Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 319
Filtrar
1.
Science ; 377(6601): eabp9960, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35771903

RESUMO

Changes in the composition of the gut microbiota are associated with many human diseases. So far, however, we have failed to define homeostasis or dysbiosis by the presence or absence of specific microbial species. The composition and function of the adult gut microbiota is governed by diet and host factors that regulate and direct microbial growth. The host delivers oxygen and nitrate to the lumen of the small intestine, which selects for bacteria that use respiration for energy production. In the colon, by contrast, the host limits the availability of oxygen and nitrate, which results in a bacterial community that specializes in fermentation for growth. Although diet influences microbiota composition, a poor diet weakens host control mechanisms that regulate the microbiota. Hence, quantifying host parameters that control microbial growth could help define homeostasis or dysbiosis and could offer alternative strategies to remediate dysbiosis.


Assuntos
Bactérias , Colo , Disbiose , Microbioma Gastrointestinal , Homeostase , Intestino Delgado , Bactérias/metabolismo , Colo/microbiologia , Colo/fisiopatologia , Disbiose/microbiologia , Disbiose/fisiopatologia , Interações entre Hospedeiro e Microrganismos , Humanos , Intestino Delgado/microbiologia , Intestino Delgado/fisiopatologia , Nitratos/metabolismo , Oxigênio/metabolismo
2.
World J Gastroenterol ; 28(12): 1204-1219, 2022 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-35431513

RESUMO

Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder in which recurrent abdominal pain is associated with defecation or a change in bowel habits (constipation, diarrhea, or both), and it is often accompanied by symptoms of abdominal bloating and distension. IBS is an important health care issue because it negatively affects the quality of life of patients and places a considerable financial burden on health care systems. Despite extensive research, the etiology and underlying pathophysiology of IBS remain incompletely understood. Proposed mechanisms involved in its pathogenesis include increased intestinal permeability, changes in the immune system, visceral hypersensitivity, impaired gut motility, and emotional disorders. Recently, accumulating evidence has highlighted the important role of the gut microbiota in the development of IBS. Microbial dysbiosis within the gut is thought to contribute to all aspects of its multifactorial pathogenesis. The last few decades have also seen an increasing interest in the impact of antibiotics on the gut microbiota. Moreover, antibiotics have been suggested to play a role in the development of IBS. Extensive research has established that antibacterial therapy induces remarkable shifts in the bacterial community composition that are quite similar to those observed in IBS. This suggestion is further supported by data from cohort and case-control studies, indicating that antibiotic treatment is associated with an increased risk of IBS. This paper summarizes the main findings on this issue and contributes to a deeper understanding of the link between antibiotic use and the development of IBS.


Assuntos
Antibacterianos , Disbiose , Microbioma Gastrointestinal , Síndrome do Intestino Irritável , Antibacterianos/efeitos adversos , Disbiose/etiologia , Disbiose/microbiologia , Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Humanos , Síndrome do Intestino Irritável/etiologia , Síndrome do Intestino Irritável/microbiologia , Síndrome do Intestino Irritável/fisiopatologia , Qualidade de Vida
3.
BMC Nephrol ; 23(1): 10, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34979948

RESUMO

BACKGROUND: The pathogenesis of Henoch-Schönlein purpura nephritis (HSPN) is closely associated with mucosal infection. But whether intestinal microbiota dysbiosis plays a role in it is not clear. METHODS: A total of 52 participants including 26 HSPN patients and 26 healthy controls were included. By using 16S ribosomal RNA gene sequencing, the intestinal microbiota composition between HSPN and healthy controls was compared. The diagnostic potency was evaluated by Receiver operating characteristic (ROC) with area under curves (AUC). Meanwhile, correlation analysis was also performed. RESULTS: The lower community richness and diversity of fecal microbiota was displayed in HSPN patients and the structure of gut microbiota was remarkedly different. A genus-level comparison indicated a significant increase in the proportions of g-Bacteroides, g-Escherichia-Shigella and g-Streptococcus, and a marked reduction of g-Prevotella_9 in HSPN patients, suggesting that the overrepresentation of potential pathogens and reduction of profitable strains were the main feature of the dysbiosis. The differential taxonomic abundance might make sense for distinguishing HSPN from healthy controls, with AUC of 0.86. The relative abundance of the differential bacteria was also concerned with clinical indices. Among them, Streptococcus spp. was positively associated with the severity of HSPN (P < 0.050). It was found that HSPN patients with higher level of Streptococcus spp. were more likely to suffering from hematuria and hypoalbuminemia (P < 0.050). CONCLUSIONS: The dysbiosis of gut microbiota was obvious in HSPN patients, and the intestinal mucosal streptococcal infection was distinctive, which was closely related to its severity.


Assuntos
Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Vasculite por IgA/microbiologia , Vasculite por IgA/fisiopatologia , Streptococcus/isolamento & purificação , Adulto , Estudos de Casos e Controles , Fezes/microbiologia , Feminino , Humanos , Vasculite por IgA/diagnóstico , Masculino , RNA Ribossômico 16S , Análise de Sequência de RNA , Índice de Gravidade de Doença
4.
J Exp Clin Cancer Res ; 41(1): 31, 2022 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-35065652

RESUMO

The role of the microbiota in human health and disease is well established, including its effects on several cancer types. However, the role of microbial dysbiosis in prostate cancer development, progression, and response to treatment is less well understood. This knowledge gap could perhaps be implicated in the lack of better risk stratification and prognostic tools that incorporate risk factors such as bacterial infections and inflammatory signatures. With over a decade's research investigating associations between microbiome and prostate carcinogenesis, we are ever closer to finding the crucial biological link between the two. Yet, definitive answers remain elusive, calling for continued research into this field. In this review, we outline the three frequently used NGS based analysis methodologies that are used for microbiome profiling, thereby serving as a quick guide for future microbiome research. We next provide a detailed overview of the current knowledge of the role of the human microbiome in prostate cancer development, progression, and treatment response. Finally, we describe proposed mechanisms of host-microbe interactions that could lead to prostate cancer development, progression or treatment response.


Assuntos
Disbiose/fisiopatologia , Microbiota/fisiologia , Neoplasias da Próstata/fisiopatologia , Humanos , Masculino
5.
Tissue Barriers ; 10(3): 2000299, 2022 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-34775911

RESUMO

The gut-brain axis hypothesis suggests that interactions in the intestinal milieu are critically involved in regulating brain function. Several studies point to a gut-microbiota-brain connection linking an impaired intestinal barrier and altered gut microbiota composition to neurological disorders involving neuroinflammation. Increased gut permeability allows luminal antigens to cross the gut epithelium, and via the blood stream and an impaired blood-brain barrier (BBB) enters the brain impacting its function. Pre-haptoglobin 2 (pHP2), the precursor protein to mature HP2, is the first characterized member of the zonulin family of structurally related proteins. pHP 2 has been identified in humans as the thus far only endogenous regulator of epithelial and endothelial tight junctions (TJs). We have leveraged the Zonulin-transgenic mouse (Ztm) that expresses a murine pHP2 (zonulin) to determine the role of increased gut permeability and its synergy with a dysbiotic intestinal microbiota on brain function and behavior. Here we show that Ztm mice display sex-dependent behavioral abnormalities accompanied by altered gene expression of BBB TJs and increased expression of brain inflammatory genes. Antibiotic depletion of the gut microbiota in Ztm mice downregulated brain inflammatory markers ameliorating some anxiety-like behavior. Overall, we show that zonulin-dependent alterations in gut permeability and dysbiosis of the gut microbiota are associated with an altered BBB integrity, neuroinflammation, and behavioral changes that are partially ameliorated by microbiota depletion. Our results suggest the Ztm model as a tool for the study of the cross-talk between the microbiome/gut and the brain in the context of neurobehavioral/neuroinflammatory disorders.


Assuntos
Comportamento Animal , Barreira Hematoencefálica , Microbioma Gastrointestinal , Haptoglobinas , Animais , Disbiose/fisiopatologia , Haptoglobinas/genética , Humanos , Mucosa Intestinal/fisiopatologia , Camundongos , Camundongos Transgênicos , Precursores de Proteínas/genética
6.
Shock ; 57(1): 15-23, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34726875

RESUMO

ABSTRACT: Sepsis is currently defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. The skeletal muscle system is among the host organ systems compromised by sepsis. The resulting neuromuscular dysfunction and impaired regenerative capacity defines sepsis-induced myopathy and manifests as atrophy, loss of strength, and hindered regeneration after injury. These outcomes delay recovery from critical illness and confer increased vulnerability to morbidity and mortality. The mechanisms underlying sepsis-induced myopathy, including the potential contribution of peripheral organs, remain largely unexplored. The gut microbiome is an immunological and homeostatic entity that interacts with and controls end-organ function, including the skeletal muscle system. Sepsis induces alterations in the gut microbiota composition, which is globally termed a state of "dysbiosis" for the host compared to baseline microbiota composition. In this review, we critically evaluate existing evidence and potential mechanisms linking sepsis-induced myopathy with gut microbiota dysbiosis.


Assuntos
Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Doenças Musculares/fisiopatologia , Sepse/fisiopatologia , Humanos , Músculo Esquelético/metabolismo , Regeneração/fisiologia , Sarcopenia/fisiopatologia , Células Satélites de Músculo Esquelético/fisiologia , Nicho de Células-Tronco/fisiologia
7.
Am J Clin Dermatol ; 23(1): 27-36, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34687433

RESUMO

Diet has long been understood to have an intricate association with atopic dermatitis, although much remains unelucidated. Skin barrier dysfunction with dysbiosis and consequent impairment of immune tolerance likely underly the pathogenesis of coincident atopic dermatitis and food allergy. There is a wide range of possible skin reactions to food, complicating the diagnosis and understanding of food allergies. Many patients, parents, and providers incorrectly suspect diet as causative of atopic dermatitis symptoms and many have tried elimination diets. This frequently leads to inaccurate labeling of food allergies, contributing to a dangerous spiral of inappropriate testing, referrals, and dietary changes, while neglecting established atopic dermatitis treatment essentials. Alternatively, certain dietary supplements or the introduction of certain foods may be beneficial for atopic dermatitis management or prevention. Greater consensus on the role of diet among providers of patients with atopic dermatitis is strongly encouraged to improve the management of atopic dermatitis.


Assuntos
Dermatite Atópica/dietoterapia , Dieta , Alérgenos/análise , Cannabis , Dermatite Atópica/fisiopatologia , Suplementos Nutricionais , Disbiose/fisiopatologia , Epitélio/fisiopatologia , Hipersensibilidade Alimentar/diagnóstico , Histidina/uso terapêutico , Humanos , Extratos Vegetais/uso terapêutico , Guias de Prática Clínica como Assunto , Chá
8.
Mucosal Immunol ; 15(1): 10-26, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34316007

RESUMO

Crohn's disease (CD) and ulcerative colitis (UC) are both characterized by chronic inflammation and severe dysfunction of the gastrointestinal tract. These two forms of inflammatory bowel disease (IBD) represent distinct clinical disorders with diverse driving mechanisms; however, this divergence is not reflected in currently approved therapeutics that commonly target general proinflammatory pathways. A compelling need therefore remains to understand factors that differentiate the topology and the distinct clinical manifestations of CD versus UC, in order to develop more effective and specialized therapies. Animal models provide valuable platforms for studying IBD heterogeneity and deciphering disease-specific mechanisms. Both the established and the newly developed ileitis mouse models are characterized by various disease initiating mechanisms and diverse phenotypic outcomes that reflect the complexity of human CD-ileitis. Microbial dysbiosis, destruction of epithelial barrier integrity, immune cell deregulation, as well as the recently described genome instability and stromal cell activation have all been proposed as the triggering factors for the development of ileitis-associated pathology. In this review, we aim to critically evaluate the mechanistic underpinnings of murine models of CD-ileitis, discuss their phenotypic similarities to human disease, and envisage their further exploitation for the development of novel targeted and personalized therapeutics.


Assuntos
Doença de Crohn/fisiopatologia , Disbiose/fisiopatologia , Ileíte/fisiopatologia , Animais , Doença de Crohn/terapia , Modelos Animais de Doenças , Disbiose/terapia , Humanos , Ileíte/terapia , Inflamação , Camundongos , Fenótipo
9.
Gut ; 71(1): 78-87, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33504491

RESUMO

OBJECTIVE: Effects of the diet-induced gut microbiota dysbiosis reach far beyond the gut. We aim to uncover the direct evidence involving the gut-testis axis in the aetiology of impaired spermatogenesis. DESIGN: An excessive-energy diet-induced metabolic syndrome (MetS) sheep model was established. The testicular samples, host metabolomes and gut microbiome were analysed. Faecal microbiota transplantation (FMT) confirmed the linkage between gut microbiota and spermatogenesis. RESULTS: We demonstrated that the number of arrested spermatogonia was markedly elevated by using 10× single-cell RNA-seq in the MetS model. Furthermore, through using metabolomics profiling and 16S rDNA-seq, we discovered that the absorption of vitamin A in the gut was abolished due to a notable reduction of bile acid levels, which was significantly associated with reduced abundance of Ruminococcaceae_NK4A214_group. Notably, the abnormal metabolic effects of vitamin A were transferable to the testicular cells through the circulating blood, which contributed to abnormal spermatogenesis, as confirmed by FMT. CONCLUSION: These findings define a starting point for linking the testicular function and regulation of gut microbiota via host metabolomes and will be of potential value for the treatment of male infertility in MetS.


Assuntos
Microbioma Gastrointestinal/fisiologia , Síndrome Metabólica/fisiopatologia , Espermatogênese/fisiologia , Testículo/fisiologia , Vitamina A/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Modelos Animais de Doenças , Disbiose/fisiopatologia , Masculino , Metaboloma , Ovinos
10.
Int J Obes (Lond) ; 46(1): 30-38, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34471225

RESUMO

BACKGROUND: Functional connectivity alterations in the lateral and medial hypothalamic networks have been associated with the development and maintenance of obesity, but the possible impact on the structural properties of these networks remains largely unexplored. Also, obesity-related gut dysbiosis may delineate specific hypothalamic alterations within obese conditions. We aim to assess the effects of obesity, and obesity and gut-dysbiosis on the structural covariance differences in hypothalamic networks, executive functioning, and depressive symptoms. METHODS: Medial (MH) and lateral (LH) hypothalamic structural covariance alterations were identified in 57 subjects with obesity compared to 47 subjects without obesity. Gut dysbiosis in the subjects with obesity was defined by the presence of high (n = 28) and low (n = 29) values in a BMI-associated microbial signature, and posthoc comparisons between these groups were used as a proxy to explore the role of obesity-related gut dysbiosis on the hypothalamic measurements, executive function, and depressive symptoms. RESULTS: Structural covariance alterations between the MH and the striatum, lateral prefrontal, cingulate, insula, and temporal cortices are congruent with previously functional connectivity disruptions in obesity conditions. MH structural covariance decreases encompassed postcentral parietal cortices in the subjects with obesity and gut-dysbiosis, but increases with subcortical nuclei involved in the coding food-related hedonic information in the subjects with obesity without gut-dysbiosis. Alterations for the structural covariance of the LH in the subjects with obesity and gut-dysbiosis encompassed increases with frontolimbic networks, but decreases with the lateral orbitofrontal cortex in the subjects with obesity without gut-dysbiosis. Subjects with obesity and gut dysbiosis showed higher executive dysfunction and depressive symptoms. CONCLUSIONS: Obesity-related gut dysbiosis is linked to specific structural covariance alterations in hypothalamic networks relevant to the integration of somatic-visceral information, and emotion regulation.


Assuntos
Disbiose/complicações , Doenças Hipotalâmicas/etiologia , Vias Neurais/fisiologia , Obesidade/complicações , Obesidade/fisiopatologia , Adulto , Índice de Massa Corporal , Estudos Transversais , Disbiose/fisiopatologia , Feminino , Humanos , Hipotálamo/fisiopatologia , Masculino , Pessoa de Meia-Idade , Vias Neurais/anormalidades
11.
Eur Rev Med Pharmacol Sci ; 25(1 Suppl): 101-107, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34890040

RESUMO

OBJECTIVE: The aim of the study was to show the importance of developing techniques that could exploit the potential of bacteriophages as therapeutics or food supplements. MATERIALS AND METHODS: PubMed database was searched using the following combination of keywords: (bacteriophage) AND (human therapy); (natural bacteriophage) AND (application). RESULTS: The increasing antibiotic resistance of many bacterial strains is making standard antibiotic treatments less effective. Phage therapy provides a non-antibiotic alternative with greater specificity and without harmful effects on the human microbiota. Phages target their specific bacteria, replicate, and then, destroy the host pathogen. Bacteriophages may be administered by several routes, including topical, oral and intravenous. They not only destroy the host pathogen but, in some cases, increase the sensitivity of host bacteria to antibiotics. Various studies have shown that combining phage therapy and antibiotic treatment can be effective against bacterial infections. Clinical trials of phage therapy have shown promising results for various human diseases and conditions. With advances in genetic engineering and molecular techniques, bacteriophages will be able to target a wide range of bacteria. CONCLUSIONS: In the future, phage therapy promises to become an effective therapeutic option for bacterial infections. Since many potentially beneficial bacteriophages can be found in food, supplements containing bacteriophages could be designed to remodel gut microbiota and eliminate pathogenic bacteria. Remodeling of gut microbiota could correct gut dysbiosis. The order of phages known to have these promising activities is Caudovirales, especially the families Siphoviridae and Myoviridae.


Assuntos
Infecções Bacterianas/terapia , Bacteriófagos , Terapia por Fagos/métodos , Infecções Bacterianas/fisiopatologia , Infecções Bacterianas/virologia , Bacteriófagos/isolamento & purificação , Bacteriófagos/fisiologia , Técnicas de Cultura/métodos , Técnicas de Cultura/tendências , Disbiose/fisiopatologia , Disbiose/terapia , Disbiose/virologia , Microbioma Gastrointestinal/fisiologia , Humanos , Terapia por Fagos/tendências
12.
Eur J Pharm Biopharm ; 169: 220-240, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34736984

RESUMO

The healthy human body is inhabited with a large number of bacteria, forming natural flora. It is even estimated that for a human body, its amount of DNA is less important that its bacterial genetic material. This flora plays major roles in the sickness and health of the human body and any change in its composition may lead to different diseases. Nanoparticles are widely used in numerous fields: cosmetics, food, industry, and as drug delivery carrier in the medical field. Being included in these various applications, nanoparticles may interact with the human body at various levels and with different mechanisms. These interactions differ depending on the nanoparticle nature, its structure, its concentration and manifest in different ways on the microbiota, leading to its destabilization, its restoring or showing no toxic effect. Nanoparticles may also be used as a vehicle to regulate the microbiota or to treat some of its diseases.


Assuntos
Microbiota , Nanopartículas , Disbiose/etiologia , Disbiose/fisiopatologia , Disbiose/prevenção & controle , Saúde , Humanos , Metagenoma , Microbiota/efeitos dos fármacos , Microbiota/fisiologia , Nanopartículas/efeitos adversos , Nanopartículas/metabolismo , Nanopartículas/uso terapêutico
13.
Gut Microbes ; 13(1): 1996848, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34812107

RESUMO

Amyotrophic Lateral Sclerosis is a neuromuscular disease characterized by the progressive death of motor neurons and muscle atrophy. The gastrointestinal symptoms in ALS patients were largely ignored or underestimated. The relationship between the enteric neuromuscular system and microbiome in ALS progression is unknown. We performed longitudinal studies on the enteric neuron system (ENS) and microbiome in the ALS human-SOD1G93A (Superoxide Dismutase 1) transgenic mice. We treated age-matched wild-type and ALS mice with butyrate or antibiotics to investigate the microbiome and neuromuscular functions. We examined intestinal mobility, microbiome, an ENS marker GFAP (Glial Fibrillary Acidic Protein), a smooth muscle marker (SMMHC, Smooth Muscle Myosin Heavy Chain), and human colonoids. The distribution of human-G93A-SOD1 protein was tested as an indicator of ALS progression. At 2-month-old before ALS onset, SOD1G93A mice had significantly lower intestinal mobility, decreased grip strength, and reduced time in the rotarod. We observed increased GFAP and decreased SMMHC expression. These changes correlated with consistent increased aggregation of mutated SOD1G93A in the colon, small intestine, and spinal cord. Butyrate or antibiotics treated SOD1G93A mice had a significantly longer latency to fall in the rotarod test, reduced SOD1G93A aggregation, and enhanced enteric neuromuscular function. Feces from 2-month-old SOD1G93A mice significantly enhanced SOD1G93A aggregation in human colonoids transfected with a SOD1G93A-GFP plasmid. Longitudinal studies of microbiome data further showed the altered bacterial community related to autoimmunity (e.g., Clostridium sp. ASF502, Lachnospiraceae bacterium A4), inflammation (e.g., Enterohabdus Muris,), and metabolism (e.g., Desulfovibrio fairfieldensis) at 1- and 2-month-old SOD1G93A mice, suggesting the early microbial contribution to the pathological changes. We have demonstrated a novel link between the microbiome, hSOD1G93A aggregation, and intestinal mobility. Dysbiosis occurred at the early stage of the ALS mice before observed mutated-SOD1 aggregation and dysfunction of ENS. Manipulating the microbiome improves the muscle performance of SOD1G93A mice. We provide insights into the fundamentals of intestinal neuromuscular function and microbiome in ALS.


Assuntos
Esclerose Lateral Amiotrófica/microbiologia , Disbiose/microbiologia , Sistema Nervoso Entérico/fisiopatologia , Músculo Liso/fisiopatologia , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Antibacterianos/uso terapêutico , Butiratos/uso terapêutico , Modelos Animais de Doenças , Disbiose/tratamento farmacológico , Disbiose/fisiopatologia , Sistema Nervoso Entérico/efeitos dos fármacos , Sistema Nervoso Entérico/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Humanos , Intestino Delgado/inervação , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Intestino Delgado/fisiopatologia , Estudos Longitudinais , Camundongos , Camundongos Transgênicos , Força Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/microbiologia , Agregação Patológica de Proteínas/fisiopatologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
14.
Sci Rep ; 11(1): 21397, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725349

RESUMO

Metabolic syndrome a lifestyle disease, where diet and gut microbiota play a prodigious role in its initiation and progression. Prophylactic bio-interventions employing probiotics and prebiotics offer an alternate nutritional approach towards attenuating its progression. The present study aimed to evaluate the protective efficacy of a novel synbiotic (Lactiplantibacillus pentosus GSSK2 + isomalto-oligosaccharides) in comparison to orlistat in an experimental model of metabolic syndrome. It was observed that supplementation of synbiotic for 12 weeks to Sprague Dawley rats fed with high fat diet (HFD), ameliorated the morphometric parameters i.e. weight gain, abdominal circumference, Lee's index, BMI and visceral fat deposition along with significantly increased fecal Bacteroidetes to Firmicutes ratio, elevated population of Lactobacillus spp., Akkermansia spp., Faecalibacterium spp., Roseburia spp. and decreased Enterobacteriaceae compared with HFD animals. Additionally, synbiotic administration to HFD animals exhibited improved glucose clearance, lipid biomarkers, alleviated oxidative stress, prevented leaky gut phenotype, reduced serum lipopolysaccharides and modulated the inflammatory, lipid and glucose metabolism genes along with restored histomorphology of adipose tissue, colon and liver compared with HFD animals. Taken together, the study highlights the protective potential of synbiotic in comparison with its individual components in ameliorating HFD-induced metabolic complications.


Assuntos
Disbiose/terapia , Lactobacillus/fisiologia , Síndrome Metabólica/terapia , Oligossacarídeos/uso terapêutico , Prebióticos , Probióticos/uso terapêutico , Animais , Disbiose/fisiopatologia , Microbioma Gastrointestinal , Masculino , Síndrome Metabólica/fisiopatologia , Prebióticos/administração & dosagem , Ratos Sprague-Dawley
15.
Sci Rep ; 11(1): 20571, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663882

RESUMO

Intestinal functions demonstrate circadian rhythms thought to be entrained, in part, by an organisms' intrinsic feeding and fasting periods as well as by the intestinal microbiome. Circadian disruption as a result of ill-timed nutrient exposure and obesogenic feeding poses an increased risk to disease. As such, the aim of this study was to assess the relationships between dietary timing, composition, and the microbiome with regard to rhythmic small intestinal structure and mucosal immunity. Rodent chow (RC)-mice exhibited time-dependent increases in small intestinal weight, villus height, and crypt depth as well as an increased proportion of CD8αα+ cells and concomitant decrease in CD8αß+ cells at the onset of the feeding period (p < 0.05-0.001). Western diet (WD)-animals displayed disrupted time-dependent patterns in intestinal structure and lymphocyte populations (p < 0.05-0.01). Antibiotic-induced microbial depletion abrogated the time- and diet-dependent patterns in both RC- and WD-mice (p < 0.05-0.001). However, although germ-free-mice displayed altered rhythms, fecal microbial transfer from RC-mice was generally unsuccessful in restoring structural and immune changes in these animals. This study shows that adaptive changes in the small intestine at the onset of the feeding and fasting periods are disrupted by WD-feeding, and that these changes are dependent, in part, on the intestinal microbiome.


Assuntos
Ritmo Circadiano/fisiologia , Microbioma Gastrointestinal/fisiologia , Intestino Delgado/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Disbiose/fisiopatologia , Jejum , Comportamento Alimentar/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/fisiopatologia
17.
Bioengineered ; 12(1): 6484-6498, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34517785

RESUMO

To explore mechanism of intestinal flora dysregulation promoting constipation, 60 specific pathogen-free (SPF) mice were used as research objects and were treated with constipation population fecal fluid gavage and distilled water gavage. Then, relationship between intestinal dysregulation and constipation in mice with biofilm-mediated intestinal flora was investigated in vitro. The results showed that recombinant serotonin transporter (SERT) messenger ribonucleic acid (mRNA) level of the constipation population fecal fluid gavage group and the relative expression level of SERT mRNA were 1.61 ± 0.08 and 1.49 ± 0.06, which were higher markedly than those of distilled water group (P < 0.05). The level of 5-hydroxytryptamine (5-HT) in colonic tissue of the constipation population fecal fluid gavage group was 145.36 ± 14.12 ng/mL, and the expression level of 5-HT on the surface of epithelial cells of biofilm-positive colonic tissue was 20.11 ± 2.03, which were significantly lower than those of the distilled water group, with statistical significance (P < 0.05). Besides, the microbial sequencing of fecal flora indicated that The Akk and bacteroidetes ofconstipation population fecal fluid gavage group were higher hugely than those of distilled water group (P < 0.05).In conclusion, after the occurrence of constipation, the diversity of intestinal microflora decreased, and the probiotics reduced. Iintestinal microflora dysregulation would lead to increase of SERT expression level in defecation function and intestinal motility in mice, and the decrease of 5-HT, thereby changing the intestinal movement resulting in mucosal protective barrier damage,thereby causing changes in intestinal movement and the destruction of the intestinal mucosal protective barrier, which eventually resulted in constipation. The occurrence of constipation could be improved by regulating balance of intestinal flora, increasing the diversity of flora, and reducing the genus of opportunistic pathogens.


Assuntos
Constipação Intestinal , Disbiose , Microbioma Gastrointestinal/fisiologia , Animais , Biofilmes , Constipação Intestinal/microbiologia , Constipação Intestinal/fisiopatologia , Disbiose/microbiologia , Disbiose/fisiopatologia , Fezes/microbiologia , Feminino , Mucosa Intestinal/fisiopatologia , Masculino , Camundongos , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
18.
Neurobiol Dis ; 159: 105495, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34478848

RESUMO

Long-term high-fat diet (HFD) consumption commonly leads to obesity, a major health concern of western societies and a risk factor for Alzheimer's disease (AD). Both conditions present glial activation and inflammation and show sex differences in their incidence, clinical manifestation, and disease course. HFD intake has an important impact on gut microbiota, the bacteria present in the gut, and microbiota dysbiosis is associated with inflammation and certain mental disorders such as anxiety. In this study, we have analyzed the effects of a prolonged (18 weeks, starting at 7 months of age) HFD on male and female mice, both wild type (WT) and TgAPP mice, a model for AD, investigating the behavioral profile, gut microbiota composition and inflammatory/phagocytosis-related gene expression in hippocampus. In the open-field test, no overt differences in motor activity were observed between male and female or WT and TgAPP mice on a low-fat diet (LFD). However, HFD induced anxiety, as judged by decreased motor activity and increased time in the margins in the open-field, and a trend towards increased immobility time in the tail suspension test, with increased defecation. Intriguingly, female TgAPP mice on HFD showed less immobility and defecation compared to female WT mice on HFD. HFD induced dysbiosis of gut microbiota, resulting in reduced microbiota diversity and abundance compared with LFD fed mice, with some significant differences due to sex and little effect of genotype. Gene expression of pro-inflammatory/phagocytic markers in the hippocampus were not different between male and female WT mice, and in TgAPP mice of both sexes, some cytokines (IL-6 and IFNγ) were higher than in WT mice on LFD, more so in female TgAPP (IL-6). HFD induced few alterations in mRNA expression of inflammatory/phagocytosis-related genes in male mice, whether WT (IL-1ß, MHCII), or TgAPP (IL-6). However, in female TgAPP, altered gene expression returned towards control levels following prolonged HFD (IL-6, IL-12ß, TNFα, CD36, IRAK4, PYRY6). In summary, we demonstrate that HFD induces anxiogenic symptoms, marked alterations in gut microbiota, and increased expression of inflammatory genes, except for female TgAPP that appear to be resistant to the diet effects. Lifestyle interventions should be introduced to prevent AD onset or exacerbation by reducing inflammation and its associated symptoms; however, our results suggest that the eventual goal of developing prevention and treatment strategies should take sex into consideration.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Comportamento Animal/fisiologia , Dieta Hiperlipídica , Disbiose/genética , Microbioma Gastrointestinal/fisiologia , Inflamação/genética , Estresse Psicológico/genética , Doença de Alzheimer/fisiopatologia , Animais , Modelos Animais de Doenças , Disbiose/fisiopatologia , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Interleucina-1beta/genética , Interleucina-6/genética , Masculino , Camundongos , Camundongos Transgênicos , Fagocitose/genética , RNA Mensageiro/metabolismo , Caracteres Sexuais , Estresse Psicológico/fisiopatologia
19.
Nutrients ; 13(9)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34578939

RESUMO

Patients with irritable bowel syndrome (IBS) are increasingly presenting with a wide range of neuropsychiatric symptoms, such as deterioration in gastroenteric physiology, including visceral hypersensitivity, altered intestinal membrane permeability, and gastrointestinal motor dysfunction. Functional imaging of IBS patients has revealed several abnormalities in various brain regions, such as significant activation of amygdala, thinning of insular and anterior cingulate cortex, and increase in hypothalamic gray matter, which results in poor psychiatric and cognitive outcomes. Interrelations between the enteric and central events in IBS-related gastrointestinal, neurological, and psychiatric pathologies have compelled researchers to study the gut-brain axis-a bidirectional communication that maintains the homeostasis of the gastrointestinal and central nervous system with gut microbiota as the protagonist. Thus, it can be disrupted by any alteration owing to the gut dysbiosis or loss of diversity in microbial composition. Available evidence indicates that the use of probiotics as a part of a balanced diet is effective in the management of IBS and IBS-associated neurodegenerative and psychiatric comorbidities. In this review, we delineate the pathogenesis and complications of IBS from gastrointestinal and neuropsychiatric standpoints while also discussing the neurodegenerative events in enteric and central nervous systems of IBS patients and the therapeutic potential of gut microbiota-based therapy established on clinical and preclinical data.


Assuntos
Depressão/epidemiologia , Disbiose/epidemiologia , Microbioma Gastrointestinal , Síndrome do Intestino Irritável/epidemiologia , Doenças Neurodegenerativas/epidemiologia , Encéfalo/fisiopatologia , Sistema Nervoso Central/fisiopatologia , Cognição , Comunicação , Comorbidade , Depressão/fisiopatologia , Disbiose/fisiopatologia , Feminino , Humanos , Síndrome do Intestino Irritável/fisiopatologia , Masculino , Doenças Neurodegenerativas/fisiopatologia , Probióticos/administração & dosagem
20.
Mol Brain ; 14(1): 133, 2021 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-34481527

RESUMO

Cumulative evidence shows that gut microbiome can influence brain function and behavior via the inflammatory processes. However, the role of interaction between gut dysbiosis and C-reactive protein (CRP) in the development of anxiety and depression remains to be elucidated. In this study, a total of 3321 independent single nucleotide polymorphism (SNP) loci associated with gut microbiome were driven from genome-wide association study (GWAS). Using individual level genotype data from UK Biobank, we then calculated the polygenetic risk scoring (PRS) of 114 gut microbiome related traits. Moreover, regression analysis was conducted to evaluate the possible effect of interaction between gut microbiome and CRP on the risks of Patient Health Questionnaire-9 (PHQ-9) (N = 113,693) and Generalized Anxiety Disorder-7 (GAD-7) (N = 114,219). At last, 11 candidate CRP × gut microbiome interaction with suggestive significance was detected for PHQ-9 score, such as F_Ruminococcaceae (ß = - 0.009, P = 2.2 × 10-3), G_Akkermansia (ß = - 0.008, P = 7.60 × 10-3), F_Acidaminococcaceae (ß = 0.008, P = 1.22 × 10-2), G_Holdemanella (ß = - 0.007, P = 1.39 × 10-2) and O_Lactobacillales (ß = 0.006, P = 1.79× 10-2). 16 candidate CRP × gut microbiome interaction with suggestive significance was detected for GAD-7 score, such as O_Bacteroidales (ß = 0.010, P = 4.00× 10-4), O_Selenomonadales (ß = - 0.010, P = 1.20 × 10-3), O_Clostridiales (ß = 0.009, P = 2.70 × 10-3) and G_Holdemanella (ß = - 0.008, P = 4.20 × 10-3). Our results support the significant effect of interaction between CRP and gut microbiome on the risks of anxiety and depression, and identified several candidate gut microbiomes for them.


Assuntos
Transtornos de Ansiedade/microbiologia , Eixo Encéfalo-Intestino , Proteína C-Reativa/fisiologia , Transtorno Depressivo/microbiologia , Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Genes Bacterianos , Adulto , Idoso , Transtornos de Ansiedade/etiologia , Transtornos de Ansiedade/genética , Transtornos de Ansiedade/fisiopatologia , Bactérias/genética , Bactérias/isolamento & purificação , Transtorno Depressivo/etiologia , Transtorno Depressivo/genética , Transtorno Depressivo/fisiopatologia , Disbiose/genética , Disbiose/fisiopatologia , Feminino , Fermentação , Microbioma Gastrointestinal/genética , Estudo de Associação Genômica Ampla , Humanos , Inflamação , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Risco , Índice de Gravidade de Doença , Especificidade da Espécie , Inquéritos e Questionários
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA