Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Gastrointestin Liver Dis ; 33(1): 115-122, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38554422

ABSTRACT

The role of gut microbiota in autoimmune disorders like multiple sclerosis is gaining attention. Multiple sclerosis is characterized by inflammation, demyelination, and neurodegeneration in the central nervous system. Alterations in gut microbiota have been linked to multiple sclerosis development, with decreased beneficial bacteria and increased harmful species. The gut-brain axis is a complex interface influencing bidirectional interactions between the gut and the brain. Dysbiosis, an imbalance in gut microbiota, has been associated with autoimmune diseases. The influence of gut microbiota in multiple sclerosis is reversible, making it a potential therapeutic target. Probiotics, prebiotics, and fecal microbiota transplantation have shown promise in multiple sclerosis treatment, with positive effects on inflammation and immune regulation. Immunoglobulin Y (IgY) supplements derived from chicken egg yolk have potential as nutraceuticals or dietary supplements. IgY technology has been effective against various infections, and studies have highlighted its role in modulating gut microbiota and immune responses. Clinical trials using IgY supplements in multiple sclerosis are limited but have shown positive outcomes, including reduced symptoms, and altered immune responses. Future research directions involve understanding the mechanisms of IgY's interaction with gut microbiota, optimal dosage determination, and long-term safety assessments. Combining IgY therapy with other interventions and investigating correlations between microbiota changes and clinical outcomes are potential avenues for advancing multiple sclerosis treatment with IgY supplements.


Subject(s)
Autoimmune Diseases , Immunoglobulins , Multiple Sclerosis , Probiotics , Humans , Multiple Sclerosis/therapy , Dysbiosis/microbiology , Dysbiosis/therapy , Dietary Supplements/adverse effects , Probiotics/therapeutic use , Inflammation
2.
NPJ Biofilms Microbiomes ; 9(1): 99, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38092763

ABSTRACT

Spinal cord injury (SCI) can reshape gut microbial composition, significantly affecting clinical outcomes in SCI patients. However, mechanisms regarding gut-brain interactions and their clinical implications have not been elucidated. We hypothesized that short-chain fatty acids (SCFAs), intestinal microbial bioactive metabolites, may significantly affect the gut-brain axis and enhance functional recovery in a mouse model of SCI. We enrolled 59 SCI patients and 27 healthy control subjects and collected samples. Thereafter, gut microbiota and SCFAs were analyzed using 16 S rDNA sequencing and gas chromatography-mass spectrometry, respectively. We observed an increase in Actinobacteriota abundance and a decrease in Firmicutes abundance. Particularly, the SCFA-producing genera, such as Faecalibacterium, Megamonas, and Agathobacter were significantly downregulated among SCI patients compared to healthy controls. Moreover, SCI induced downregulation of acetic acid (AA), propionic acid (PA), and butyric acid (BA) in the SCI group. Fecal SCFA contents were altered in SCI patients with different injury course and injury segments. Main SCFAs (AA, BA, and PA) were administered in combination to treat SCI mice. SCFA supplementation significantly improved locomotor recovery in SCI mice, enhanced neuronal survival, promoted axonal formation, reduced astrogliosis, and suppressed microglial activation. Furthermore, SCFA supplementation downregulated NF-κB signaling while upregulating neurotrophin-3 expression following SCI. Microbial sequencing and metabolomics analysis showed that SCI patients exhibited a lower level of certain SCFAs and related bacterial strains than healthy controls. SCFA supplementation can reduce inflammation and enhance nourishing elements, facilitating the restoration of neurological tissues and the improvement of functional recuperation. Trial registration: This study was registered in the China Clinical Trial Registry ( www.chictr.org.cn ) on February 13, 2017 (ChiCTR-RPC-17010621).


Subject(s)
Dysbiosis , Spinal Cord Injuries , Humans , Mice , Animals , Dysbiosis/microbiology , Fatty Acids, Volatile , Acetic Acid/metabolism , Bacteria/genetics , Bacteria/metabolism , Butyric Acid/metabolism
3.
Shock ; 60(4): 585-593, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37548929

ABSTRACT

ABSTRACT: The Earth's population is aging, and by 2050, one of six people will be 65 years or older. Therefore, proper treatment of injuries that disproportionately impact people of advanced age will be more important. Clinical studies reveal people 65 years or older account for 16.5% of all burn injuries and experience higher morbidity, including neurocognitive decline, and mortality that we and others believe are mediated, in part, by heightened intestinal permeability. Herein, we used our clinically relevant model of scald burn injury in young and aged mice to determine whether age and burn injury cooperate to induce heightened colonic damage, alterations to the fecal microbiome, and whether resultant changes in the microbiome correlate with neuroinflammation. We found that aged, burn-injured mice have an increase in colonic lymphoid aggregates, inflammation, and proinflammatory chemokine expression when compared with young groups and sham-injured aged mice. We then performed fecal microbiota sequencing and found a striking reduction in gut protective bacterial taxa, including Akkermansia , in the aged burn group compared with all other groups. This reduction correlated with an increase in serum fluorescein isothiocyanate-Dextran administered by gavage, indicating heightened intestinal permeability. Furthermore, loss of Akkermansia was highly correlated with increased messenger RNA expression of neuroinflammatory markers in the brain, including chemokine ligand 2, TNF-α, CXC motif ligand 1, and S100 calcium-binding protein A8. Finally, we discovered that postburn alterations in the microbiome correlated with measures of strength in all treatment groups, and those that performed better on the rotarod and hanging wire tests had higher abundance of Akkermansia than those that performed worse. Taken together, these findings indicate that loss of protective bacteria after burn injury in aged mice contributes to alterations in the colon, gut leakiness, neuroinflammation, and strength. Therefore, supplementation of protective bacteria, such as Akkermansia , after burn injury in aged patients may have therapeutic benefit.


Subject(s)
Burns , Microbiota , Humans , Aged , Neuroinflammatory Diseases , Dysbiosis/microbiology , Ligands , Burns/microbiology , Bacteria/genetics , Chemokines , Colon
4.
Nutrients ; 15(14)2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37513683

ABSTRACT

Autism Spectrum Disorder (ASD) is characterized by varying degrees of difficulty in social interaction and communication. These deficits are often associated with gastrointestinal symptoms, indicating alterations in both intestinal microbiota composition and metabolic activities. The intestinal microbiota influences the function and development of the nervous system. In individuals with ASD, there is an increase in bacterial genera such as Clostridium, as well as species involved in the synthesis of branched-chain amino acids (BCAA) like Prevotella copri. Conversely, decreased amounts of Akkermansia muciniphila and Bifidobacterium spp. are observed. Epigallocatechin-3-gallate (EGCG) is one of the polyphenols with the greatest beneficial activity on microbial growth, and its consumption is associated with reduced psychological distress. Therefore, the objective of this review is to analyze how EGCG and its metabolites can improve the microbial dysbiosis present in ASD and its impact on the pathology. The analysis reveals that EGCG inhibits the growth of pathogenic bacteria like Clostridium perfringens and Clostridium difficile. Moreover, it increases the abundance of Bifidobacterium spp. and Akkermansia spp. As a result, EGCG demonstrates efficacy in increasing the production of metabolites involved in maintaining epithelial integrity and improving brain function. This identifies EGCG as highly promising for complementary treatment in ASD.


Subject(s)
Autism Spectrum Disorder , Gastrointestinal Microbiome , Humans , Child , Autism Spectrum Disorder/microbiology , Dysbiosis/microbiology , Bacteria
5.
Arch Microbiol ; 205(7): 257, 2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37280443

ABSTRACT

Concept of microorganisms has largely been perceived from their pathogenic view point. Nevertheless, it is being gradually revisited in terms of its significance to human health and now appears to be the most dominant force that shapes the immune system of the human body and also determines an individual's predisposition to diseases. Human inhabits bacterial diversity (which is predominant among all microbial communities in human body) occupying 0.3% of body mass, known as microbiota. On birth, a part of microbiota that child obtains is essentially a mother's legacy. So, the review was initiated with this critical topic of microbiotal inheritance. Since, each body site has distinct physiological specifications; therefore, they contain discrete microbiome composition that has been separately discussed along with dysbiosis-induced pathologies originating in different body organs. Factors affecting microbiome composition and may cause dysbiosis like antibiotics, delivery, feeding method etc. as well as the strategies that immune system adopts to prevent dysbiosis have been highlighted. We also tried to bring into attention the topic of dysbiosis induced biofilms, that enables cohort to survive stresses, evolve, disseminate and infection resurgence that is still in dormancy. Eventually, we put spotlight on microbiome significance in medical therapeutics. We didn't merely confine article to gut microbiota, that is being studied more extensively. Numerous community forms at diverse body sites are inter-related, and being exposed to awfully variable perturbations appear to be challenging to evaluate perturbation risks holistically. All aspects have been elaborately discussed to achieve a global depiction of human microbiota in order to meet urgent necessity for protocol standardisation. Demonstrates that environmental challenges (antibiotic use, alterations in diet, stress, smoking etc.) might cause dysbiosis i.e. transition of healthy microbiome composition to the one in which pathogenic microorganisms become more abundant, and eventually results in an infected state.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Bacteria/genetics , Biofilms , Dysbiosis/microbiology , Microbiota/physiology , Infant, Newborn
6.
Pharm Biol ; 61(1): 531-540, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36994999

ABSTRACT

CONTEXT: A Chinese herbal formula, Tiaopi Xiezhuo decoction (TXD), is developed from a classical Chinese prescription Sanhuang Xiexin decoction. OBJECTIVE: To investigate the regulatory effect of TXD on gut dysbiosis, as a treatment of constipation in patients with peritoneal dialysis (PD). MATERIALS AND METHODS: The chemical content of TXD was assessed by high-performance liquid chromatography. A total of 29 PD patients were enrolled and treated with TXD orally (3 g crude drug/each/twice/day) for 3 months. Blood and faecal samples were collected at the beginning and end, to determine the changes in biochemical characteristics and gut microbial composition. The stool conditions were asked to be scored. Additional 30 healthy individuals were recruited as a control for the analysis of gut microbiota. RESULTS: Although having no significant effects on serum biochemical characteristics, 3-month TXD intervention improved constipation in PD patients: decreased 80% abdominal distention (p < 0.01), increased 2.6-fold sloppy stools (p < 0.05) and eliminated hard stool completely (p < 0.01). The analysis of gut microbiota showed that, compared to the healthy group, the microbial richness was reduced in PD patients. After a 3-month TXD treatment, this reduced richness was raised, and Paraprevotella clara, Lachnospiraceae bacterium 2-146FA, Phascolarctobaterium succinatutens, Lachnospiraceae bacterium 2-1-58FAA, Fusobacterium mortiferum, and Prevotella copri were accumulated in the intestinal flora. Furthermore, the bacterial species enriched by TXD correlated with the improvement of constipation. DISCUSSION AND CONCLUSIONS: TXD treatment may improve constipation by modulating gut dysbiosis in PD patients. These findings provide data to support the further application of TXD in the adjuvant treatment of PD.


Subject(s)
Constipation , Drugs, Chinese Herbal , Dysbiosis , Gastrointestinal Microbiome , Peritoneal Dialysis , Humans , Constipation/drug therapy , Dysbiosis/drug therapy , Dysbiosis/microbiology , Feces , Peritoneal Dialysis/adverse effects , Drugs, Chinese Herbal/therapeutic use
7.
Microbiol Res ; 271: 127346, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36921399

ABSTRACT

The gut microbiome is the community of healthy, and infectious organisms in the gut and its interaction in the host gut intestine (GI) environment. The balance of microbial richness with beneficial microbes is very important to perform healthy body functions like digesting food, controlling metabolism, and precise immune function. Alternately, this microbial dysbiosis occurs due to changes in the physiochemical condition, substrate avidity, and drugs. Moreover, various categories of diet such as "plant-based", "animal-based", "western", "mediterranean", and various drugs (antibiotic and common drugs) also contribute to maintaining microbial flora inside the gut. The imbalance (dysbiosis) in the microbiota of the GI tract can cause several disorders (such as diabetes, obesity, cancer, inflammation, and so on). Recently, the major interest is to use prebiotic, probiotic, postbiotic, and herbal supplements to balance such microbial community in the GI tract. But, there has still a large gap in understanding the microbiome function, and its relation to the host diet, drugs, and herbal supplements to maintain the healthy life of the host. So, the present review is about the updates on the microbiome concerns related to diet, drug, and herbal supplements, and also gives research evidence to improve our daily habits regarding diet, drugs, and herbal supplements. Because our regular dietary plan and traditional herbal supplements can improve our health by balancing the bacteria in our gut.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Dysbiosis/microbiology , Dietary Supplements , Obesity/microbiology
8.
Pharmacol Res ; 190: 106714, 2023 04.
Article in English | MEDLINE | ID: mdl-36863429

ABSTRACT

Ischemic stroke is closely associated with gut microbiota dysbiosis and intestinal barrier dysfunction. Prebiotic intervention could modulate the intestinal microbiota, thus considered a practical strategy for neurological disorders. Puerariae Lobatae Radix-resistant starch (PLR-RS) is a potential novel prebiotic; however, its role in ischemic stroke remains unknown. This study aimed to clarify the effects and underlying mechanisms of PLR-RS in ischemic stroke. Middle cerebral artery occlusion surgery was performed to establish a model of ischemic stroke in rats. After gavage for 14 days, PLR-RS attenuated ischemic stroke-induced brain impairment and gut barrier dysfunction. Moreover, PLR-RS rescued gut microbiota dysbiosis and enriched Akkermansia and Bifidobacterium. We transplanted the fecal microbiota from PLR-RS-treated rats into rats with ischemic stroke and found that the brain and colon damage were also ameliorated. Notably, we found that PLR-RS promoted the gut microbiota to produce a higher level of melatonin. Intriguingly, exogenous gavage of melatonin attenuated ischemic stroke injury. In particular, melatonin attenuated brain impairment via a positive co-occurrence pattern in the intestinal microecology. Specific beneficial bacteria served as leaders or keystone species to promoted gut homeostasis, such as Enterobacter, Bacteroidales_S24-7_group, Prevotella_9, Ruminococcaceae and Lachnospiraceae. Thus, this new underlying mechanism could explain that the therapeutic efficacy of PLR-RS on ischemic stroke at least partly attributed to gut microbiota-derived melatonin. In summary, improving intestinal microecology by prebiotic intervention and melatonin supplementation in the gut were found to be effective therapies for ischemic stroke.


Subject(s)
Central Nervous System Depressants , Gastrointestinal Microbiome , Ischemic Stroke , Melatonin , Pueraria , Animals , Rats , Dysbiosis/microbiology , Ischemic Stroke/drug therapy , Melatonin/pharmacology , Melatonin/therapeutic use , Prebiotics , Resistant Starch , Central Nervous System Depressants/pharmacology , Central Nervous System Depressants/therapeutic use
9.
World J Gastroenterol ; 29(6): 967-996, 2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36844143

ABSTRACT

A growing body of evidence from multiple areas proposes that periodontal disease, accompanied by oral inflammation and pathological changes in the microbiome, induces gut dysbiosis and is involved in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). A subgroup of NAFLD patients have a severely progressive form, namely nonalcoholic steatohepatitis (NASH), which is characterized by histological findings that include inflammatory cell infiltration and fibrosis. NASH has a high risk of further progression to cirrhosis and hepatocellular carcinoma. The oral microbiota may serve as an endogenous reservoir for gut microbiota, and transport of oral bacteria through the gastro-intestinal tract can set up a gut microbiome dysbiosis. Gut dysbiosis increases the production of potential hepatotoxins, including lipopolysaccharide, ethanol, and other volatile organic compounds such as acetone, phenol and cyclopentane. Moreover, gut dysbiosis increases intestinal permeability by disrupting tight junctions in the intestinal wall, leading to enhanced translocation of these hepatotoxins and enteric bacteria into the liver through the portal circulation. In particular, many animal studies support that oral administration of Porphyromonas gingivalis, a typical periodontopathic bacterium, induces disturbances in glycolipid metabolism and inflammation in the liver with gut dysbiosis. NAFLD, also known as the hepatic phenotype of metabolic syndrome, is strongly associated with metabolic complications, such as obesity and diabetes. Periodontal disease also has a bidirectional relationship with metabolic syndrome, and both diseases may induce oral and gut microbiome dysbiosis with insulin resistance and systemic chronic inflammation cooperatively. In this review, we will describe the link between periodontal disease and NAFLD with a focus on basic, epidemiological, and clinical studies, and discuss potential mechanisms linking the two diseases and possible therapeutic approaches focused on the microbiome. In conclusion, it is presumed that the pathogenesis of NAFLD involves a complex crosstalk between periodontal disease, gut microbiota, and metabolic syndrome. Thus, the conventional periodontal treatment and novel microbiome-targeted therapies that include probiotics, prebiotics and bacteriocins would hold great promise for preventing the onset and progression of NAFLD and subsequent complications in patients with periodontal disease.


Subject(s)
Metabolic Syndrome , Microbiota , Non-alcoholic Fatty Liver Disease , Periodontitis , Animals , Dysbiosis/microbiology , Fibrosis , Inflammation/pathology , Liver/pathology , Metabolic Syndrome/complications , Non-alcoholic Fatty Liver Disease/pathology , Periodontitis/therapy , Periodontitis/complications , Periodontitis/metabolism , Intestines
10.
Front Endocrinol (Lausanne) ; 14: 1122709, 2023.
Article in English | MEDLINE | ID: mdl-36814581

ABSTRACT

Background: Polycystic ovarian syndrome (PCOS) is a common endocrine disorder characterized by hyperandrogenism, ovarian dysfunction and polycystic ovarian morphology. Gut microbiota dysbiosis and metabolite are associated with PCOS clinical parameters. Yulin Tong Bu formula (YLTB), a traditional Chinese medicine formula, has been recently indicated to be capable of ameliorating polycystic ovary symptoms and correcting abnormal glucose metabolism. However, the therapeutic mechanism of YLTB on PCOS has not been fully elucidated. Methods: A pseudo sterile mouse model was established during this four-day acclimatization phase by giving the animals an antibiotic cocktail to remove the gut microbiota. Here, the therapeutic effects of YLTB on PCOS were investigated using dehydroepiandrosterone plus high-fat diet-induced PCOS mice model. Female prepuberal mice were randomly divided into three groups; namely, the control group, PCOS group and YLTB (38.68 g·kg-1·day-1) group. To test whether this effect is associated with the gut microbiota, we performed 16S rRNA sequencing studies to analyze the fecal microbiota of mice. The relationships among metabolites, gut microbiota, and PCOS phenotypes were further explored by using Spearman correlation analysis. Then, the effect of metabolite ferulic acid was then validated in PCOS mice. Results: Our results showed that YLTB treatment ameliorated PCOS features (ovarian dysfunction, delayed glucose clearance, decreased insulin sensitivity, deregulation of glucolipid metabolism and hormones, etc.) and significantly attenuated PCOS gut microbiota dysbiosis. Spearman correlation analysis showed that metabolites such as ferulic acid and folic acid are negatively correlated with PCOS clinical parameters. The effect of ferulic acid was similar to that of YLTB. In addition, the bacterial species such as Bacteroides dorei and Bacteroides fragilis were found to be positively related to PCOS clinical parameters, using the association study analysis. Conclusion: These results suggest that YLTB treatment systematically regulates the interaction between the gut microbiota and the associated metabolites to ameliorate PCOS, providing a solid theoretical basis for further validation of YLTB effect on human PCOS trials.


Subject(s)
Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Mice , Female , Humans , Animals , Polycystic Ovary Syndrome/metabolism , Gastrointestinal Microbiome/physiology , Dysbiosis/microbiology , RNA, Ribosomal, 16S
11.
Comb Chem High Throughput Screen ; 26(5): 892-905, 2023.
Article in English | MEDLINE | ID: mdl-35786331

ABSTRACT

For several decades, studies have reported that n-3 polyunsaturated fatty acids (PUFAs) play a beneficial role in cardiovascular, immune, cognitive, visual, mental and metabolic health. The mammalian intestine is colonized by microbiota, including bacteria, archaea, viruses, protozoans, and fungi. The composition of the gut microbiota is influenced by long-term dietary habits, disease-associated dysbiosis, and the use of antibiotics. Accumulating evidence suggests a relationship between n-3 PUFAs and the gut microbiota. N-3 PUFAs can alter the diversity and abundance of the gut microbiome, and gut microbiota can also affect the metabolism and absorption of n-3 PUFAs. Changes in the populations of certain gut microbiota can lead to negative effects on inflammation, obesity, and metabolic diseases. An imbalanced consumption of n-3/n-6 PUFAs may lead to gut microbial dysbiosis, in particular, a significant increase in the ratio of Firmicutes to Bacteroidetes, which eventually results in being overweight and obesity. N-3 PUFA deficiency disrupts the microbiota community in metabolic disorders. In addition, accumulating evidence indicates that the interplay between n-3 PUFAs, gut microbiota, and immune reactions helps to maintain the integrity of the intestinal wall and interacts with host immune cells. Supplementation with n-3 PUFAs may be an effective therapeutic measure to restore gut microbiota homeostasis and correct metabolic disturbances associated with modern chronic diseases. In particular, marine extracts from seaweed contain a considerable dry weight of lipids, including n-3 PUFAs such as eicosapentaenoic acid (EPA, C20: 5) and docosahexaenoic acid (DHA, C22: 6). This review describes how gut microbiota function in intestinal health, how n-3 PUFAs interact with the gut microbiota, and the potential of n-3 PUFAs to influence the gut-brain axis, acting through gut microbiota composition.


Subject(s)
Fatty Acids, Omega-3 , Gastrointestinal Microbiome , Metabolic Diseases , Dysbiosis/drug therapy , Dysbiosis/microbiology , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Unsaturated , Metabolic Diseases/drug therapy , Obesity/drug therapy , Obesity/metabolism , Humans
12.
Inflamm Bowel Dis ; 29(3): 470-479, 2023 03 01.
Article in English | MEDLINE | ID: mdl-35851921

ABSTRACT

Inflammatory bowel disease (IBD), a disorder characterized by chronic inflammation of the gastrointestinal (GI) tract and a range of adverse health effects including diarrhea, abdominal pain, vomiting, and bloody stools, affects nearly 3.1 million genetically susceptible adults in the United States today. Although the etiology of IBD remains unclear, genetics, stress, diet, and gut microbiota dysbiosis- especially in immunocompromised individuals- have been identified as possible causes of disease. Although previous research has largely focused on the role of bacteria in IBD pathogenesis, recently observed alterations of fungal load and biodiversity in the GI tract of afflicted individuals suggest interkingdom interactions amongst different gut microbial communities, particularly between bacteria and fungi. These discoveries point to the potential utilization of treatment approaches such as antibiotics, antifungals, probiotics, and postbiotics that target both bacteria and fungi in managing IBD. In this review, we discuss the impact of specific fungi on disease pathogenesis, with a focus on the highly virulent genus Candida and how the presence of certain co-enzymes impacts its virulence. In addition, we evaluate current gut microbiome-based therapeutic approaches with the intention of better understanding the mechanisms behind novel therapies.


Recently observed alterations of fungal load in the gastrointestinal tract of IBD patients suggest interkingdom interactions amongst different gut microbial communities. These discoveries point to the potential utilization of antifungals and probiotics that target bacteria and fungi in managing IBD.


Subject(s)
Inflammatory Bowel Diseases , Mycobiome , Adult , Humans , Inflammatory Bowel Diseases/therapy , Inflammation , Bacteria , Dysbiosis/microbiology
13.
J Med Food ; 25(12): 1102-1111, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36516056

ABSTRACT

Alcoholic liver disease (ALD) is a major chronic liver disease. Chronic alcohol consumption induces dysbiosis, disruption of gut barrier function, oxidative stress, inflammation, and changes in lipid metabolism, thereby leading to ALD. In this study, we investigated whether the commercial Morinda citrifolia extract Nonitri can ameliorate ALD symptoms through the gut-liver axis. We used mice chronically administered EtOH and found a marked increase in serum endotoxin levels and biomarkers of liver pathology. Moreover, the EtOH-treated group showed significantly altered gut microbial composition particularly that of Alistipes, Bacteroides, and Muribaculum and disrupted gut barrier function. However, Nonitri improved serum parameters, restored the microbial proportions, and regulated levels of zonula occludens1, occludin, and claudin1. Furthermore, Nonitri suppressed inflammation by inhibiting endotoxin-triggered toll-like receptor 4-signaling pathway and fat deposition by reducing lipogenesis through activating AMP-activated protein kinase in the liver. Furthermore, Pearson's correlation analysis showed that gut microbiota and ALD-related markers were correlated, and Nonitri regulated these bacteria. Taken together, our results indicate that the hepatoprotective effect of Nonitri reduces endotoxin levels by improving gut health, and inhibits fat deposition by regulating lipid metabolism.


Subject(s)
Fatty Liver, Alcoholic , Liver Diseases, Alcoholic , Morinda , Mice , Animals , Fatty Liver, Alcoholic/drug therapy , Fatty Liver, Alcoholic/metabolism , Dysbiosis/microbiology , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/prevention & control , Liver/metabolism , Ethanol/metabolism , Endotoxins , Inflammation/metabolism , Mice, Inbred C57BL
14.
Nutrients ; 14(19)2022 Oct 08.
Article in English | MEDLINE | ID: mdl-36235833

ABSTRACT

As a traditional Chinese medicine, licorice is often used in functional foods for its health benefits. However, the role of gut microbiota in the efficacy of licorice has not yet been fully elucidated. We hypothesized that the involvement of intestinal flora may be a key link in licorice ethanol extract (LEE)-induced health benefits. The aim of this study was to investigate whether LEE improves hepatic lipid accumulation in obese mice fed a high-fat diet (HFD) and whether the gut microbiota plays a key role in LEE treatment. Male C57BL/6J mice were fed HFD for liver fat accumulation and then treated with LEE. The same experiments were later performed using pseudo-sterile mice to verify the importance of gut flora. Supplementation with LEE improved the obesity profile, lipid profile and liver fat accumulation in HFD mice. In addition, LEE treatment improved intestinal flora dysbiosis caused by HFD in mice, as evidenced by a decrease in the percentage of Firmicutes/Bacteroidetes and an increase in the abundance of known anti-obesity-related bacteria. However, LEE failed to exhibit a therapeutic effect in pseudo-sterile mice. The results of the cellular assay showed that glycyrrhetic acid (GA), the main conversion product of glycyrrhizin (GL), was more effective in reducing fat accumulation and intracellular TG content in hepatocytes compared to GL. In conclusion, our data suggest that LEE attenuates obesity and hepatic fat accumulation in HFD mice, which may be associated with modulating the composition of gut microbiota and the conversion of LLE by the intestinal flora.


Subject(s)
Gastrointestinal Microbiome , Glycyrrhetinic Acid , Glycyrrhiza , Animals , Diet, High-Fat/adverse effects , Dysbiosis/microbiology , Ethanol/pharmacology , Glycyrrhetinic Acid/pharmacology , Glycyrrhizic Acid/pharmacology , Lipids/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Plant Extracts/therapeutic use
15.
Front Biosci (Landmark Ed) ; 27(9): 278, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36224015

ABSTRACT

BACKGROUND: Impaired glucose regulation (IGR) represents the prediabetic state and is associated with gut microbiota (GM) dysbiosis and chronic inflammation. Tangning Ziyabitusi Tablet (TZT) is a Chinese Uyghur herbal medicine with preventative and therapeutic effects on diabetes, but its hypoglycemic mechanisms are unclear. METHODS: Thirty-six male Wistar rats were divided into the normal diet (ND) and IGR groups. The IGR group was given a high-fat diet (HFD). After the IGR model establishment, the ND group was divided into ND and ND+TZT groups, and the IGR group into IGR and IGR+TZT groups. After 8 weeks of TZT administration, 16S rRNA sequencing and untargeted metabolomics were performed on fecal samples. Mesenteric lymph nodes were also collected, and T lymphocytes separated after rats were sacrificed. Flow cytometry was used to characterize different CD4+ T cell subsets in mesenteric lymph nodes. Finally, we analyzed the correlation between GM and characteristic fecal metabolites. RESULTS: Impaired glucose tolerance and insulin resistance were improved in the IGR+TZT group when compared with the IGR group. Bacterial 16S rRNA sequencing results showed that Sobs and Chao1 indices in the IGR group were significantly decreased, but were increased in the IGR+TZT group. The relative abundance of Bacteroidetes was decreased while the relative abundance of Firmicutes was increased in the IGR group. Adlercreutzia abundance was decreased after TZT administration, while the abundance of Christensenellaceae_R-7_group, norank_f_norank_o_Clostridia_UCG-014, UCG-005, and Eubacterium_nodatum_group was increased in the IGR+TZT group. Lymph node CD4+ T cell proportions in the IGR group were significantly increased, while they were significantly decreased in the IGR+TZT group. Correlation analysis showed that tumor necrosis factor-α, interleukin-6, T helper cells (Th1, Th2, Treg), and insulin had a greater impact on GM community structure. CONCLUSIONS: TZT improved glucose tolerance and ameliorated GM dysbiosis in IGR rats. Additionally, TZT significantly modulated CD4+ T cell subset proportions in rat mesenteric lymph nodes and fecal metabolism. Moreover, correlation analysis showed that key microbiota was closely related to IGR indices. Thus, TZT modulated GM composition and immune functions of the intestinal mucosa. We provide useful information for the investigation of active mechanisms and the clinical application of TZT.


Subject(s)
Gastrointestinal Microbiome , Insulins , Animals , Dysbiosis/microbiology , Glucose/pharmacology , Hypoglycemic Agents/pharmacology , Insulins/pharmacology , Insulins/therapeutic use , Interleukin-6 , Male , RNA, Ribosomal, 16S/genetics , Rats , Rats, Wistar , T-Lymphocyte Subsets/metabolism , Tablets/pharmacology , Tablets/therapeutic use , Tumor Necrosis Factor-alpha
16.
World J Gastroenterol ; 28(27): 3370-3382, 2022 Jul 21.
Article in English | MEDLINE | ID: mdl-36158273

ABSTRACT

Colorectal cancer (CRC) is a leading cause of human mortality worldwide. As conventional anticancer therapy not always being effective, there is growing interest in innovative "drug-free" cancer treatments or interventions that improve the efficacy of established therapy. CRC is associated with microbiome alterations, a process known as dysbiosis that involves depletion and/or enrichment of particular gut bacterial species and their metabolic functions. Supplementing patient treatment with traditional probiotics (with or without prebiotics), next-generation probiotics (NGP), or postbiotics represents a potentially effective and accessible complementary anticancer strategy by restoring gut microbiota composition and/or by signaling to the host. In this capacity, restoration of the gut microbiota in cancer patients can stabilize and enhance intestinal barrier function, as well as promote anticarcinogenic, anti-inflammatory, antimutagenic or other biologically important biochemical pathways that show high specificity towards tumor cells. Potential benefits of traditional probiotics, NGP, and postbiotics include modulating gut microbiota composition and function, as well as the host inflammatory response. Their application in CRC prevention is highlighted in this review, where we consider supportive in vitro, animal, and clinical studies. Based on emerging research, NGP and postbiotics hold promise in establishing innovative treatments for CRC by conferring physiological functions via the production of dominant natural products and metabolites that provide new host-microbiota signals to combat CRC. Although favorable results have been reported, further investigations focusing on strain and dose specificity are required to ensure the efficacy and safety of traditional probiotics, NGP, and postbiotics in CRC prevention and treatment.


Subject(s)
Biological Products , Colorectal Neoplasms , Complementary Therapies , Probiotics , Animals , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/prevention & control , Dysbiosis/microbiology , Humans , Prebiotics , Probiotics/therapeutic use
17.
Food Funct ; 13(12): 6648-6664, 2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35642970

ABSTRACT

Poria cocos (P. cocos) has been traditionally used as folk medicine and functional food in China for more than 2000 years. The water-soluble polysaccharide is the main component of P. cocos decoction. The effects and mechanisms of the water-soluble polysaccharide from P. cocos (PCWP) were investigated in chronic sleep deprivation (CSD)-induced anxiety in rats. CSD induced anxiety, gut dysbiosis, and inflammatory responses, and reduced neurotransmitter levels, whereas PCWP intervention ameliorated anxiety-like behaviors, increased the levels of 5-hydroxytryptamine, dopamine, norepinephrine, and γ-aminobutyric acid in the hypothalamus, regulated gastrointestinal peptide levels, reduced inflammatory factors, and inhibited the tumor necrosis factor (TNF)-α/nuclear factor (NF)-κB signaling pathway in rats with CSD. The changes in the intestinal flora composition were determined using 16S rDNA sequencing, and indicated that PCWP significantly improved species richness and diversity in the intestinal flora of rats with anxiety, and adjusted the abundance of the following dysregulated bacteria closer to that of the normal group: Rikenellaceae_RC9_gut_group, Ruminococcus, Prevotellaceae_UCG-001, Prevotellaceae_NK3B31_group, Fusicatenibacter. Metabolomics was used to analyze fecal samples to identify significantly altered metabolites in the PCWP-treated groups. Thirty-eight PCWP-related metabolites and four metabolic pathways such as sphingolipid metabolism, taurine and hypotaurine metabolism, vitamin B6 metabolism, and glycerophospholipid metabolism were explored. The results of serum metabolomics showed that 26 biomarkers were significantly changed after PCWP intervention compared with the model group. The regulatory effects of metabolic pathway enrichment on sphingolipid, phenylalanine, and taurine and hypotaurine metabolism, and validation results showed that PCWP intervention regulated the activity of enzymes involved in the above metabolic pathways. A strong correlation between intestinal bacteria and potential biomarkers was found. Our findings present new evidence supporting the potential effect of PCWP in preventing the progression of anxiety by inhibiting the TNF-α/NF-κB signaling pathway, alleviating metabolic disorders, and ameliorating the gut microflora imbalance.


Subject(s)
Metabolic Diseases , Wolfiporia , Animals , Anxiety/drug therapy , Biomarkers/metabolism , Dysbiosis/microbiology , Intestinal Diseases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Polysaccharides/pharmacology , Rats , Signal Transduction , Sleep Deprivation , Sphingolipids , Taurine/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Water/pharmacology , Wolfiporia/chemistry
18.
Food Chem ; 393: 133438, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35696951

ABSTRACT

In this study, the mitigative effect of Rhodomyrtus tomentosa (Ait.) Hassk fruit extract rich in phenolic compounds (RTE) on high fat diet (HFD)-induced intestinal barrier dysfunction of mice and the underlying mechanism were explored. The results revealed that RTE supplementation obviously improved gut microbiota dysbiosis induced by HFD, which was evidenced by elevated alpha diversity, suppressed Firmicutes/Bacteroidetes ratio, enriched short-chain fatty acid-producing bacteria (Odoribacter, Parabacteroides, Blautia and Akkermansia), and depleted harmful bacteria (Helicobacter, norank_f_ Desulfovibrionaceae and Mucispirillum). RTE intervention mitigated intestinal barrier dysfunction and inflammation by elevating tight junction proteins expression levels and decreasing proinflammatory cytokines levels. Furthermore, RTE administration inhibited the HFD-induced trigger of the lipopolysaccharide-toll-like receptor 4-nuclear factor kappa-B (LPS-TLR4-NF-κB) pathway in colonic tissue. Therefore, RTE supplementation may be an effective way to protect the intestinal tract in HFD-induced obese individuals.


Subject(s)
Gastrointestinal Diseases , Myrtaceae , Animals , Diet, High-Fat , Dysbiosis/microbiology , Fruit/metabolism , Inflammation/drug therapy , Inflammation/genetics , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Myrtaceae/genetics , Myrtaceae/metabolism , NF-kappa B/metabolism , Phenols , Plant Extracts/pharmacology
19.
Theranostics ; 12(8): 3637-3655, 2022.
Article in English | MEDLINE | ID: mdl-35664075

ABSTRACT

Rationale: The high fat and sucrose diet, known as the obesogenic diet (OD), has been related to low-grade chronic inflammation and neurodevelopmental disorders. Emerging evidence suggests that OD influences cognitive and social function via the gut-brain axis. However, the effects of OD during adolescence on future health have been unclear. Meanwhile, the underlying mechanisms and effective interventions are not fully understood. Polysaccharides, one of the most abundant substances in the Eucommiae cortex, exhibit potential immunomodulatory and neuroprotective effects. Here, we aimed to investigate the impact of OD on adolescents, explore the modulating roles of Eucommiae cortex polysaccharides (EPs) on OD-induced behavioral dysfunction, and elucidate the underlying molecular mechanisms. Methods: In the present study, four-week-old mice were fed with OD for four weeks to simulate persistent OD in adolescents. The behavioral features were accessed by open field test and Morris water maze. The gut bacterial structure was identified by 16S rRNA gene amplicon sequencing. The gene and protein expression in colonic tissues and hippocampus were detected by qRT-PCR, immunoblotting, enzyme-linked immunosorbent assay, and immunofluorescence staining. Detection of biological metabolites in serum and hippocampal tissues was performed by widely targeted metabolomics and targeted metabolomics. Results: We found that OD-fed mice showed cognitive and social-behavioral deficits accompanied by gut dysbiosis and systematic tryptophan (Trp) metabolism disorders, which increased kynurenine (Kyn) concentration in the hippocampus. Bacteria-derived lipopolysaccharide (LPS, endotoxin) induced microglia-mediated neuroinflammation, directing the metabolism of Kyn in the hippocampus toward quinolinic acid (QA), which led to glutamate-mediated hyperactivation of mossy cells (MCs) in hippocampal hilus. Furthermore, OD impaired parvalbumin (PV) interneurons-related local circuits in the hippocampal granule cell layer. These resulted in hippocampal neurogenesis deficits and related behavioral dysfunction in mice. EPs supplementation ameliorated OD-induced gut dysbiosis, as evidenced by inhibiting the expansion of Escherichia coli (E.coli) and reducing the concentration of LPS in colonic contents and serum, thereby inhibiting the subsequent neuroinflammation. In addition, oral EPs suppressed the peripheral Kyn pathway to reduce the concentration of QA and glutamic acid in the hippocampus of OD-fed mice, thereby rescuing the glutamic acid-triggered neuroexcitotoxicity. These contributed to remodeling the rhythm of hippocampal neurogenesis and mitigated behavioral dysfunction in OD-fed mice. Conclusions: The present study addresses a gap in the understanding of neuronal dysfunction associated with OD during adolescence and provides the first evidence that EPs improved cognitive and social behavior via modulation of gut microbiota and tryptophan metabolism in adolescent mice fed with OD, which may represent novel preemptive therapy for neurodevelopmental disorders via manipulation of the tryptophan metabolite.


Subject(s)
Gastrointestinal Microbiome , Animals , Cognition , Diet , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Glutamic Acid , Kynurenine/pharmacology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S , Tryptophan
20.
Nutrients ; 14(12)2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35745182

ABSTRACT

Celiac disease (CD) is an autoimmune disease with the destruction of small intestinal villi, which occurs in genetically predisposed individuals. At the present moment, a gluten-free diet (GFD) is the only way to restore the functionality of gut mucosa. However, there is an open debate on the effects of long-term supplementation through a GFD, because some authors report an unbalance in microbial taxa composition. METHODS: For microbiome analysis, fecal specimens were collected from 46 CD individuals in GFD for at least 2 years and 30 specimens from the healthy controls (HC). Data were analyzed using an ensemble of software packages: QIIME2, Coda-lasso, Clr-lasso, Selbal, PICRUSt2, ALDEx2, dissimilarity-overlap analysis, and dysbiosis detection tests. RESULTS: The adherence to GFD restored the alpha biodiversity of the gut microbiota in celiac people but microbial composition at beta diversity resulted as different to HC. The microbial composition of the CD subjects was decreased in a number of taxa, namely Bifidobacterium longum and several belonging to Lachnospiraceae family, whereas Bacteroides genus was found to be more abundant. Predicted metabolic pathways among the CD bacterial communities revealed an important role in tetrapyrrole biosynthesis. CONCLUSIONS: CD patients in GFD had a non-dysbiotic microbial composition for the crude alpha diversity metrics. We found significant differences in beta diversity, in certain taxon, and pathways between subjects with inactive CD in GFD and controls. Collectively, our data may suggest the development of new GFD products by modulating the gut microbiota through diet, supplements of vitamins, and the addition of specific prebiotics.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Microbiota , Diet, Gluten-Free , Dysbiosis/microbiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL