Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.165
Filtrar
1.
Gut Microbes ; 16(1): 2360233, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38949979

RESUMO

Functional gastrointestinal disorders (FGIDs), chronic disorders characterized by either abdominal pain, altered intestinal motility, or their combination, have a worldwide prevalence of more than 40% and impose a high socioeconomic burden with a significant decline in quality of life. Recently, FGIDs have been reclassified as disorders of gut-brain interaction (DGBI), reflecting the key role of the gut-brain bidirectional communication in these disorders and their impact on psychological comorbidities. Although, during the past decades, the field of DGBIs has advanced significantly, the molecular mechanisms underlying DGBIs pathogenesis and pathophysiology, and the role of the gut microbiome in these processes are not fully understood. This review aims to discuss the latest body of literature on the complex microbiota-gut-brain interactions and their implications in the pathogenesis of DGBIs. A better understanding of the existing communication pathways between the gut microbiome and the brain holds promise in developing effective therapeutic interventions for DGBIs.


Assuntos
Eixo Encéfalo-Intestino , Encéfalo , Gastroenteropatias , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiologia , Humanos , Eixo Encéfalo-Intestino/fisiologia , Gastroenteropatias/microbiologia , Gastroenteropatias/fisiopatologia , Encéfalo/microbiologia , Encéfalo/fisiopatologia , Animais , Trato Gastrointestinal/microbiologia
5.
Asia Pac J Clin Nutr ; 33(3): 397-404, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38965727

RESUMO

BACKGROUND AND OBJECTIVES: Hashimoto's thyroiditis (HT) is an autoimmune disease, characterized by abnormal elevation in thyroid peroxidase antibody and/or thyroglobulin antibody. In recent decades, HT disease has become more and more widespread. Patients always report multiple symptoms, even though their thyroid hormone levels are kept in normal ranges. However, no treatment exists to effectively reduce the levels of thyroid antibodies. Our study aims to determine whether calorie-restricted diet is helpful in improving health of HT patients. METHODS AND STUDY DESIGN: This is a 3-month randomized controlled trial. HT patients will be randomized into a calorie-restricted (CR) group or a calorie-unrestricted control group. All the participants will be instructed to consume a diet that includes a combination of 45-55% calories from carbohydrates, 20-30% from fats, and 15-25% from proteins, according to current Chinese Dietary Guidelines. Participants in CR group need to limit their calories intake equal to their basal energy expenditure, which means that their daily caloric intake will be limited by about 20-30%. RESULTS: The study population is planned to be 66 HT patients aged 18 to 65 years. The primary outcome is change of thyroid antibody levels from baseline. Secondary outcomes include the changes of non-hypothyroid symptoms scores, thyroid function indexes, morphology of thyroid, T lymphocyte subpopulations, inflammatory biomarkers and lipids from baseline to 12 weeks. CONCLUSIONS: This trial will have implications for nutrition treatment policy in regard to thyroid antibodies control, immune dysfunction and related non-hypothyroid symptoms improvement among HT patients.


Assuntos
Restrição Calórica , Microbioma Gastrointestinal , Doença de Hashimoto , Adolescente , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Restrição Calórica/métodos , Microbioma Gastrointestinal/fisiologia , Doença de Hashimoto/dietoterapia , Doença de Hashimoto/imunologia , Nível de Saúde , Ensaios Clínicos Controlados Aleatórios como Assunto
6.
Crit Care ; 28(1): 220, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965622

RESUMO

The gut serves as a vital immunological organ orchestrating immune responses and influencing distant mucosal sites, notably the respiratory mucosa. It is increasingly recognized as a central driver of critical illnesses, with intestinal hyperpermeability facilitating bacterial translocation, systemic inflammation, and organ damage. The "gut-lung" axis emerges as a pivotal pathway, where gut-derived injurious factors trigger acute lung injury (ALI) through the systemic circulation. Direct and indirect effects of gut microbiota significantly impact immune responses. Dysbiosis, particularly intestinal dysbiosis, termed as an imbalance of microbial species and a reduction in microbial diversity within certain bodily microbiomes, influences adaptive immune responses, including differentiating T regulatory cells (Tregs) and T helper 17 (Th17) cells, which are critical in various lung inflammatory conditions. Additionally, gut and bone marrow immune cells impact pulmonary immune activity, underscoring the complex gut-lung interplay. Moreover, lung microbiota alterations are implicated in diverse gut pathologies, affecting local and systemic immune landscapes. Notably, lung dysbiosis can reciprocally influence gut microbiota composition, indicating bidirectional gut-lung communication. In this review, we investigate the pathophysiology of ALI/acute respiratory distress syndrome (ARDS), elucidating the role of immune cells in the gut-lung axis based on recent experimental and clinical research. This exploration aims to enhance understanding of ALI/ARDS pathogenesis and to underscore the significance of gut-lung interactions in respiratory diseases.


Assuntos
Microbioma Gastrointestinal , Síndrome do Desconforto Respiratório , Humanos , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/fisiopatologia , Síndrome do Desconforto Respiratório/microbiologia , Microbioma Gastrointestinal/fisiologia , Microbioma Gastrointestinal/imunologia , Pulmão/imunologia , Pulmão/fisiopatologia , Pulmão/microbiologia , Disbiose/fisiopatologia , Disbiose/imunologia , Disbiose/complicações , Animais
7.
CNS Neurosci Ther ; 30(7): e14840, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38973202

RESUMO

BACKGROUND: Heat stress (HS) commonly occurs as a severe pathological response when the body's sensible temperature exceeds its thermoregulatory capacity, leading to the development of chronic brain inflammation, known as neuroinflammation. Emerging evidence suggests that HS leads to the disruption of the gut microbiota, whereas abnormalities in the gut microbiota have been demonstrated to affect neuroinflammation. However, the mechanisms underlying the effects of HS on neuroinflammation are poorly studied. Meanwhile, effective interventions have been unclear. ß-Hydroxybutyric acid (BHBA) has been found to have neuroprotective and anti-inflammatory properties in previous studies. This study aims to explore the modulatory effects of BHBA on neuroinflammation induced by HS and elucidate the underlying molecular mechanisms. METHODS: An in vivo and in vitro model of HS was constructed under the precondition of BHBA pretreatment. The modulatory effects of BHBA on HS-induced neuroinflammation were explored and the underlying molecular mechanisms were elucidated by flow cytometry, WB, qPCR, immunofluorescence staining, DCFH-DA fluorescent probe assay, and 16S rRNA gene sequencing of colonic contents. RESULTS: Heat stress was found to cause gut microbiota disruption in HS mouse models, and TM7 and [Previotella] spp. may be the best potential biomarkers for assessing the occurrence of HS. Fecal microbiota transplantation associated with BHBA effectively reversed the disruption of gut microbiota in HS mice. Moreover, BHBA may inhibit microglia hyperactivation, suppress neuroinflammation (TNF-α, IL-1ß, and IL-6), and reduce the expression of cortical endoplasmic reticulum stress (ERS) markers (GRP78 and CHOP) mainly through its modulatory effects on the gut microbiota (TM7, Lactobacillus spp., Ruminalococcus spp., and Prevotella spp.). In vitro experiments revealed that BHBA (1 mM) raised the expression of the ERS marker GRP78, enhanced cellular activity, and increased the generation of reactive oxygen species (ROS) and anti-inflammatory cytokines (IL-10), while also inhibiting HS-induced apoptosis, ROS production, and excessive release of inflammatory cytokines (TNF-α and IL-1ß) in mouse BV2 cells. CONCLUSION: ß-Hydroxybutyric acid may be an effective agent for preventing neuroinflammation in HS mice, possibly due to its ability to inhibit ERS and subsequent microglia neuroinflammation via the gut-brain axis. These findings lay the groundwork for future research and development of BHBA as a preventive drug for HS and provide fresh insights into techniques for treating neurological illnesses by modifying the gut microbiota.


Assuntos
Ácido 3-Hidroxibutírico , Eixo Encéfalo-Intestino , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Doenças Neuroinflamatórias , Animais , Camundongos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/fisiologia , Eixo Encéfalo-Intestino/fisiologia , Eixo Encéfalo-Intestino/efeitos dos fármacos , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Masculino , Ácido 3-Hidroxibutírico/farmacologia , Transtornos de Estresse por Calor/metabolismo , Chaperona BiP do Retículo Endoplasmático , Fármacos Neuroprotetores/farmacologia , Resposta ao Choque Térmico/fisiologia , Resposta ao Choque Térmico/efeitos dos fármacos
8.
Cell Host Microbe ; 32(7): 1048-1049, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38991502

RESUMO

Gestational diabetes mellitus (GDM) is associated with increased risk of metabolic and neurodevelopmental disorders in offspring. In this issue of Cell Host & Microbe, Wang et al. provide evidence that changes in the gut microbiome of mothers with GDM may lead to dysbiosis in their infants and altered development in a sex-dependent manner.


Assuntos
Diabetes Gestacional , Disbiose , Microbioma Gastrointestinal , Diabetes Gestacional/microbiologia , Diabetes Gestacional/metabolismo , Gravidez , Microbioma Gastrointestinal/fisiologia , Humanos , Feminino , Disbiose/microbiologia , Lactente , Masculino , Recém-Nascido
9.
Cell Host Microbe ; 32(7): 1041-1043, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38991499

RESUMO

Multiple host and microbial factors dictate whether Candida albicans can colonize the mammalian gastrointestinal tract. In this issue of Cell Host & Microbe, Savage et al. demonstrate that restoration of intestinal epithelial hypoxia is sufficient to restore Candida albicans colonization resistance, even when other Candida inhibitory effectors remain depleted.


Assuntos
Candida albicans , Candidíase , Trato Gastrointestinal , Candida albicans/crescimento & desenvolvimento , Candida albicans/fisiologia , Humanos , Trato Gastrointestinal/microbiologia , Candidíase/microbiologia , Animais , Hipóxia/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Camundongos , Interações Hospedeiro-Patógeno , Microbioma Gastrointestinal/fisiologia
10.
PeerJ ; 12: e17597, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38974417

RESUMO

The huhu beetle (Prionoplus reticularis) is the largest endemic beetle found throughout Aotearoa New Zealand, and is characterised by feeding on wood during its larval stage. It has been hypothesised that its gut microbiome plays a fundamental role in the degradation of wood. To explore this idea we examined the fungal and bacterial community composition of huhu grubs' frass, using amplicon sequencing. Grubs were reared on an exclusive diet of either a predominantly cellulose source (cotton) or lignocellulose source (pine) for 4 months; subsequently a diet switch was performed and the grubs were grown for another 4 months. The fungal community of cellulose-reared huhu grubs was abundant in potential cellulose degraders, contrasting with the community of lignocellulose-reared grubs, which showed abundant potential soft rot fungi, yeasts, and hemicellulose and cellulose degraders. Cellulose-reared grubs showed a less diverse fungal community, however, diet switch from cellulose to lignocellulose resulted in a change in community composition that showed grubs were still capable of utilising this substrate. Conversely, diet seemed to have a limited influence on huhu grub gut bacterial communities.


Assuntos
Besouros , Microbioma Gastrointestinal , Lignina , Microbioma Gastrointestinal/fisiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Animais , Lignina/metabolismo , Besouros/microbiologia , Celulose/metabolismo , Dieta , Nova Zelândia , Fungos/genética , Fungos/metabolismo , Bactérias/genética , Bactérias/classificação , Bactérias/metabolismo
11.
CNS Neurosci Ther ; 30(7): e14823, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38992870

RESUMO

BACKGROUND: Caloric restriction (CR) might be effective for alleviating/preventing Alzheimer's disease (AD), but the biological mechanisms remain unclear. In the current study, we explored whether CR caused an alteration of gut microbiome and resulted in the attenuation of cognitive impairment of AD animal model. METHODS: Thirty-week-old male APP/PS1 transgenic mice were used as AD models (AD mouse). CR was achieved by 30% reduction of daily free feeding (ad libitum, AL) amount. The mice were fed with CR protocol or AL protocol for six consecutive weeks. RESULTS: We found that with CR treatment, AD mice showed improved ability of learning and spatial memory, and lower levels of Aß40, Aß42, IL-1ß, TNF-α, and ROS in the brain. By sequencing 16S rDNA, we found that CR treatment resulted in significant diversity in composition and abundance of gut flora. At the phylum level, Deferribacteres (0.04%), Patescibacteria (0.14%), Tenericutes (0.03%), and Verrucomicrobia (0.5%) were significantly decreased in CR-treated AD mice; at the genus level, Dubosiella (10.04%), Faecalibaculum (0.04%), and Coriobacteriaceae UCG-002 (0.01%) were significantly increased in CR-treated AD mice by comparing with AL diet. CONCLUSIONS: Our results demonstrate that the attenuation of AD following CR treatment in APP/PS1 mice may result from alterations in the gut microbiome. Thus, gut flora could be a new target for AD prevention and therapy.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Restrição Calórica , Microbioma Gastrointestinal , Camundongos Transgênicos , Animais , Microbioma Gastrointestinal/fisiologia , Restrição Calórica/métodos , Doença de Alzheimer/microbiologia , Doença de Alzheimer/dietoterapia , Doença de Alzheimer/prevenção & controle , Masculino , Camundongos , Precursor de Proteína beta-Amiloide/genética , Presenilina-1/genética , Peptídeos beta-Amiloides/metabolismo , Modelos Animais de Doenças , Aprendizagem em Labirinto/fisiologia , Encéfalo/metabolismo , Camundongos Endogâmicos C57BL
12.
Int J Mol Sci ; 25(13)2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-39000530

RESUMO

The fish gut microbiome is well known for its role in degrading nutrients to improve the host's digestion and absorption efficiency. In this study, we focused on the core physiological adaptability during the various reproductive stages of the black Amur bream (Megalobrama terminalis) to explore the interaction mechanisms among the fish host gut mucosal structure, gut enzyme activity, and gut microbial metabolism in the course of the host's reproductive cycle. Our findings showed that M. terminalis exhibited locomotion metabolic type (aids in sporting) in the reproductive stage, and a change to visceral metabolic type (aids in digestion) during non-reproductive and post-reproductive stage phases. The impact of metabolic type selection and energy demand during various reproductive stages on fish nutrition strategy and digestive function was substantial. Our resulted showed that mitochondria in intestinal epithelial cells of reproductive M. terminalis appeared autophagy phenomenon, and the digestive enzyme activities in the intestines of reproductive M. terminalis were lower than those in the non-reproductive and post-reproductive individuals. Moreover, these differences in nutrition strategy have a prominent impact on the gut microbiome of reproductive M. terminalis, compared to non-reproductive and post-reproductive samples. Our findings showed that reproductive females had lower levels of alpha diversity compared to non-reproductive and post-reproductive females. Our results also showed a greater functional variety and an increase in functional genes related to carbohydrate, lipid, amino acid, cofactors, and vitamin metabolic pathways in the NRS and PRS group. It is noteworthy that an enrichment of genes encoding putative enzymes implicated in the metabolism of taurine and hypotaurine was observed in the RS samples. Our findings illustrated that the stability and resilience of the gut bacterial community could be shaped in the wild fish host-microbiome interactions during reproductive life history.


Assuntos
Microbioma Gastrointestinal , Reprodução , Animais , Microbioma Gastrointestinal/fisiologia , Feminino , Cyprinidae/microbiologia , Cyprinidae/fisiologia , Cyprinidae/metabolismo , Água Doce/microbiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia
13.
Int J Mol Sci ; 25(13)2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-39000578

RESUMO

Aging clocks are predictive models of biological age derived from age-related changes, such as epigenetic changes, blood biomarkers, and, more recently, the microbiome. Gut and skin microbiota regulate more than barrier and immune function. Recent studies have shown that human microbiomes may predict aging. In this narrative review, we aim to discuss how the gut and skin microbiomes influence aging clocks as well as clarify the distinction between chronological and biological age. A literature search was performed on PubMed/MEDLINE databases with the following keywords: "skin microbiome" OR "gut microbiome" AND "aging clock" OR "epigenetic". Gut and skin microbiomes may be utilized to create aging clocks based on taxonomy, biodiversity, and functionality. The top contributing microbiota or metabolic pathways in these aging clocks may influence aging clock predictions and biological age. Furthermore, gut and skin microbiota may directly and indirectly influence aging clocks through the regulation of clock genes and the production of metabolites that serve as substrates or enzymatic regulators. Microbiome-based aging clock models may have therapeutic potential. However, more research is needed to advance our understanding of the role of microbiota in aging clocks.


Assuntos
Envelhecimento , Microbioma Gastrointestinal , Microbiota , Pele , Humanos , Pele/microbiologia , Pele/metabolismo , Microbioma Gastrointestinal/fisiologia , Epigênese Genética , Animais , Relógios Biológicos
14.
Zool Res ; 45(4): 857-874, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39004863

RESUMO

Emerging evidence indicates that sleep deprivation (SD) can lead to Alzheimer's disease (AD)-related pathological changes and cognitive decline. However, the underlying mechanisms remain obscure. In the present study, we identified the existence of a microbiota-gut-brain axis in cognitive deficits resulting from chronic SD and revealed a potential pathway by which gut microbiota affects cognitive functioning in chronic SD. Our findings demonstrated that chronic SD in mice not only led to cognitive decline but also induced gut microbiota dysbiosis, elevated NLRP3 inflammasome expression, GSK-3ß activation, autophagy dysfunction, and tau hyperphosphorylation in the hippocampus. Colonization with the "SD microbiota" replicated the pathological and behavioral abnormalities observed in chronic sleep-deprived mice. Remarkably, both the deletion of NLRP3 in NLRP3 -/- mice and specific knockdown of NLRP3 in the hippocampus restored autophagic flux, suppressed tau hyperphosphorylation, and ameliorated cognitive deficits induced by chronic SD, while GSK-3ß activity was not regulated by the NLRP3 inflammasome in chronic SD. Notably, deletion of NLRP3 reversed NLRP3 inflammasome activation, autophagy deficits, and tau hyperphosphorylation induced by GSK-3ß activation in primary hippocampal neurons, suggesting that GSK-3ß, as a regulator of NLRP3-mediated autophagy dysfunction, plays a significant role in promoting tau hyperphosphorylation. Thus, gut microbiota dysbiosis was identified as a contributor to chronic SD-induced tau pathology via NLRP3-mediated autophagy dysfunction, ultimately leading to cognitive deficits. Overall, these findings highlight GSK-3ß as a regulator of NLRP3-mediated autophagy dysfunction, playing a critical role in promoting tau hyperphosphorylation.


Assuntos
Autofagia , Disbiose , Microbioma Gastrointestinal , Proteína 3 que Contém Domínio de Pirina da Família NLR , Privação do Sono , Proteínas tau , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Microbioma Gastrointestinal/fisiologia , Privação do Sono/metabolismo , Privação do Sono/fisiopatologia , Privação do Sono/complicações , Camundongos , Autofagia/fisiologia , Proteínas tau/metabolismo , Proteínas tau/genética , Masculino , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inflamassomos/metabolismo
15.
Medicine (Baltimore) ; 103(28): e38689, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38996098

RESUMO

Acute pancreatitis (AP), a severe inflammatory condition affecting the pancreas requires investigation into its predictors. Melatonin, a compound with anti-inflammatory and antioxidant properties, has shown promise in managing AP. Additionally, the gut microbiota, a community of microorganisms residing in the intestines has been linked to AP development. This study aims to explore the correlation between melatonin and gut microbiota in predicting AP severity. This study involved 199 participants, with 99 diagnosed with AP and 100 serving as healthy controls. The AP patients were categorized into 2 groups based on the severity of their condition: mild AP (MAP) and severe AP (SAP). Serum melatonin levels were measured on Days 1, 3, and 5 of hospitalization, and gut microbiota composition was examined via 16S rRNA gene sequencing. Other parameters were evaluated, such as the Acute Physiology and Chronic Health Evaluation (APACHE) score, Ranson, and Acute Gastrointestinal Injury (AGI) scores. Melatonin levels were significantly lower in subjects with severe AP compared those with mild AP (18.2 ng/mL vs 32.2 ng/mL, P = .001), and melatonin levels decreased significantly in patients with AP on Days 3 and 5. The study also revealed that individuals with AP exhibited a significantly altered gut microbiota composition compared to control individuals, with a lower Shannon index and higher Simpson index. The AUCs for Simpson index and F/B ratio were significantly higher than those for other biomarkers, indicating that these gut microbiota markers may also be useful for AP prediction. The study proposes that there is a relationship between melatonin levels and the dynamics of gut microbiota profiles in relation to the severity of AP. As a result, the severity of the disease can be assessed by assessing the levels of serum melatonin and gut microbiota profiles.


Assuntos
Microbioma Gastrointestinal , Melatonina , Pancreatite , Índice de Gravidade de Doença , Humanos , Melatonina/sangue , Microbioma Gastrointestinal/fisiologia , Masculino , Feminino , Estudos de Casos e Controles , Pancreatite/microbiologia , Pancreatite/sangue , Pessoa de Meia-Idade , Adulto , Idoso , Transdução de Sinais , Doença Aguda , RNA Ribossômico 16S
16.
BMC Med ; 22(1): 300, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39020393

RESUMO

BACKGROUND: Multiple high doses of 131I therapy in patients with differentiated thyroid cancer (DTC) might disrupt the balance of gut microbiota and metabolites. This study aimed to investigate the alterations of intestinal bacteria and metabolism over two courses of 131I therapy, explore the interactions, and construct diagnostic models reflecting enteric microecology based on 131I therapy. METHODS: A total of 81 patients were recruited for the first 131I therapy (131I-1st), among whom 16 received a second course (131I-2nd) after half a year. Fecal samples were collected 1 day before (Pre-131I-1st/2nd) and 3 days after (Post-131I-1st/2nd) 131I therapy for microbiome (16S rRNA gene sequencing) and metabolomic (LC-MS/MS) analyses. RESULTS: A total of six microbial genera and 11 fecal metabolites enriched in three pathways were identified to show significant differences between Pre-131I-1st and other groups throughout the two courses of 131I treatment. In the Post-131I-1st group, the beneficial bacteria Bifidobacterium, Lachnoclostridium, uncultured_bacterium_f_Lachnospiraceae, and Lachnospiraceae_UCG004 were abundant and the radiation-sensitive pathways of linoleic acid (LA), arachidonic acid, and tryptophan metabolism were inhibited compared with the Pre-131I-1st group. Compared with the Pre-131I-1st group, the Pre-131I-2nd group exhibited a reduced diversity of flora and differentially expressed metabolites, with a low abundance of beneficial bacteria and dysregulated radiation-sensitive pathways. However, less significant differences in microbiota and metabolites were found between the Pre/Post-131I-2nd groups compared with those between the Pre/Post-131I-1st groups. A complex co-occurrence was observed between 6 genera and 11 metabolites, with Lachnoclostridium, Lachnospiraceae_UCG004, Escherichia-Shigella, and LA-related metabolites contributing the most. Furthermore, combined diagnostic models of charactered bacteria and metabolites answered well in the early, long-term, and dose-dependent responses for 131I therapy. CONCLUSIONS: Different stages of 131I therapy exert various effects on gut microecology, which play an essential role in regulating radiotoxicity and predicting the therapeutic response.


Assuntos
Fezes , Microbioma Gastrointestinal , Radioisótopos do Iodo , Neoplasias da Glândula Tireoide , Humanos , Microbioma Gastrointestinal/fisiologia , Neoplasias da Glândula Tireoide/radioterapia , Neoplasias da Glândula Tireoide/microbiologia , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Fezes/microbiologia , Idoso , RNA Ribossômico 16S/genética , Adulto Jovem
17.
Alcohol Alcohol ; 59(5)2024 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-39042929

RESUMO

The human intestine is colonized by a variety of microorganisms that influence the immune system, the metabolic response, and the nervous system, with consequences for brain function and behavior. Unbalance in this microbial ecosystem has been shown to be associated with psychiatric disorders, and altered gut microbiome composition related to bacteria, viruses, and fungi has been well established in patients with alcohol use disorder. This review describes the gut microbiome-brain communication pathways, including the ones related to the vagus nerve, the inflammatory cytokines, and the gut-derived metabolites. Finally, the potential benefits of microbiota-based therapies for the management of alcohol use disorder, such as probiotics, prebiotics, and fecal microbiota transplantation, are also discussed.


Assuntos
Alcoolismo , Eixo Encéfalo-Intestino , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Probióticos , Humanos , Microbioma Gastrointestinal/fisiologia , Eixo Encéfalo-Intestino/fisiologia , Alcoolismo/microbiologia , Alcoolismo/terapia , Probióticos/uso terapêutico , Prebióticos , Animais , Encéfalo/metabolismo
18.
Front Cell Infect Microbiol ; 14: 1371312, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39035357

RESUMO

The symbiotic relationship between the human digestive system and its intricate microbiota is a captivating field of study that continues to unfold. Comprising predominantly anaerobic bacteria, this complex microbial ecosystem, teeming with trillions of organisms, plays a crucial role in various physiological processes. Beyond its primary function in breaking down indigestible dietary components, this microbial community significantly influences immune system modulation, central nervous system function, and disease prevention. Despite the strides made in microbiome research, the precise mechanisms underlying how bacterial effector functions impact mammalian and microbiome physiology remain elusive. Unlike the traditional DNA-RNA-protein paradigm, bacteria often communicate through small molecules, underscoring the imperative to identify compounds produced by human-associated bacteria. The gut microbiome emerges as a linchpin in the transformation of natural products, generating metabolites with distinct physiological functions. Unraveling these microbial transformations holds the key to understanding the pharmacological activities and metabolic mechanisms of natural products. Notably, the potential to leverage gut microorganisms for large-scale synthesis of bioactive compounds remains an underexplored frontier with promising implications. This review serves as a synthesis of current knowledge, shedding light on the dynamic interplay between natural products, bacteria, and human health. In doing so, it contributes to our evolving comprehension of microbiome dynamics, opening avenues for innovative applications in medicine and therapeutics. As we delve deeper into this intricate web of interactions, the prospect of harnessing the power of the gut microbiome for transformative medical interventions becomes increasingly tantalizing.


Assuntos
Produtos Biológicos , Microbioma Gastrointestinal , Humanos , Produtos Biológicos/farmacologia , Produtos Biológicos/metabolismo , Microbioma Gastrointestinal/fisiologia , Bactérias/metabolismo , Bactérias/classificação , Animais , Interações entre Hospedeiro e Microrganismos , Simbiose
19.
J Vis Exp ; (208)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38975757

RESUMO

Gut microbial products are known to act both locally within the intestine and get absorbed into circulation, where their effects can extend to numerous distant organ systems. Short-chain fatty acids (SCFA) are one class of metabolites produced by gut microbes during the fermentation of indigestible dietary fiber. They are now recognized as important contributors to how the gut microbiome influences extra-intestinal organ systems via the gut-lung, gut-brain, and other gut-organ axes throughout the host. SCFAs are absorbed from the colon, through intestinal tissue, into the portal vein (PV). They then pass through the liver, and are consumed in various organs such as the brain, muscle, adipose tissue, and lungs. SCFAs are most easily measured in the expelled fecal material however, more accurate measurements have been obtained from intra-colonic fecal contents. Here we propose that sampling PV and systemic circulating plasma of a single subject may be preferable for studying the absorption, transport, and systemic levels of SCFAs in mice. We present a new technique for efficient blood sampling from the PV and inferior vena cava (IVC) that allows for the collection of relatively large volumes of blood from the portal and systemic circulations. This is accomplished by ligating the PV, thereby allowing for the dilation or enlargement of the PV as it backfills from the mesenteric veins that drain into it. Using this method, we were able to improve the rate of successful collection as well as the total amount of blood collected (up to 0.3 mL from IVC and 0.5 mL from PV).


Assuntos
Microbioma Gastrointestinal , Veia Porta , Veia Cava Inferior , Animais , Camundongos , Veia Porta/metabolismo , Microbioma Gastrointestinal/fisiologia , Veia Cava Inferior/metabolismo , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/análise , Coleta de Amostras Sanguíneas/métodos , Masculino
20.
Crit Rev Food Sci Nutr ; 64(20): 7067-7084, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38975869

RESUMO

Multiple beneficial effects have been attributed to green tea catechins (GTCs). However, the bioavailability of GTCs is generally low, with only a small portion directly absorbed in the small intestine. The majority of ingested GTCs reaches the large intestinal lumen, and are extensively degraded via biotransformation by gut microbiota, forming many low-molecular-weight metabolites such as phenyl-γ-valerolactones, phenolic acids, butyrate, and acetate. This process not only improves the overall bioavailability of GTC-derived metabolites but also enriches the biological activities of GTCs. Therefore, the intra- and inter-individual differences in human gut microbiota as well as the resulting biological contribution of microbial metabolites are crucial for the ultimate health benefits. In this review, the microbial degradation of major GTCs was characterized and an overview of the in vitro models used for GTC metabolism was summarized. The intra- and inter-individual differences of human gut microbiota composition and the resulting divergence in the metabolic patterns of GTCs were highlighted. Moreover, the potential beneficial effects of GTCs and their gut microbial metabolites were also discussed. Overall, the microbial metabolites of GTCs with higher bioavailability and bioactive potency are key factors for the observed beneficial effects of GTCs and green tea consumption.


Assuntos
Disponibilidade Biológica , Catequina , Microbioma Gastrointestinal , Chá , Microbioma Gastrointestinal/fisiologia , Humanos , Chá/química , Catequina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA