicrobianos/genética , Peptídeos Antimicrobianos/imunologia , Citocinas/imunologia , Disbiose/induzido quimicamente , Disbiose/genética , Disbiose/imunologia , Disbiose/microbiologia , Etanol/farmacologia , Etanol/toxicidade , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Intestinos/microbiologia , Camundongos Endogâmicos C57BL , Permeabilidade/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , alfa-Defensinas/genética , alfa-Defensinas/imunologia
2.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35163417

RESUMO

Some say that all diseases begin in the gut. Interestingly, this concept is actually quite old, since it is attributed to the Ancient Greek physician Hippocrates, who proposed the hypothesis nearly 2500 years ago. The continuous breakthroughs in modern medicine have transformed our classic understanding of the gastrointestinal tract (GIT) and human health. Although the gut microbiota (GMB) has proven to be a core component of human health under standard metabolic conditions, there is now also a strong link connecting the composition and function of the GMB to the development of numerous diseases, especially the ones of musculoskeletal nature. The symbiotic microbes that reside in the gastrointestinal tract are very sensitive to biochemical stimuli and may respond in many different ways depending on the nature of these biological signals. Certain variables such as nutrition and physical modulation can either enhance or disrupt the equilibrium between the various species of gut microbes. In fact, fat-rich diets can cause dysbiosis, which decreases the number of protective bacteria and compromises the integrity of the epithelial barrier in the GIT. Overgrowth of pathogenic microbes then release higher quantities of toxic metabolites into the circulatory system, especially the pro-inflammatory cytokines detected in osteoarthritis (OA), thereby promoting inflammation and the initiation of many disease processes throughout the body. Although many studies link OA with GMB perturbations, further research is still needed.


Assuntos
Disbiose , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal , Osteoartrite , Animais , Disbiose/imunologia , Disbiose/microbiologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Osteoartrite/etiologia , Osteoartrite/imunologia , Osteoartrite/microbiologia
3.
Int J Mol Sci ; 23(3)2022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-35163596

RESUMO

Because both endotoxemia and gut dysbiosis post-splenectomy might be associated with systemic infection, the susceptibility against infection was tested by dextran sulfate solution (DSS)-induced colitis and lipopolysaccharide (LPS) injection models in splenectomy mice with macrophage experiments. Here, splenectomy induced a gut barrier defect (FITC-dextran assay, endotoxemia, bacteria in mesenteric lymph nodes, and the loss of enterocyte tight junction) and gut dysbiosis (increased Proteobacteria by fecal microbiome analysis) without systemic inflammation (serum IL-6). In parallel, DSS induced more severe mucositis in splenectomy mice than sham-DSS mice, as indicated by mortality, stool consistency, gut barrier defect, serum cytokines, and blood bacterial burdens. The presence of green fluorescent-producing (GFP) E. coli in the spleen of sham-DSS mice after an oral gavage supported a crucial role of the spleen in the control of bacteria from gut translocation. Additionally, LPS administration in splenectomy mice induced lower serum cytokines (TNF-α and IL-6) than LPS-administered sham mice, perhaps due to LPS tolerance from pre-existing post-splenectomy endotoxemia. In macrophages, LPS tolerance (sequential LPS stimulation) demonstrated lower cell activities than the single LPS stimulation, as indicated by the reduction in supernatant cytokines, pro-inflammatory genes (iNOS and IL-1ß), cell energy status (extracellular flux analysis), and enzymes of the glycolysis pathway (proteomic analysis). In conclusion, a gut barrier defect after splenectomy was vulnerable to enterocyte injury (such as DSS), which caused severe bacteremia due to defects in microbial control (asplenia) and endotoxemia-induced LPS tolerance. Hence, gut dysbiosis and gut bacterial translocation in patients with a splenectomy might be associated with systemic infection, and gut-barrier monitoring or intestinal tight-junction strengthening may be useful.


Assuntos
Bacteriemia/imunologia , Colite/imunologia , Sulfato de Dextrana/toxicidade , Disbiose/imunologia , Tolerância Imunológica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Esplenectomia , Animais , Colite/induzido quimicamente , Disbiose/induzido quimicamente , Masculino , Camundongos
4.
J Immunol Res ; 2022: 6839356, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35224112

RESUMO

Intestinal bacterial compositions of rheumatoid arthritis (RA) patients have been reported to be different from those of healthy people. Dysbiosis, imbalance of the microbiota, is widely known to cause gut barrier damage, resulting in an influx of bacteria and their substances into host bloodstreams in animal studies. However, few studies have investigated the effect of bacterial substances on the pathophysiology of RA. In this study, eighty-seven active RA patients who had inadequate responses to conventional synthetic disease-modifying antirheumatic drugs or severe comorbidities were analyzed for correlations between many factors such as disease activities, disease biomarkers, intestinal bacterial counts, fecal and serum lipopolysaccharide (LPS), LPS-binding protein (LBP), endotoxin neutralizing capacity (ENC), and serum antibacterial substance IgG and IgA antibody levels by multiple regression analysis with consideration for demographic factors such as age, sex, smoking, and methotrexate treatment. Serum LBP levels, fecal LPS levels, total bacteria counts, serum anti-LPS from Porphyromonas gingivalis (Pg-LPS) IgG antibody levels, and serum anti-Pg-LPS IgA antibody levels were selected for multiple regression analysis using Spearman's correlation analysis. Serum LBP levels were correlated with disease biomarker levels, such as erythrocyte sedimentation rate (p < 0.001), C-reactive protein (p < 0.001), matrix metalloproteinase-3 (p < 0.001), and IL-6 (p = 0.001), and were inversely correlated with hemoglobin (p = 0.005). Anti-Pg-LPS IgG antibody levels were inversely correlated with activity indices such as patient global assessments using visual analogue scale (VAS) (p = 0.002) and painVAS (p < 0.001). Total bacteria counts were correlated with ENC (p < 0.001), and inversely correlated with serum LPS (p < 0.001) and anti-Pg-LPS IgA antibody levels (p < 0.001). These results suggest that substances from oral and gut microbiota may influence disease activity in RA patients.


Assuntos
Artrite Reumatoide/microbiologia , Infecções por Bacteroidaceae/microbiologia , Disbiose/microbiologia , Boca/microbiologia , Porphyromonas gingivalis/fisiologia , Proteínas de Fase Aguda/metabolismo , Idoso , Artrite Reumatoide/imunologia , Autoanticorpos/sangue , Carga Bacteriana , Infecções por Bacteroidaceae/imunologia , Biomarcadores/metabolismo , Proteínas de Transporte/metabolismo , Estudos Transversais , Disbiose/imunologia , Feminino , Microbioma Gastrointestinal , Humanos , Imunoglobulina A/metabolismo , Lipopolissacarídeos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade
5.
Front Immunol ; 13: 1096551, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36726985

RESUMO

The gastrointestinal tract is inhabited by trillions of commensal microorganisms that constitute the gut microbiota. As a main metabolic organ, the gut microbiota has co-evolved in a symbiotic relationship with its host, contributing to physiological homeostasis. Recent advances have provided mechanistic insights into the dual role of the gut microbiota in cancer pathogenesis. Particularly, compelling evidence indicates that the gut microbiota exerts regulatory effects on the host immune system to fight against cancer development. Some microbiota-derived metabolites have been suggested as potential activators of antitumor immunity. On the contrary, the disequilibrium of intestinal microbial communities, a condition termed dysbiosis, can induce cancer development. The altered gut microbiota reprograms the hostile tumor microenvironment (TME), thus allowing cancer cells to avoid immunosurvelliance. Furthermore, the gut microbiota has been associated with the effects and complications of cancer therapy given its prominent immunoregulatory properties. Therapeutic measures that aim to manipulate the interplay between the gut microbiota and tumor immunity may bring new breakthroughs in cancer treatment. Herein, we provide a comprehensive update on the evidence for the implication of the gut microbiota in immune-oncology and discuss the fundamental mechanisms underlying the influence of intestinal microbial communities on systemic cancer therapy, in order to provide important clues toward improving treatment outcomes in cancer patients.


Assuntos
Microbioma Gastrointestinal , Neoplasias , Microambiente Tumoral , Humanos , Microbioma Gastrointestinal/imunologia , Sistema Imunitário , Neoplasias/imunologia , Resultado do Tratamento , Microambiente Tumoral/imunologia , Disbiose/imunologia
6.
Pharmacol Rep ; 74(1): 111-123, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34757518

RESUMO

BACKGROUND: Obesity is still a worldwide public health problem, requiring the development of adjuvant therapies to combat it. In this context, modulation of the intestinal microbiota seems prominent, given that the composition of the intestinal microbiota contributes to the outcome of this disease. The aim of this work is to investigate the treatment with an antimicrobial and/or a potential probiotic against overweight. METHODS: Male C57BL/6J mice were subjected to a 12-week overweight induction protocol. After that, 4-week treatment was started, with mice divided into four groups: control, treated with distilled water; potential probiotic, with Lactobacillus gasseri LG-G12; antimicrobial, with ceftriaxone; and antimicrobial + potential probiotic with ceftriaxone in the first 2 weeks and L. gasseri LG-G12 in the subsequent weeks. RESULTS: The treatment with ceftriaxone in isolated form or in combination with the potential probiotic provided a reduction in body fat. However, such effect is supposed to be a consequence of the negative action of ceftriaxone on the intestinal microbiota composition, and this intestinal dysbiosis may have contributed to the destruction of the intestinal villi structure, which led to a reduction in the absorptive surface. Also, the effects of L. gasseri LG-G12 apparently have been masked by the consumption of the high-fat diet. CONCLUSIONS: The results indicate that the use of a ceftriaxone in the adjuvant treatment of overweight is not recommended due to the potential risk of developing inflammatory bowel disease.


Assuntos
Ceftriaxona/farmacologia , Disbiose , Microbioma Gastrointestinal , Absorção Intestinal , Obesidade , Adjuvantes Farmacêuticos/farmacologia , Animais , Antibacterianos/farmacologia , Disbiose/induzido quimicamente , Disbiose/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Doenças Inflamatórias Intestinais/imunologia , Absorção Intestinal/efeitos dos fármacos , Absorção Intestinal/imunologia , Lactobacillus gasseri/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/tratamento farmacológico , Obesidade/microbiologia , Probióticos/farmacologia , Medição de Risco
7.
Dig Dis Sci ; 67(1): 42-55, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33527328

RESUMO

Inflammatory bowel disease (IBD) is a group of chronic intestinal inflammatory disorders with a prolonged duration characterized by recurrent relapse and remission. The exact etiology of IBD remains poorly understood despite the identification of relevant risk factors, including individual genetic susceptibility, environmental triggers, and disruption of immune homeostasis. Dysbiosis of the gut microbiota is believed to exacerbate the progression of IBD. Recently, increasing evidence has also linked oral microbiota dysbiosis with the development of IBD. On the one hand, IBD patients show significantly unbalanced composition and function of the oral microbiota known as dysbiosis. On the other, overabundances of oral commensal bacteria with opportunistic pathogenicity have been found in the gut microbiota of IBD patients. Herein, we review the current information on the causative factors of IBD, especially recent evidence of IBD-associated oral microbiota dysbiosis, which has seldom been covered in the previous literature review, highlighting the pathogenic mechanisms of specific oral bacteria in the development of IBD. Ectopic colonization of several oral bacteria, including a subset of Porphyromonas gingivalis, Streptococcus mutans, Fusobacterium nucleatum, Campylobacter concisus, and Klebsiella pneumoniae, may lead to destruction of the intestinal epithelial barrier, excessive secretion of inflammatory cytokines, disruption of the host immune system, and dysbiosis of gut microbiota, consequently aggravating chronic intestinal inflammation. Studying oral microbiota dysbiosis may open future horizons for understanding IBD pathogenesis and provide novel biomarkers for IBD. This review also presents the current treatment and new perspectives for IBD treatment.


Assuntos
Disbiose , Microbioma Gastrointestinal/fisiologia , Doenças Inflamatórias Intestinais , Boca/microbiologia , Disbiose/imunologia , Disbiose/microbiologia , Humanos , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/imunologia , Análise de Mediação
8.
Pediatr Res ; 91(1): 35-43, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33731809

RESUMO

Primary immunodeficiency diseases (PIDs) caused by a single-gene defect generally are referred to as monogenic autoimmune disorders. For example, mutations in the transcription factor autoimmune regulator (AIRE) result in a condition called autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy; while mutations in forkhead box P3 lead to regulatory T cell (Treg)-deficiency-induced multiorgan inflammation, which in humans is called "immune dysregulation, polyendocrinopathy, enteropathy with X-linked inheritance" (or IPEX syndrome). Previous studies concluded that monogenic diseases are insensitive to commensal microbial regulation because they develop even in germ-free (GF) animals, a conclusion that has limited the number of studies determining the role of microbiota in monogenic PIDs. However, emerging evidence shows that although the onset of the disease is independent of the microbiota, several monogenic PIDs vary in severity in association with the microbiome. In this review, we focus on monogenic PIDs associated with Treg deficiency/dysfunction, summarizing the gut microbial dysbiosis that has been shown to be linked to these diseases. From limited studies, we have gleaned several mechanistic insights that may prove to be of therapeutic importance in the early stages of life. IMPACT: This review paper serves to refute the concept that monogenic PIDs are not linked to the microbiome. The onset of monogenic PIDs is independent of microbiota; single-gene mutations such as AIRE or Foxp3 that affect central or peripheral immune tolerance produce monogenic diseases even in a GF environment. However, the severity and outcome of PIDs are markedly impacted by the microbial composition. We suggest that future research for these conditions may focus on targeting the microbiome.


Assuntos
Doenças Autoimunes/genética , Disbiose/imunologia , Microbioma Gastrointestinal , Linfócitos T Reguladores/imunologia , Doenças Autoimunes/imunologia , Humanos , Lactente , Recém-Nascido
9.
J. investig. allergol. clin. immunol ; 32(5): 327-344, 2022. ilus
Artigo em Inglês | IBECS | ID: ibc-212729

RESUMO

The role of the microbiome in the molecular mechanisms underlying allergy has become highly relevant in recent years. Studies areincreasingly suggesting that altered composition of the microbiota, or dysbiosis, may result in local and systemic alteration of the immuneresponse to specific allergens. In this regard, a link has been established between lung microbiota and respiratory allergy, between skinmicrobiota and atopic dermatitis, and between gut microbiota and food allergy.The composition of the human microbiota is dynamic and depends on host-associated factors such as diet, diseases, and lifestyle. Omics arethe techniques of choice for the analysis and understanding of the microbiota. Microbiota analysis techniques have advanced considerablyin recent decades, and the need for multiple approaches to explore and comprehend multifactorial diseases, including allergy, has increased.Thus, more and more studies are proposing mechanisms for intervention in the microbiota.In this review, we present the latest advances with respect to the human microbiota in the literature, focusing on the intestinal, cutaneous,and respiratory microbiota. We discuss the relationship between the microbiome and the immune system, with emphasis on allergic diseases.Finally, we discuss the main technologies for the study of the microbiome and interventions targeting the microbiota for prevention of allergy. (AU)


El papel del microbioma en los mecanismos moleculares de las enfermedades alérgicas se ha vuelto muy relevante en los últimos años.Cada vez más estudios sugieren que una composición alterada de la microbiota, o disbiosis, puede resultar en una alteración local ysistémica de la respuesta inmune a alérgenos específicos. En este sentido, se ha establecido un vínculo entre la microbiota pulmonar y laalergia respiratoria, así como la microbiota cutánea y el desarrollo de dermatitis atópica, y la microbiota intestinal y la alergia alimentaria.La composición de la microbiota humana es dinámica y depende de diversos factores asociados al huésped como la dieta, las enfermedadesy el estilo de vida, entre otros. Para el análisis y comprensión de la microbiota, las ómicas son las técnicas de elección. En las últimasdécadas, las técnicas de análisis de microbiota han tenido un gran avance y han aumentado la necesidad de múltiples enfoques paraexplorar y comprender las enfermedades multifactoriales, incluidas las enfermedades alérgicas. De esta manera, cada vez son más losestudios que proponen mecanismos de intervención sobre la microbiota de pacientes.En esta revisión, presentamos los últimos avances encontrados en la literatura sobre la microbiota humana, centrándose en las microbiotasintestinal, cutánea y respiratoria. Discutimos la relación entre el microbioma y el sistema inmunológico, con especial énfasis en lasenfermedades alérgicas. Finalmente, discutimos las principales tecnologías para el estudio del microbioma y los estudios de intervencióndirigidos a la microbiota propuestos para la prevención de alergias. (AU)


Assuntos
Humanos , Hipersensibilidade Alimentar , Microbioma Gastrointestinal/imunologia , Alérgenos/imunologia , Disbiose/imunologia
10.
Front Immunol ; 12: 796865, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925383

RESUMO

Besides its contribution to the development of rheumatic diseases, the gut microbiota interact with anti-rheumatic drugs. The intestinal microbiota can directly metabolize many drugs and indirectly change drug metabolism through a complex multi-dimensional interaction with the host, thus affecting individual response to drug therapy and adverse effects. The focus of the current review is to address recent advances and important progress in our understanding of how the gut microbiota interact with anti-rheumatic drugs and provide perspectives on promoting precision treatment, drug discovery, and better therapy for rheumatic diseases.


Assuntos
Antirreumáticos/uso terapêutico , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Animais , Humanos , Medicina de Precisão , Doenças Reumáticas/tratamento farmacológico
11.
Front Immunol ; 12: 799788, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925385

RESUMO

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that was traditionally thought to be closely related to genetic and environmental risk factors. Although treatment options for SLE with hormones, immunosuppressants, and biologic drugs are now available, the rates of clinical response and functional remission of these drugs are still not satisfactory. Currently, emerging evidence suggests that gut microbiota dysbiosis may play crucial roles in the occurrence and development of SLE, and manipulation of targeting the gut microbiota holds great promises for the successful treatment of SLE. The possible mechanisms of gut microbiota dysbiosis in SLE have not yet been well identified to date, although they may include molecular mimicry, impaired intestinal barrier function and leaky gut, bacterial biofilms, intestinal specific pathogen infection, gender bias, intestinal epithelial cells autophagy, and extracellular vesicles and microRNAs. Potential therapies for modulating gut microbiota in SLE include oral antibiotic therapy, fecal microbiota transplantation, glucocorticoid therapy, regulation of intestinal epithelial cells autophagy, extracellular vesicle-derived miRNA therapy, mesenchymal stem cell therapy, and vaccination. This review summarizes novel insights into the mechanisms of microbiota dysbiosis in SLE and promising therapeutic strategies, which may help improve our understanding of the pathogenesis of SLE and provide novel therapies for SLE.


Assuntos
Disbiose/complicações , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Lúpus Eritematoso Sistêmico/microbiologia , Animais , Humanos , Lúpus Eritematoso Sistêmico/imunologia
12.
Front Immunol ; 12: 759679, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34868000

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder, affecting 1-2% of the population aged 65 and over. Additionally, non-motor symptoms such as pain and gastrointestinal dysregulation are also common in PD. These impairments might stem from a dysregulation within the gut-brain axis that alters immunity and the inflammatory state and subsequently drives neurodegeneration. There is increasing evidence linking gut dysbiosis to the severity of PD's motor symptoms as well as to somatosensory hypersensitivities. Altogether, these interdependent features highlight the urgency of reviewing the links between the onset of PD's non-motor symptoms and gut immunity and whether such interplays drive the progression of PD. This review will shed light on maladaptive neuro-immune crosstalk in the context of gut dysbiosis and will posit that such deleterious interplays lead to PD-induced pain hypersensitivity.


Assuntos
Disbiose/imunologia , Dor/imunologia , Doença de Parkinson/imunologia , Humanos
13.
J Immunol Res ; 2021: 5578958, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869782

RESUMO

BACKGROUND: As a transitional state between normal aging and Alzheimer's disease (AD), mild cognitive impairment (MCI) is characterized by a worse cognitive decline than that of natural aging. The association between AD and gut microbiota has been reported in a number of studies; however, microbial research regarding MCI remains limited. METHODS: This study examined 48 participants, of whom 22 were MCI cases and 26 were normal control cases. Fecal samples were collected for 16S ribosomal RNA (rRNA) quantitative arrays and bioinformatics analysis. RESULTS: A principal coordinates analysis (PCoA) and nonmetric multidimensional scaling (NMDS) both demonstrated that the microbial composition of participants with MCI deviated from that of healthy control participants. Multiple bacterial species were significantly increased (e.g., Staphylococcus intermedius) or decreased (e.g., Bacteroides salyersiae) in samples from the MCI group. CONCLUSION: The composition of gut microbiota differed between normal control and MCI cases. This is the first study to identify a signature series of species in the gut microbiota of individuals with MCI. The results provide a new direction for the future development of an early diagnosis and probiotic regimen.


Assuntos
Envelhecimento/imunologia , Disfunção Cognitiva/imunologia , Disbiose/complicações , Microbioma Gastrointestinal/imunologia , Idoso , Estudos de Casos e Controles , Disfunção Cognitiva/microbiologia , Disfunção Cognitiva/prevenção & controle , Disbiose/dietoterapia , Disbiose/imunologia , Disbiose/microbiologia , Fezes/microbiologia , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Probióticos/administração & dosagem
14.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34884634

RESUMO

Chronic alcohol abuse causes an inflammatory response in the intestinal tract with damage to the integrity of the mucosa and epithelium, as well as dysbiosis in the gut microbiome. However, the role of gut bacteria in ethanol effects and how these microorganisms interact with the immune system are not well understood. The aim of the present study was to evaluate if TLR4 alters the ethanol-induced intestinal inflammatory response, and whether the response of this receptor affects the gut microbiota profile. We analyzed the 16S rRNA sequence of the fecal samples from wild-type (WT) and TLR4-knockout (TLR4-KO) mice with and without ethanol intake for 3 months. The results demonstrated that chronic ethanol consumption reduces microbiota diversity and causes dysbiosis in WT mice. Likewise, ethanol upregulates several inflammatory genes (IL-1ß, iNOS, TNF-α) and miRNAs (miR-155-5p, miR-146a-5p) and alters structural and permeability genes (INTL1, CDH1, CFTR) in the colon of WT mice. Our results further demonstrated that TLR4-KO mice exhibit a different microbiota that can protect against the ethanol-induced activation of the immune system and colon integrity dysfunctions. In short, our results reveal that TLR4 is a key factor for determining the gut microbiota, which can participate in dysbiosis and the inflammatory response induced by alcohol consumption.


Assuntos
Alcoolismo/microbiologia , Microbioma Gastrointestinal , Mucosa Intestinal/imunologia , Receptor 4 Toll-Like/deficiência , Alcoolismo/imunologia , Alcoolismo/metabolismo , Animais , Depressores do Sistema Nervoso Central/toxicidade , Modelos Animais de Doenças , Disbiose/imunologia , Disbiose/metabolismo , Disbiose/microbiologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 4 Toll-Like/metabolismo
15.
Int J Mol Sci ; 22(23)2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34884961

RESUMO

Lipocalin 2 (LCN2) mediates key roles in innate immune responses. It has affinity for many lipophilic ligands and binds various siderophores, thereby limiting bacterial growth by iron sequestration. Furthermore, LCN2 protects against obesity and metabolic syndrome by interfering with the composition of gut microbiota. Consequently, complete or hepatocyte-specific ablation of the Lcn2 gene is associated with higher susceptibility to bacterial infections. In the present study, we comparatively profiled microbiota in fecal samples of wild type and Lcn2 null mice and show, in contrast to previous reports, that the quantity of DNA in feces of Lcn2 null mice is significantly lower than that in wild type mice (p < 0.001). By using the hypervariable V4 region of the 16S rDNA gene and Next-Generation Sequencing methods, we found a statistically significant change in 16 taxonomic units in Lcn2-/- mice, including eight gender-specific deviations. In particular, members of Clostridium, Escherichia, Helicobacter, Lactococcus, Prevotellaceae_UCG-001 and Staphylococcus appeared to expand in the intestinal tract of knockout mice. Interestingly, the proportion of Escherichia (200-fold) and Staphylococcus (10-fold) as well as the abundance of intestinal bacteria encoding the LCN2-sensitive siderphore enterobactin (entA) was significantly increased in male Lcn2 null mice (743-fold, p < 0.001). This was accompanied by significant higher immune cell infiltration in the ileum as demonstrated by increased immunoreactivity against the pan-leukocyte protein CD45, the lymphocyte transcription factor MUM-1/IRF4, and the macrophage antigen CD68/Macrosialin. In addition, we found a higher expression of mucosal mast cell proteases indicating a higher number of those innate immune cells. Finally, the ileum of Lcn2 null mice displayed a high abundance of segmented filamentous bacteria, which are intimately associated with the mucosal cell layer, provoking epithelial antimicrobial responses and affecting T-helper cell polarization.


Assuntos
Bactérias/classificação , Disbiose/microbiologia , Lipocalina-2/genética , Mutação com Perda de Função , Análise de Sequência de DNA/métodos , Animais , Bactérias/genética , Bactérias/isolamento & purificação , DNA Bacteriano/genética , DNA Ribossômico/genética , Modelos Animais de Doenças , Disbiose/genética , Disbiose/imunologia , Fezes/microbiologia , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos , Camundongos Knockout , Filogenia , RNA Ribossômico 16S/genética , Fatores Sexuais
16.
Front Immunol ; 12: 765965, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721437

RESUMO

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulted in an unprecedented global crisis. Although primarily a respiratory illness, dysregulated immune responses may lead to multi-organ dysfunction. Prior data showed that the resident microbial communities of gastrointestinal and respiratory tracts act as modulators of local and systemic inflammatory activity (the gut-lung axis). Evolving evidence now signals an alteration in the gut microbiome, brought upon either by cytokines from the infected respiratory tract or from direct infection of the gut, or both. Dysbiosis leads to a "leaky gut". The intestinal permeability then allows access to bacterial products and toxins into the circulatory system and further exacerbates the systemic inflammatory response. In this review, we discuss the available data related to the role of the gut microbiome in the development and progression of COVID-19. We provide mechanistic insights into early data with a focus on immunological crosstalk and the microbiome's potential as a biomarker and therapeutic target.


Assuntos
COVID-19/microbiologia , Síndrome da Liberação de Citocina/microbiologia , Disbiose/microbiologia , Microbioma Gastrointestinal/imunologia , SARS-CoV-2/fisiologia , COVID-19/imunologia , Síndrome da Liberação de Citocina/imunologia , Disbiose/imunologia , Humanos , Imunidade , Inflamação
17.
J Immunol Res ; 2021: 4973589, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34722779

RESUMO

METHOD: This study included 74 Chinese male patients with HCC. They were divided into early (n = 19), intermediate (n = 37), and terminal (n = 18) groups, referred to as Barcelona Clinic Liver Cancer stage 0+A, B, and C+D, respectively. Paired fecal and plasma samples were collected. Microbial composition and profiles were analyzed by 16S rRNA gene sequencing. The levels of gut damage marker (regenerating islet-derived protein 3α (REG3α)) and microbial translocation markers (soluble CD14 (sCD14), lipopolysaccharide-binding protein (LBP), peptidoglycan recognition proteins (PGRPs)) were determined in plasma samples of patients by ELISA. Twenty plasma cytokine and chemokines were determined by Luminex. RESULTS: In early, intermediate, and terminal groups, the abundance of the Bifidobacteriaceae family decreased significantly (3.52%, 1.55%, and 0.56%, respectively, P = 0.003), while the abundance of the Enterococcaceae family increased significantly (1.6%, 2.9%, and 13.4%, respectively, P = 0.022). Levels of REG3α and sCD14 were markedly elevated only in the terminal group compared with the early (P = 0.025 and P = 0.048) and intermediate groups (P = 0.023 and P = 0.046). The level of LBP significantly increased in the intermediate (P = 0.035) and terminal (P = 0.025) groups compared with the early group. The PGRP levels were elevated only in the terminal group compared with the early group (P = 0.018). The ratio of Enterococcaceae to Bifidobacteriaceae was significantly associated with the levels of REG3α, LBP, sCD14, and PGRPs. With HCC progression, increased levels of inflammatory cytokines accompanied by a T cell-immunosuppressive response and microbial translocation were observed. CONCLUSION: Gut microbiota compositional and functional shift, together with elevated gut damage and microbial translocation, may promote HCC development by stimulating inflammatory response and suppressing T cell response.


Assuntos
Translocação Bacteriana/imunologia , Carcinoma Hepatocelular/imunologia , Disbiose/complicações , Microbioma Gastrointestinal/imunologia , Neoplasias Hepáticas/imunologia , Actinobacteria/genética , Actinobacteria/imunologia , Actinobacteria/isolamento & purificação , Idoso , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/microbiologia , Carcinoma Hepatocelular/patologia , DNA Bacteriano/isolamento & purificação , Progressão da Doença , Disbiose/diagnóstico , Disbiose/imunologia , Disbiose/microbiologia , Enterococcaceae/genética , Enterococcaceae/imunologia , Enterococcaceae/isolamento & purificação , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/microbiologia , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , RNA Ribossômico 16S
18.
J Immunol Res ; 2021: 8124593, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34722781

RESUMO

OBJECTIVE: Periodontitis is an inflammatory disease of microbial etiology caused primarily by dysbiosis of the oral microbiota. Our aim was to compare variations in the composition of the oral microbiomes of youths with severe periodontitis according to gender. METHODS: Subgingival plaque samples collected from 17 patients with severe periodontitis (11 males and 6 females) were split for 16S rRNA gene sequencing. The composition, α-diversity, and ß-diversity of the patients' oral microbiomes were compared between the males and the females. Linear discriminant analysis effect size (LEfSe) was used to analyze the specific taxa enriched in the two groups. Functional profiles (KEGG pathways) were obtained using PICRUSt based on 16S rRNA gene sequencing data. RESULTS: The Chao1 index and phylogenetic diversity whole tree were significantly higher in males than in females. The Simpson and Shannon indices were not significantly different between the two groups. ß-Diversity suggested that the samples were reasonably divided into groups. The Kruskal-Wallis test based on the relative abundance of species, combined with the LEfSe analysis showed that the dominant bacteria in males were Pseudomonas and Papillibacter, whereas the dominant bacteria in women were Fusobacteriales and Tannerella. KEGG analysis predicted that the variation in the oral microbiome may be related to the immune system in women, whereas immune system diseases were the dominant pathway in men. CONCLUSION: We found sex-specific differences in the oral microbiome in a sample of youths with severe periodontitis. The differences may be related to changes in immune homeostasis and lead to a better understanding of periodontitis.


Assuntos
Disbiose/diagnóstico , Microbiota/imunologia , Mucosa Bucal/microbiologia , Periodontite/microbiologia , Adulto , DNA Bacteriano/isolamento & purificação , Disbiose/complicações , Disbiose/imunologia , Disbiose/microbiologia , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Microbiota/genética , Periodontite/diagnóstico , Periodontite/imunologia , Filogenia , RNA Ribossômico 16S/genética , Índice de Gravidade de Doença , Fatores Sexuais
19.
Front Immunol ; 12: 747914, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745119

RESUMO

The human body and its microbiome constitute a highly delicate system. The gut microbiome participates in the absorption of the host's nutrients and metabolism, maintains the microcirculation, and modulates the immune response. Increasing evidence shows that gut microbiome dysbiosis in the body not only affects the occurrence and development of tumors but also tumor prognosis and treatment. Microbiome have been implicated in tumor control in patients undergoing anti- angiogenesis therapy and immunotherapy. In cases with unsatisfactory responses to chemotherapy, radiotherapy, and targeted therapy, appropriate adjustment of microbes abundance is considered to enhance the treatment response. Here, we review the current research progress in cancer immunotherapy and anti- angiogenesis therapy, as well as the unlimited potential of their combination, especially focusing on how the interaction between intestinal microbiota and the immune system affects cancer pathogenesis and treatment. In addition, we discuss the effects of microbiota on anti-cancer immune response and anti- angiogenesis therapy, and the potential value of these interactions in promoting further research in this field.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Imunoterapia , Microbiota , Neoplasias/terapia , Inibidores da Angiogênese/farmacologia , Carcinogênese/imunologia , Ensaios Clínicos Fase III como Assunto , Terapia Combinada , Dieta , Medicamentos de Ervas Chinesas/farmacologia , Disbiose/imunologia , Disbiose/metabolismo , Humanos , Inibidores de Checkpoint Imunológico , Microbiota/efeitos dos fármacos , Microbiota/imunologia , Microbiota/fisiologia , Neoplasias/irrigação sanguínea , Neoplasias/imunologia , Neoplasias/microbiologia , Probióticos , Simbiose , Evasão Tumoral
20.
Front Immunol ; 12: 766296, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745141

RESUMO

Cyclophosphamide (CTX), a common anticancer drug, can cause a variety of side effects such as immunosuppression and intestinal mucosal injury. Polysaccharides are the major bioactive components of the roots of Millettia Speciosa Champ and have gained attention for their immunomodulatory activity. This study was designed to evaluate the immunomodulatory effect of Millettia Speciosa Champ polysaccharide (MSCP) on CTX-induced mice and the possible mechanism. The results showed that MSCP attenuated the CTX-induced decrease in body weight and immune organ indices in mice and promoted the secretion of immune-related cytokines (IL-2, IL-4, IL-10, TNF-α, and IgG). Meanwhile, MSCP restored intestinal morphology, increased the ratio of villus height/crypt depth (V/C), and improved the number of goblet cells and mucins expression. At the mRNA level, MSCP activated the TLRs/MyD88/NF-κB p65 pathway and enhanced the expression of genes related to intestinal mucosal integrity (Occludin1, Claudin1, and MUC-2). In addition, MSCP as a prebiotic improved microbial community diversity, regulated the relative abundance of dominant microbiota from the phylum level to the genus level, restored CTX-induced gut microbial dysbiosis, and promoted short-chain fatty acid production in mice. Based on the present findings, MSCP may modulate the immune response depending on enhancing intestinal health, suggesting that MSCP holds promise as a promising immunostimulant in functional foods and drugs.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Ciclofosfamida/efeitos adversos , Disbiose/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Imunossupressores/efeitos adversos , Millettia , Polissacarídeos/uso terapêutico , Prebióticos , Adjuvantes Imunológicos/farmacologia , Animais , Citocinas/sangue , Disbiose/induzido quimicamente , Disbiose/imunologia , Disbiose/microbiologia , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Feminino , Imunoglobulina G/sangue , Terapia de Imunossupressão , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Intestinos/patologia , Masculino , Camundongos , Raízes de Plantas , Polissacarídeos/farmacologia , Baço/efeitos dos fármacos , Baço/patologia , Timo/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...