Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 446
Filtrar
1.
Cureus ; 16(8): e67010, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39280497

RESUMEN

Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide. Recent research has identified gut dysbiosis - an imbalance in the gut microbiota - as a significant factor in the development of CVDs. This complex relationship between gut microbiota and cardiovascular health involves various mechanisms, including the production of metabolites such as trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFAs). These metabolites influence lipid metabolism, inflammation, and blood pressure regulation. In addition, the gut-brain axis and neurohormonal pathways play crucial roles in cardiovascular function. Epidemiological studies have linked gut dysbiosis to various cardiovascular conditions, highlighting the potential for therapeutic interventions. Dietary changes, probiotics, and prebiotics have shown promise in modulating gut microbiota and reducing cardiovascular risk factors. This underscores the critical role of gut health in preventing and treating CVDs. However, further research is needed to develop targeted therapies that can enhance cardiovascular outcomes.

2.
Front Microbiol ; 15: 1407991, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39234554

RESUMEN

Cirrhosis, a pathological stage that develops from various chronic liver diseases, is characterized by liver fibrosis, pseudolobular formation, and chronic inflammation. When it progresses to the decompensated phase, the mortality rate of cirrhosis can reach 80%. The role of gut microbiota in the progression of liver diseases has received significant attention. Numerous studies have shown that regulating gut microbiota has significant therapeutic effects on preventing and reversing liver cirrhosis. This article reviewed the mechanisms by which gut microbiota influence liver cirrhosis, explaining the effective therapeutic effects of traditional Chinese medicine. Through multi-directional regulation involving signaling pathways, gut microbiota diversity, and restoration of intestinal barrier function, traditional Chinese medicine has been promising in ameliorating liver cirrhosis, providing treatment options and pharmacological guidance for the occurrence and development of liver cirrhosis.

3.
Mucosal Immunol ; 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39265892

RESUMEN

The host-microbiome axis has been implicated in promoting anti-inflammatory immune responses. Yet, the underlying molecular mechanisms of commensal-mediated IL-10 production by regulatory B cells (Bregs) are not fully elucidated. Here, we demonstrate that bacterial CpG motifs trigger the signaling downstream of TLR9 promoting IκBNS-mediated expression of Blimp-1, a transcription regulator of IL-10. Surprisingly, this effect was counteracted by the NF-κB transcription factor c-Rel. A functional screen for intestinal bacterial species identified the commensal Clostridium sporogenes, secreting high amounts of short-chain fatty acids (SCFAs) and branched-chain fatty acids (BCFAs), as an amplifier of IL-10 production by promoting sustained mTOR signaling in B cells. Consequently, enhanced Breg functionality was achieved by combining CpG with the SCFA butyrate or the BCFA isovalerate thereby synergizing TLR- and mTOR-mediated pathways. Collectively, Bregs required two bacterial signals (butyrate and CpG) to elicit their full suppressive capacity and ameliorate T cell-mediated intestinal inflammation. Our study has dissected the molecular pathways induced by bacterial factors, which might contribute not only to better understanding of host-microbiome interactions, but also to exploration of new strategies for improvement of anti-inflammatory cellular therapy.

4.
J Clin Med ; 13(17)2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39274267

RESUMEN

Background: Early diagnosis of post-operative complications is an urgent task, allowing timely prescribing of appropriate therapy and reducing the cost of patient treatment. The purpose of this study was to determine whether an integrated approach based on clinical data, along with metabolites and biomarkers, had greater predictive value than the models built on fewer data in the early diagnosis of post-operative complications after cardiac surgery. Methods: The study included patients (n = 62) admitted for planned cardiac surgery (coronary artery bypass grafting with cardiopulmonary bypass) with (n = 26) or without (n = 36) post-operative complications. Clinical and laboratory data on the first day after surgery were analyzed. Additionally, patients' blood samples were collected before and on the first day after surgery to determine biomarkers and metabolites. Results: Multivariate PLS-DA models, predicting the presence or absence of post-operative complications, were built using clinical data, concentrations of metabolites and biomarkers, and the entire data set (ROC-AUC = 0.80, 0.71, and 0.85, respectively). For comparison, we built univariate models using the EuroScore2 and SOFA scales, concentrations of lactate, the dynamic changes of 4-hydroxyphenyllactic acid, and the sum of three sepsis-associated metabolites (ROC-AUC = 0.54, 0.79, 0.62, 0.58, and 0.70, respectively). Conclusions: The proposed complex model using the entire dataset had the best characteristics, which confirms the expediency of searching for new predictive models based on a variety of factors.

5.
Trends Mol Med ; 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256110

RESUMEN

Increasing evidence suggests that the gut microbiome plays a key role in a host of pathological conditions, including cancer. Indeed, the bidirectional communication that occurs between the gut and the brain, known as the 'gut-brain axis,' has recently been implicated in brain tumour pathology. Here, we focus on current research that supports a gut microbiome-brain tumour link with emphasis on high-grade gliomas, the most aggressive of all brain tumours, and the impact on the glioma tumour microenvironment. We discuss the potential use of gut-brain axis signals to improve responses to current and future therapeutic approaches. We highlight that the success of novel treatment strategies may rely on patient-specific microbiome profiles, and these should be considered for personalised treatment approaches.

6.
Cell Host Microbe ; 32(9): 1488-1501.e5, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39214086

RESUMEN

The pathogenic outcome of enteric virus infections is governed by a complex interplay between the virus, intestinal microbiota, and host immune factors, with metabolites serving as a key mediator. Noroviruses bind bile acid metabolites, which are produced by the host and then modified by commensal bacteria. Paradoxically, bile acids can have both proviral and antiviral roles during norovirus infections. Working in an infant mouse model of norovirus infection, we demonstrate that microbiota and their bile acid metabolites protect from norovirus diarrhea, whereas host bile acids promote disease. We also find that maternal bile acid metabolism determines the susceptibility of newborn mice to norovirus diarrhea during breastfeeding. Finally, targeting maternal and neonatal bile acid metabolism can protect newborn mice from norovirus disease. In summary, neonatal metabolic immaturity and breastmilk bile acids are central determinants of heightened newborn vulnerability to norovirus disease.


Asunto(s)
Animales Recién Nacidos , Ácidos y Sales Biliares , Infecciones por Caliciviridae , Diarrea , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Leche Humana , Norovirus , Animales , Ratones , Ácidos y Sales Biliares/metabolismo , Infecciones por Caliciviridae/metabolismo , Infecciones por Caliciviridae/virología , Leche Humana/virología , Leche Humana/metabolismo , Diarrea/virología , Diarrea/metabolismo , Femenino , Humanos , Ratones Endogámicos C57BL
7.
Microbiol Res ; 288: 127871, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39137590

RESUMEN

The gut microbiota plays a critical role in numerous biochemical processes essential for human health, such as metabolic regulation and immune system modulation. An increasing number of research suggests a strong association between the gut microbiota and carcinogenesis. The diverse metabolites produced by gut microbiota can modulate cellular gene expression, cell cycle dynamics, apoptosis, and immune system functions, thereby exerting a profound influence on cancer development and progression. A healthy gut microbiota promotes substance metabolism, stimulates immune responses, and thereby maintains the long-term homeostasis of the intestinal microenvironment. When the gut microbiota becomes imbalanced and disrupts the homeostasis of the intestinal microenvironment, the risk of various diseases increases. This review aims to elucidate the impact of gut microbial metabolites on cancer initiation and progression, focusing on short-chain fatty acids (SCFAs), polyamines (PAs), hydrogen sulfide (H2S), secondary bile acids (SBAs), and microbial tryptophan catabolites (MTCs). By detailing the roles and molecular mechanisms of these metabolites in cancer pathogenesis and therapy, this article sheds light on dual effects on the host at different concentrations of metabolites and offers new insights into cancer research.


Asunto(s)
Ácidos y Sales Biliares , Progresión de la Enfermedad , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Neoplasias , Humanos , Neoplasias/microbiología , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Ácidos Grasos Volátiles/metabolismo , Ácidos y Sales Biliares/metabolismo , Sulfuro de Hidrógeno/metabolismo , Poliaminas/metabolismo , Triptófano/metabolismo , Carcinogénesis , Animales , Microambiente Tumoral
8.
J Environ Manage ; 368: 122184, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39128358

RESUMEN

Ion adsorption rare earth ore nearly satisfy global market demand for heavy rare earth elements (HREEs). Bio-leaching has important potential for the clean and efficient extraction of ion-adsorption rare earth ore. However, the complexities of in-situ mining restrict the use of contact/direct bio-leaching, and non-contact/indirect bio-leaching would be the best choice. This study explore the potential of fermentation broths prepared by Yarrowia lipolytica (ATCC 30162) for the bio-leaching of ion-adsorption rare earth ore, and three typical metabolites (potassium citrate (K3Cit), sodium citrate (Na3Cit) and ammonium citrate ((NH4)3Cit) of Yarrowia lipolytica were further evaluated in simulated bioleaching (non-contact bioleaching) of ion-adsorption rare earth ore, including leaching behavior, seepage rule and rare earth elements (REEs) morphological transformation. The column leaching experiments shown that direct leaching of REEs using fermentation broths results in incomplete leaching of REEs due to the influence of impurities. Using the purified and prepared metabolites as lixiviant, REEs can be effectively extracted (leaching efficiency >90%) at cation concentration was only 10 % of the commonly used ammonium sulfate concentration (45 mM). Cation type had less effect on leaching efficiency. During the ion-adsorption rare earth ore leaching process, rare earth ions form a variety of complex chelates with citrate, thus transferring rare earth elements from the mineral surface to the leachate. Experimental results showed that pH and concentration together determined the type and form of rare earth chelates, which in turn affect the leaching behavior of REEs and solution seepage rule. This study helps to provide a theoretical basis for the regulation and enhancement of ion-adsorption rare earth ore non-contact bioleaching process.


Asunto(s)
Metales de Tierras Raras , Metales de Tierras Raras/metabolismo , Adsorción , Minería , Yarrowia/metabolismo , Fermentación , Iones
9.
Arch Anim Nutr ; : 1-17, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39087698

RESUMEN

This study investigated the effects of dietary L-valine (Val) supplementation and sanitary conditions with lipopolysaccharide injection on growth performance, immune response, and intestinal bacterial profiles and metabolites in weaned pigs. Thirty-two weaned pigs (6.98 ± 0.47 kg) were randomly assigned to treatments in a 2 × 2 factorial arrangement based on dietary Val levels and sanitary conditions (low or high). The pigs were fed either a basal diet containing the standard levels of Val suggested by (NRC), (2012) or a basal diet supplemented with 0.1% L-Val. A room designated as a high sanitary room was washed weekly, whereas the designated low sanitary room was not washed throughout the experiment and 5 kg of manure from the nursery pig barn was spread on the pen floors on day 1. All data were analysed using a mixed procedure of SAS, with the individual pen as the experimental unit. The pigs raised in low sanitary conditions exhibited a lower (p < 0.05) average daily gain, average daily feed intake, and gain-to-feed ratio and a higher (p < 0.05) incidence of diarrhoea than those raised in high sanitary conditions during the 14-d experimental period. The pigs in the low sanitary group also had a lower (p < 0.05) concentration of butyrate in the jejunum and a higher (p < 0.05) concentration of NH3-N in the colon than those in the high sanitary group. Dietary Val supplementation was reduced (p < 0.05) plasma interleukin (IL)-1ß and IL-1 receptor antagonist concentrations as well as isovalerate and NH3-N concentrations in the colon, regardless of sanitary conditions. Interactions between dietary Val supplementation and sanitary conditions were observed in the abundances of mRNA-encoding ß-defensins 113, 125 and 129 (p < 0.05). In conclusion, dietary Val supplementation beneficially modulates inflammatory responses and microbial metabolites regardless of sanitary conditions while transcriptional levels of ß-defensins are regulated by dietary Val supplementation in a manner dependent on housing hygiene conditions.

10.
Cell Host Microbe ; 32(8): 1280-1300, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39146799

RESUMEN

The inhabitants of our intestines, collectively called the gut microbiome, comprise fungi, viruses, and bacterial strains. These microorganisms are involved in the fermentation of dietary compounds and the regulation of our adaptive and innate immune systems. Less known is the reciprocal interaction between the gut microbiota and type 2 diabetes mellitus (T2DM), as well as their role in modifying therapies to reduce associated morbidity and mortality. In this review, we aim to discuss the existing literature on gut microbial strains and their diet-derived metabolites involved in T2DM. We also explore the potential diagnostics and therapeutic avenues the gut microbiota presents for targeted T2DM management. Personalized treatment plans, driven by diet and medication based on the patient's microbiome and clinical markers, could optimize therapy.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Diabetes Mellitus Tipo 2/microbiología , Humanos , Dieta , Bacterias/metabolismo , Bacterias/clasificación , Animales , Disbiosis/microbiología
11.
Microorganisms ; 12(8)2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39203515

RESUMEN

Dietary fiber (DF) is an important nutrient component in pig's diet that remarkably influences their growth and slaughter performance. The ability of pigs to digest DF depends on the microbial composition of the intestinal tract, particularly in the hindgut. However, studies on how DF alters the growth and slaughter performance of pigs by shaping the gut microbial composition and metabolites are still limited. Therefore, this study aimed to investigate the effects of DF on microbial composition, functions, and metabolites, ultimately altering host growth and slaughter performance using Durco × Bamei crossbred pigs supplemented with 0%, 10%, 17%, and 24% broad bean silage in the basic diet. We found that the final weight, average daily gain, fat, and lean meat weight significantly decreased with increasing DF. Pigs with the lowest slaughter rate and fat weight were observed in the 24% fiber-supplemented group. Gut microbial communities with the highest alpha diversity were formed in the 17% fiber group. The relative abundance of fiber-degrading bacteria, bile acid, and succinate-producing bacteria, including Prevotella sp., Bacteroides sp., Ruminococcus sp., and Parabacteroides sp., and functional pathways, including the butanoate metabolism and the tricarboxylic acid [TCA] cycle, significantly increased in the high-fiber groups. The concentrations of several bile acids significantly decreased in the fiber-supplemented groups, whereas the concentrations of succinate and long-chain fatty acids increased. Our results indicate that a high-fiber diet may alter the growth and slaughter performance of Durco × Bamei crossbred pigs by modulating the composition of Prevotella sp., Bacteroides sp., Ruminococcus sp., Parabacteroides sp., and metabolite pathways of bile acids and succinate.

12.
Front Med (Lausanne) ; 11: 1412709, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39170038

RESUMEN

Background: Preclinical research has identified the mechanisms via which bacteria influence cancer treatment outcomes. Clinical studies have demonstrated the potential to modify the microbiome in cancer treatment. Herein, we systematically analyze how gut microorganisms interact with chemotherapy and immune checkpoint inhibitors, specifically focusing on how gut bacteria affect the pharmacokinetics and pharmacodynamics of cancer treatment. Method: This study searched Web of Science, Scopus, and PubMed until August 2023. Studies were screened by their title and abstract using the Rayyan intelligent tool for systematic reviews. Quality assessment of studies was done using the JBI critical appraisal tool. Result: Alterations in the gut microbiome are associated with gastric cancer and precancerous lesions. These alterations include reduced microbial alpha diversity, increased bacterial overgrowth, and decreased richness and evenness of gastric bacteria. Helicobacter pylori infection is associated with reduced richness and evenness of gastric bacteria, while eradication only partially restores microbial diversity. The gut microbiome also affects the response to cancer treatments, with higher abundances of Lactobacillus associated with better response to anti-PD-1/PD-L1 immunotherapy and more prolonged progression-free survival. Antibiotic-induced gut microbiota dysbiosis can reduce the anti-tumor efficacy of 5-Fluorouracil treatment, while probiotics did not significantly enhance it. A probiotic combination containing Bifidobacterium infantis, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereus can reduce inflammation, enhance immunity, and restore a healthier gut microbial balance in gastric cancer patients after partial gastrectomy. Conclusion: Probiotics and targeted interventions to modulate the gut microbiome have shown promising results in cancer prevention and treatment efficacy.Systematic review registration: https://osf.io/6vcjp.

13.
Front Nutr ; 11: 1411374, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39171106

RESUMEN

Childhood obesity presents a serious health concern associated with gut microbiota alterations. Dietary interventions targeting the gut microbiota have emerged as promising strategies for managing obesity in children. This study aimed to elucidate the impact of stachyose (STS) supplementation on the gut microbiota composition and metabolic processes in obese children. Fecal samples were collected from 40 obese children (20 boys and 20 girls) aged between 6 and 15 and in vitro fermentation was conducted with or without the addition of STS, respectively, followed by 16S rRNA amplicon sequencing and analysis of short-chain fatty acids (SCFAs) and gases. Notably, our results revealed that STS supplementation led to significant alterations in gut microbiota composition, including an increase in the abundance of beneficial bacteria such as Bifidobacterium and Faecalibacterium, and a decrease in harmful bacteria including Escherichia-Shigella, Parabacteroides, Eggerthella, and Flavonifractor. Moreover, STS supplementation resulted in changes in SCFAs production, with significant increases in acetate levels and reductions in propionate and propionate, while simultaneously reducing the generation of gases such as H2S, H2, and NH3. The Area Under the Curve (AUC)-Random Forest algorithm and PICRUSt 2 were employed to identify valuable biomarkers and predict associations between the gut microbiota, metabolites, and metabolic pathways. The results not only contribute to the elucidation of STS's modulatory effects on gut microbiota but also underscore its potential in shaping metabolic activities within the gastrointestinal environment. Furthermore, our study underscores the significance of personalized nutrition interventions, particularly utilizing STS supplementation, in the management of childhood obesity through targeted modulation of gut microbial ecology and metabolic function.

14.
Cancer Lett ; 598: 217096, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-38969161

RESUMEN

Metabolic derivatives of numerous microorganisms inhabiting the human gut can participate in regulating physiological activities and immune status of the lungs through the gut-lung axis. The current well-established microbial metabolites include short-chain fatty acids (SCFAs), tryptophan and its derivatives, polyamines (PAs), secondary bile acids (SBAs), etc. As the study continues to deepen, the critical function of microbial metabolites in the occurrence and treatment of lung cancer has gradually been revealed. Microbial derivates can enter the circulation system to modulate the immune microenvironment of lung cancer. Mechanistically, oncometabolites damage host DNA and promote the occurrence of lung cancer, while tumor-suppresive metabolites directly affect the immune system to combat the malignant properties of cancer cells and even show considerable application potential in improving the efficacy of lung cancer immunotherapy. Considering the crosstalk along the gut-lung axis, in-depth exploration of microbial metabolites in patients' feces or serum will provide novel guidance for lung cancer diagnosis and treatment selection strategies. In addition, targeted therapeutics on microbial metabolites are expected to overcome the bottleneck of lung cancer immunotherapy and alleviate adverse reactions, including fecal microbiota transplantation, microecological preparations, metabolite synthesis and drugs targeting metabolic pathways. In summary, this review provides novel insights and explanations on the intricate interplay between gut microbial metabolites and lung cancer development, and immunotherapy through the lens of the gut-lung axis, which further confirms the possible translational potential of the microbiome metabolome in lung cancer treatment.


Asunto(s)
Microbioma Gastrointestinal , Inmunoterapia , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/microbiología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Inmunoterapia/métodos , Microambiente Tumoral/inmunología , Ácidos Grasos Volátiles/metabolismo , Animales , Triptófano/metabolismo , Pulmón/microbiología , Pulmón/inmunología , Pulmón/metabolismo
15.
Mol Nutr Food Res ; 68(15): e2300845, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38966885

RESUMEN

SCOPE: The overall changes of colon under nonalcoholic fatty liver disease (NAFLD) remain to be further elucidated. METHODS AND RESULTS: This study establishes a mouse model of NAFLD through a long-term Gubra Amylin-nonalcoholic steatohepatitis (NASH) diet (GAN diet). The results show that GAN diet significantly induces weight gain, liver steatosis, colonic oxidative stress, and lipid accumulation in blood, liver, and adipose tissue in mice. GAN feeding reduces the diversity of the gut microbiota, alters the composition and abundance of the gut microbiota, and leads to an increase in microbial metabolites such as long-chain fatty acids (LCFAs) and secondary bile acids (BAs), as well as a decrease in short-chain fatty acids (SCFAs). The RNA-seq and immunofluorescence results reveal that the GAN diet alters the expression of proteins and their coding genes involved in oxidative stress, immune response, and barrier function in colon tissue, such as lipocalin-2 (Lcn2, p < 0.05), heme oxygenase-1 (HO-1/Hmox1, p < 0.05), interferon-gamma (IFN-γ), and claudin-3/7. In addition, correlation analysis indicates a strong correlation between the changes in gut microbiota and lipid biomarkers. Additionally, the expression of immune related genes in colon tissue is related to the LCFAs produced by microbial metabolism. CONCLUSION: GAN-induced NAFLD is related to microbiota and its metabolic imbalance, oxidative stress, immune disorders, and impaired barrier function in colon.


Asunto(s)
Colon , Disbiosis , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Estrés Oxidativo , Animales , Enfermedad del Hígado Graso no Alcohólico/etiología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Estrés Oxidativo/efectos de los fármacos , Colon/metabolismo , Colon/patología , Colon/efectos de los fármacos , Masculino , Ratones , Dieta , Hígado/metabolismo , Hígado/efectos de los fármacos , Modelos Animales de Enfermedad
16.
Mini Rev Med Chem ; 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38982701

RESUMEN

Globally, one of the most prevalent cancers is colorectal cancer (CRC). Chemotherapy and surgery are two common conventional CRC therapies that are frequently ineffective and have serious adverse effects. Thus, there is a need for complementary and different therapeutic approaches. The use of microbial metabolites to trigger epigenetic alterations as a way of preventing CRC is one newly emerging field of inquiry. Small chemicals called microbial metabolites, which are made by microbes and capable of altering host cell behaviour, are created. Recent research has demonstrated that these metabolites can lead to epigenetic modifications such as histone modifications, DNA methylation, and non-coding RNA regulation, which can control gene expression and affect cellular behaviour. This review highlights the current knowledge on the epigenetic modification for cancer treatment, immunomodulatory and anti-carcinogenic attributes of microbial metabolites, gut epigenetic targeting system, and the role of dietary fibre and gut microbiota in cancer treatment. It also focuses on short-chain fatty acids, especially butyrates (which are generated by microbes), and their cancer treatment perspective, challenges, and limitations, as well as state-of-the-art research on microbial metabolites-induced epigenetic changes for CRC inhibition. In conclusion, the present work highlights the potential of microbial metabolites-induced epigenetic modifications as a novel therapeutic strategy for CRC suppression and guides future research directions in this dynamic field.

17.
Nutrients ; 16(14)2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39064765

RESUMEN

The human microbiome functions as a separate organ in a symbiotic relationship with the host. Disruption of this host-microbe symbiosis can lead to serious health problems. Modifications to the composition and function of the microbiome have been linked to changes in host metabolic outcomes. Industrial lifestyles with high consumption of processed foods, alcoholic beverages and antibiotic use have significantly altered the gut microbiome in unfavorable ways. Therefore, understanding the causal relationship between the human microbiome and host metabolism will provide important insights into how we can better intervene in metabolic health. In this review, I will discuss the potential use of the human microbiome as a therapeutic target to improve host metabolism.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Metabólicas , Humanos , Microbioma Gastrointestinal/fisiología , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/terapia , Simbiosis , Disbiosis , Probióticos/uso terapéutico , Interacciones Microbiota-Huesped/fisiología
18.
Int Immunopharmacol ; 138: 112617, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38972213

RESUMEN

Severe steatosis in donor livers is contraindicated for transplantation due to the high risk of ischemia-reperfusion injury (IRI). Although Ho-1 gene-modified bone marrow mesenchymal stem cells (HO-1/BMMSCs) can mitigate IRI, the role of gut microbiota and metabolites in this protection remains unclear. This study aimed to explore how gut microbiota and metabolites contribute to HO-1/BMMSCs-mediated protection against IRI in severe steatotic livers. Using rat models and cellular models (IAR20 and THLE-2 cells) of steatotic liver IRI, this study revealed that ischemia-reperfusion led to significant liver and intestinal damage, heightened immune responses, impaired liver function, and altered gut microbiota and metabolite profiles in rats with severe steatosis, which were partially reversed by HO-1/BMMSCs transplantation. Integrated microbiome and metabolome analyses identified gut microbial metabolite oleanolic acid as a potential protective agent against IRI. Experimental validation showed that oleanolic acid administration alone alleviated IRI and inhibited ferroptosis in both rat and cellular models. Network pharmacology and molecular docking implicated KEAP1/NRF2 pathway as a potential target of oleanolic acid. Indeed, OA experimentally upregulated NRF2 activity, which underlies its inhibition of ferroptosis and protection against IRI. The gut microbial metabolite OA protects against IRI in severe steatotic liver by promoting NRF2 expression and activity, thereby inhibiting ferroptosis.


Asunto(s)
Hígado Graso , Microbioma Gastrointestinal , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2 , Ácido Oleanólico , Daño por Reperfusión , Animales , Humanos , Masculino , Ratas , Elementos de Respuesta Antioxidante , Línea Celular , Modelos Animales de Enfermedad , Hígado Graso/tratamiento farmacológico , Ferroptosis/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Hígado/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Trasplante de Células Madre Mesenquimatosas , Factor 2 Relacionado con NF-E2/metabolismo , Ácido Oleanólico/farmacología , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos
19.
J Environ Manage ; 365: 121632, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38950506

RESUMEN

Hermetia illucens larvae showcases remarkable bioremediation capabilities for both antibiotics and heavy metal contaminants. However, the distinctions in larval intestinal microbiota arising from the single and combined effects of antibiotics and heavy metals remain poorly elucidated. In this study, we delved into the details of larval intestinal bacterial communities and microbial metabolites when exposed to single and combined contaminants of oxytetracycline (OTC) and hexavalent chromium (Cr(VI)). After conversion, single contaminant-spiked substrate showed 75.5% of OTC degradation and 95.2% of Cr(VI) reductiuon, while combined contaminant-spiked substrate exhibited 71.3% of OTC degradation and 93.4% of Cr(VI) reductiuon. Single and combined effects led to differences in intestinal bacterial communities, mainly reflected in the genera of Enterococcus, Pseudogracilibacillus, Gracilibacillus, Wohlfahrtiimonas, Sporosarcina, Lysinibacillus, and Myroide. Moreover, these effects also induced differences across various categories of microbial metabolites, which categorized into amino acid and its metabolites, benzene and substituted derivatives, carbohydrates and its metabolites, heterocyclic compounds, hormones and hormone-related compounds, nucleotide and its metabolites, and organic acid and its derivatives. In particular, the differences induced OTC was greater than that of Cr(VI), and combined effects increased the complexity of microbial metabolism compared to that of single contaminant. Correlation analysis indicated that the bacterial genera, Preudogracilibacillus, Enterococcus, Sporosarcina, Lysinibacillus, Wohlfahrtiimonas, Ignatzschineria, and Fusobacterium exhibited significant correlation with significant differential metabolites, these might be used as indicators for the resistance and bioremediation of OTC and Cr(VI) contaminants. These findings are conducive to further understanding that the metabolism of intestinal microbiota determines the resistance of Hermetia illucens to antibiotics and heavy metals.


Asunto(s)
Antibacterianos , Biodegradación Ambiental , Microbioma Gastrointestinal , Larva , Metales Pesados , Animales , Antibacterianos/farmacología , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Microbioma Gastrointestinal/efectos de los fármacos , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Cromo/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-39004537

RESUMEN

The gut microbiota plays a crucial role in maintaining homeostasis and promoting health. A growing number of studies have indicated that gut microbiota can affect cancer development, prognosis, and treatment through their metabolites. By remodeling the tumor microenvironment and regulating tumor immunity, gut microbial metabolites significantly influence the efficacy of anticancer therapies, including chemo-, radio-, and immunotherapy. Several novel therapies that target gut microbial metabolites have shown great promise in cancer models. In this review, we summarize the current research status of gut microbial metabolites in cancer, aiming to provide new directions for future tumor therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA