Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 17(1): 131-148, 2024.
Article in English | MEDLINE | ID: mdl-37739064

ABSTRACT

BACKGROUND & AIMS: Altered plasma acylcarnitine levels are well-known biomarkers for a variety of mitochondrial fatty acid oxidation disorders and can be used as an alternative energy source for the intestinal epithelium when short-chain fatty acids are low. These membrane-permeable fatty acid intermediates are excreted into the gut lumen via bile and are increased in the feces of patients with inflammatory bowel disease (IBD). METHODS: Herein, based on studies in human subjects, animal models, and bacterial cultures, we show a strong positive correlation between fecal carnitine and acylcarnitines and the abundance of Enterobacteriaceae in IBD where they can be consumed by bacteria both in vitro and in vivo. RESULTS: Carnitine metabolism promotes the growth of Escherichia coli via anaerobic respiration dependent on the cai operon, and acetylcarnitine dietary supplementation increases fecal carnitine levels with enhanced intestinal colonization of the enteric pathogen Citrobacter rodentium. CONCLUSIONS: In total, these results indicate that the increased luminal concentrations of carnitine and acylcarnitines in patients with IBD may promote the expansion of pathobionts belonging to the Enterobacteriaceae family, thereby contributing to disease pathogenesis.


Subject(s)
Enterobacteriaceae , Inflammatory Bowel Diseases , Animals , Humans , Enterobacteriaceae/metabolism , Dysbiosis , Inflammatory Bowel Diseases/microbiology , Carnitine/metabolism , Fatty Acids/metabolism , Escherichia coli , Biomarkers
2.
Curr Microbiol ; 80(11): 363, 2023 Oct 08.
Article in English | MEDLINE | ID: mdl-37807005

ABSTRACT

Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) have been associated with several inflammatory conditions, including inflammatory bowel diseases (IBDs), and found to have an impact on gut microbiota. In fact, some randomized controlled studies suggest benefits to IBD patients, but others do not. Our aim was to review recent evidence on the effects of omega-3 on IBD and establish the contribution of the gut microbiome. Omega-3 mediate anti-inflammatory effects in IBD through various mechanisms, including suppression of NLR family pyrin domain-containing 3 (NLRP3) inflammasome, Toll-like receptor-4 (TLR4), and nucleotide-binding oligomerization domain 2 (NOD2) signaling; this results in the repression of the nuclear factor-kappa B (Nf-kB) pathway and the secretion of pro-inflammatory cytokines. Omega-3 can also affect gut microbiota and revert the bacterial community to patterns associated with healthy status by increasing short-chain fatty acid (SCFA)-producing bacteria and enhancing the mucosal gut barrier, thus promoting homeostasis. The combination of these immunoregulatory effects and anti-inflammation properties with the promotion of a balanced gut microbiome environment could suggest that omega-3 might benefit IBD patients. Considering the microbiota of IBD patients while using omega-3 might predict and improve omega-3 effectiveness. Combining omega-3 with bacteria-altering therapy, such as probiotics and fecal microbiota transplantation, may further enhance its efficacy; however, further studies are required to elucidate mechanisms and potential preventive or treatment roles of omega-3 in IBD.


Subject(s)
Fatty Acids, Omega-3 , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/microbiology , Bacteria/genetics , Fecal Microbiota Transplantation
3.
J Gastroenterol Hepatol ; 38(8): 1333-1345, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37210613

ABSTRACT

BACKGROUND AND AIM: Curcumin may have promising application in the prevention and amelioration of inflammatory bowel disease (IBD). However, the underlying mechanisms underpinning the ability of curcumin to interact with the gut and liver in IBD remains to be defined, which is the exploration aim of this study. METHODS: Mice with dextran sulfate sodium salt (DSS)-induced acute colitis were treated either with 100 mg/kg of curcumin or phosphate buffer saline (PBS). Hematoxylin-eosin (HE) staining, 16S rDNA Miseq sequencing, proton nuclear magnetic resonance (1 H NMR) spectroscopy, and liquid chromatography-tandem mass spectrometry (LC-MS/MS) were applied for analysis. Spearman's correlation coefficient (SCC) was utilized to assess the correlation between the modification of intestinal bacteria and hepatic metabolite parameters. RESULTS: Curcumin supplementation not only prevented further loss of body weight and colon length in IBD mice but also improved diseases activity index (DAI), colonic mucosal injury, and inflammatory infiltration. Meanwhile, curcumin restored the composition of the gut microbiota, significantly increased Akkermansia, Muribaculaceae_unclassified, and Muribaculum, and significantly elevated the concentration of propionate, butyrate, glycine, tryptophan, and betaine in the intestine. For hepatic metabolic disturbances, curcumin intervention altered 14 metabolites, including anthranilic acid and 8-amino-7-oxononanoate while enriching pathways related to the metabolism of bile acids, glucagon, amino acids, biotin, and butanoate. Furthermore, SCC analysis revealed a potential correlation between the upregulation of intestinal probiotics and alterations in liver metabolites. CONCLUSION: The therapeutic mechanism of curcumin against IBD mice occurs by improving intestinal dysbiosis and liver metabolism disorders, thus contributing to the stabilization of the gut-liver axis.


Subject(s)
Colitis , Curcumin , Inflammatory Bowel Diseases , Liver Diseases , Animals , Mice , Curcumin/pharmacology , Curcumin/therapeutic use , Dextran Sulfate , Dysbiosis/drug therapy , Chromatography, Liquid , Tandem Mass Spectrometry , Colitis/chemically induced , Colitis/drug therapy , Colitis/prevention & control , Inflammatory Bowel Diseases/microbiology , Colon/pathology , Mice, Inbred C57BL , Disease Models, Animal
4.
Gut Microbes ; 15(1): 2211501, 2023.
Article in English | MEDLINE | ID: mdl-37203220

ABSTRACT

Magnitude and diversity of gut microbiota and metabolic systems are critical in shaping human health and diseases, but it remains largely unclear how complex metabolites may selectively regulate gut microbiota and determine health and diseases. Here, we show that failures or compromised effects of anti-TNF-α therapy in inflammatory bowel diseases (IBD) patients were correlated with intestinal dysbacteriosis with more pro-inflammatory bacteria, extensive unresolved inflammation, failed mucosal repairment, and aberrant lipid metabolism, particularly lower levels of palmitoleic acid (POA). Dietary POA repaired gut mucosal barriers, reduced inflammatory cell infiltrations and expressions of TNF-α and IL-6, and improved efficacy of anti-TNF-α therapy in both acute and chronic IBD mouse models. Ex vivo treatment with POA in cultured inflamed colon tissues derived from Crohn's disease (CD) patients reduced pro-inflammatory signaling/cytokines and conferred appreciable tissue repairment. Mechanistically, POA significantly upregulated the transcriptional signatures of cell division and biosynthetic process of Akkermansia muciniphila, selectively increased the growth and abundance of Akkermansia muciniphila in gut microbiota, and further reprogrammed the composition and structures of gut microbiota. Oral transfer of such POA-reprogrammed, but not control, gut microbiota induced better protection against colitis in anti-TNF-α mAb-treated recipient mice, and co-administration of POA with Akkermansia muciniphila showed significant synergistic protections against colitis in mice. Collectively, this work not only reveals the critical importance of POA as a polyfunctional molecular force to shape the magnitude and diversity of gut microbiota and therefore promote the intestinal homeostasis, but also implicates a new potential therapeutic strategy against intestinal or abenteric inflammatory diseases.


Subject(s)
Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Animals , Mice , Tumor Necrosis Factor Inhibitors/metabolism , Colitis/microbiology , Inflammatory Bowel Diseases/microbiology , Verrucomicrobia/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Biological Therapy , Dextran Sulfate , Mice, Inbred C57BL , Disease Models, Animal
5.
J Nutr Biochem ; 101: 108926, 2022 03.
Article in English | MEDLINE | ID: mdl-34848335

ABSTRACT

Short-chain fatty acids (SCFAs) play a pivotal role in maintaining intestinal homeostasis. We aimed to investigate the effects of SCFA supplementation on gut inflammation and microbiota composition in a murine colitis model. Mice were fed with sodium butyrate or a mixture of SCFAs in the drinking water for 2 weeks, followed by 2% dextran sulfate sodium (DSS) for 7 d. After euthanasia, mouse colons were extracted to examine histological findings. Flow cytometry of the mouse colon tissues was performed to assess T cell differentiation. Changes in gut microbiota were assessed by high-throughput sequencing of the mouse feces. There were no significant differences in weight change, colonic length, or histologic inflammation score between the DSS, butyrate, and SCFA mix groups. However, flow cytometry revealed that both the expression of CD4+Foxp3+ regulatory T cells and of IL-17-producing T cells were increased in the butyrate and SCFA mix groups. Microbial compositions of the butyrate and SCFA mix groups were significantly different from those of the control and DSS groups in principal coordinate analysis. Relative abundances of the phyla Verrucomicrobia and Proteobacteria, species Akkermansia muciniphila and Escherichia fergusonii were increased in the butyrate and SCFA mix groups. Genera Roseburia and Lactobacillus showed a negative correlation with the degree of colitis, whereas genera Escherichia and Mucispirillum showed a positive correlation. SCFA supplementation did not result in a significant reduction in colon inflammation, but it promoted both regulatory T cell and IL-17-producing T cell expression, and increased both protective and aggressive gut microbiota.


Subject(s)
Butyrates/administration & dosage , Dietary Supplements , Fatty Acids, Volatile/administration & dosage , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Animals , Cell Differentiation , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colon/pathology , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Inflammatory Bowel Diseases/immunology , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
6.
Cell Host Microbe ; 30(1): 17-30.e9, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34822777

ABSTRACT

Bacterial activation of T helper 17 (Th17) cells exacerbates mouse models of autoimmunity, but how human-associated bacteria impact Th17-driven disease remains elusive. We show that human gut Actinobacterium Eggerthella lenta induces intestinal Th17 activation by lifting inhibition of the Th17 transcription factor Rorγt through cell- and antigen-independent mechanisms. E. lenta is enriched in inflammatory bowel disease (IBD) patients and worsens colitis in a Rorc-dependent manner in mice. Th17 activation varies across E. lenta strains, which is attributable to the cardiac glycoside reductase 2 (Cgr2) enzyme. Cgr2 is sufficient to induce interleukin (IL)-17a, a major Th17 cytokine. cgr2+ E. lenta deplete putative steroidal glycosides in pure culture; related compounds are negatively associated with human IBD severity. Finally, leveraging the sensitivity of Cgr2 to dietary arginine, we prevented E. lenta-induced intestinal inflammation in mice. Together, these results support a role for human gut bacterial metabolism in driving Th17-dependent autoimmunity.


Subject(s)
Colitis/metabolism , Gastrointestinal Microbiome/physiology , Lymphocyte Activation/physiology , Th17 Cells/metabolism , Actinobacteria , Animals , Bacteria/metabolism , Colitis/immunology , Cytokines , Dietary Supplements , Disease Models, Animal , Female , Humans , Inflammatory Bowel Diseases/microbiology , Interleukin-17/metabolism , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
7.
Food Funct ; 13(2): 514-529, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-34935814

ABSTRACT

Intestinal inflammation represented by inflammatory bowel disease (IBD) has become a global epidemic disease and the number of patients with IBD continues to increase. This digestive tract disease not only affects the absorption of food components by destroying the intestinal epithelial structure, but also can induce diseases in remote organs via the gut-organ axis, seriously harming human health. Nowadays, increasing attention is being paid to the nutritional and medicinal value of food components with increasing awareness among the general public regarding health. As an important member of the isothiocyanates, sulforaphane (SFN) is abundant in cruciferous plants and is famous for its excellent anti-cancer effects. With the development of clinical research, more physiological activities of SFN, such as antidepressant, hypoglycemic and anti-inflammatory activities, have been discovered, supporting the fact that SFN and SFN-rich sources have great potential to be dietary supplements that are beneficial to health. This review summarizes the characteristics of intestinal inflammation, the anti-inflammatory mechanism of SFN and its various protective effects on intestinal inflammation, and the possible future applications of SFN for promoting intestinal health have also been discussed.


Subject(s)
Anti-Inflammatory Agents , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Isothiocyanates , Sulfoxides , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Brassicaceae , Colitis/metabolism , Colitis/microbiology , Colitis/physiopathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/physiopathology , Intestines/drug effects , Intestines/physiology , Isothiocyanates/chemistry , Isothiocyanates/metabolism , Isothiocyanates/pharmacology , Mice , Sulfoxides/chemistry , Sulfoxides/metabolism , Sulfoxides/pharmacology , Vegetables
8.
United European Gastroenterol J ; 9(9): 1019-1026, 2021 11.
Article in English | MEDLINE | ID: mdl-34431607

ABSTRACT

AIM: Many therapeutic options for inflammatory bowel disease (IBD) emerged during the last 2 decades, along with the rise in disease prevalence and incidence. We aimed at assessing the published literature on different treatment options in that period. Special attention was attributed to specific medication mechanisms and geographic diversity. MATERIALS AND METHODS: We have queried PubMed for all available IBD-related entries published during 2000-2020. The following data were extracted for each entry: PubMed unique article ID (PMID), title, publishing journal, abstract text, keywords (if any), and authors' affiliations. Two gastrointestinal specialists decided in consensus on a list of terms to classify entries. The terms belonged to five treatment groups: medical, surgical, dietary, microbiome manipulation, and complementary medicine. The medical and complementary medicine groups were further sub-classified. Annual trends of publications for the years 2000-2020 were plotted for different treatment types. The slopes of publication trends were calculated by fitting regression lines to the annual number of publications. RESULTS: Overall, 77,505 IBD entries were published between 2000 and 2020. Medical treatment showed the highest number of total publications 21,540/77,505 (27.8%), followed by surgical 7605/77,505 (9.8%), microbiome research 5260/77,505 (6.8%), dietary 4819/77,505 (6.2%), and complementary medicine treatment 762/77,505 (1.0%). Interestingly, since 2012 there is a steep rise in microbiome publications that outnumbered surgery in the last 2 years. Trend analysis of medical treatment showed that biologics had the steepest slope (57.5, p < 0.001), followed by immunomodulators (4.9, p < 0.001), small molecules (3.9, p < 0.001), and 5-ASA (3.8, p < 0.001). CONCLUSION: According to our high-level publications trend analysis, the past 2 decades certainly deserve the reference as the "biologic era", as publications regarding biological therapy outnumbered all other treatment options. Interestingly, though very popular among patients, complementary medicine was not studied with correlation to its' acceptance among patients.


Subject(s)
Data Mining/methods , Inflammatory Bowel Diseases/therapy , PubMed , Biological Products/therapeutic use , Complementary Therapies , Diet , Fecal Microbiota Transplantation , Humans , Incidence , Inflammatory Bowel Diseases/epidemiology , Inflammatory Bowel Diseases/microbiology , Microbiota , Prevalence , Probiotics/therapeutic use
9.
Cell Host Microbe ; 29(8): 1294-1304.e4, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34297922

ABSTRACT

The intestinal microbiome is a key determinant of responses to biologic therapy in inflammatory bowel disease (IBD). However, diverse therapeutics and variable responses among IBD patients have posed challenges in predicting clinical therapeutic success. In this prospective study, we profiled baseline stool and blood in patients with moderate-to-severe Crohn's disease or ulcerative colitis initiating anti-cytokine therapy (anti-TNF or -IL12/23) or anti-integrin therapy. Patients were assessed at 14 weeks for clinical remission and 52 weeks for clinical and endoscopic remission. Baseline microbial richness indicated preferential responses to anti-cytokine therapy and correlated with the abundance of microbial species capable of 7α/ß-dehydroxylation of primary to secondary bile acids. Serum signatures of immune proteins reflecting microbial diversity identified patients more likely to achieve remission with anti-cytokine therapy. Remission-associated multi-omic profiles were unique to each therapeutic class. These profiles may facilitate a priori determination of optimal therapeutics for patients and serve as targets for newer therapies.


Subject(s)
Biological Therapy , Colitis, Ulcerative/therapy , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases , Antibodies, Monoclonal, Humanized , Biomarkers , Blood , Crohn Disease/therapy , Cytokines/blood , Cytokines/drug effects , Feces , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Infliximab , Metabolomics , Metagenome , Prospective Studies , Proteomics , Tumor Necrosis Factor Inhibitors/therapeutic use
10.
J Immunol Res ; 2021: 9117805, 2021.
Article in English | MEDLINE | ID: mdl-34195297

ABSTRACT

Although the strain-dependent effects of Bacteroides vulgatus on alleviating intestinal inflammatory diseases have been demonstrated, the literature has rarely focused on the underlying causes of this effect. In this study, we selected four B. vulgatus strains (FTJS5K1, FTJS7K1, FSDTA11B14, and FSDLZ51K1) with different genomic characteristics and evaluated their protective roles against dextran sulfate sodium- (DSS-) induced colitis. Compared to the other three tested strains, B. vulgatus 7K1 more strongly ameliorated the DSS-induced weight loss, shortening of the colon length, increased disease activity index scores, colonic tissue injury, and immunomodulatory disorder. In contrast, B. vulgatus 51K1 significantly worsened the DSS-induced alterations in the tumor necrosis factor-alpha (TNF-α) concentration and colonic histopathology. A comparative genomic analysis of B. vulgatus 7K1 and 51K1 showed that the beneficial effects of B. vulgatus 7K1 may be associated with some of its specific genes involved in the production of short-chain fatty acids or capsular polysaccharides and enhancement of its survivability in the gut. In conclusion, these findings indicate that the supplementation of B. vulgatus 7K1 is a potentially efficacious intervention for alleviating colitis and provides scientific support for the screening of probiotics with anticolitis effect.


Subject(s)
Bacteroides/genetics , Colitis/microbiology , Colon/pathology , Inflammatory Bowel Diseases/microbiology , Animals , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate , Disease Models, Animal , Fatty Acids/metabolism , Humans , Inflammatory Bowel Diseases/immunology , Male , Mice , Mice, Inbred C57BL , Polysaccharides/metabolism , Species Specificity , Tumor Necrosis Factor-alpha/metabolism
11.
Mol Nutr Food Res ; 65(15): e2001065, 2021 08.
Article in English | MEDLINE | ID: mdl-34075695

ABSTRACT

SCOPE: Inflammatory bowel disease (IBD) is an inflammatory gastrointestinal disorder in which endoplasmic reticulum (ER) stress and dysbiosis of the intestinal microbiota are implicated. Glycine supplementation is reported to reduce inflammatory responses in experimental colitis. However, the underlying mechanisms responsible for the beneficial effects remain unclear. METHODS AND RESULTS: Female C57BL/6 mice are orally administered with glycine (3.5 or 5.2 g kg-1 body weight) for 14 continuous days. On day 8 post-glycine supplementation, the mice are orally inoculated with 2 × 109 CFU Citrobacter rodentium (C. rodentium). The results show that glycine alleviates C. rodentium-induced body weight loss, increased disease activity index and spleen weight, colon length shortening, and colonic hyperplasia. Glycine suppresses the activation and infiltration of inflammatory cells, and secretion of pro-inflammatory cytokines in the colon tissues. The apoptosis of colon epithelial cells is also abrogated by glycine, which is associated with the inactivation of activating transcription factor 6α (ATF6α)-C/EBP homologous protein (CHOP) signaling. In addition, glycine administration increases α diversity, restores ß diversity, and abolishes the reduction in Lactobacillus, Bifidobacterium, Alistipes, Turicibacter, and Alloprevotella in the colon. CONCLUSIONS: Glycine supplementation is a nutritional strategy that may ameliorate C. rodentium-induced colitis by regulating ATF6α-CHOP-mediated ER stress and enhancing the abundance of Lactobacillus.


Subject(s)
Activating Transcription Factor 6/metabolism , Colitis/drug therapy , Endoplasmic Reticulum Stress/drug effects , Gastrointestinal Microbiome/drug effects , Glycine/pharmacology , Animals , Antimicrobial Peptides/genetics , Cell Death/drug effects , Citrobacter rodentium/pathogenicity , Colitis/metabolism , Colitis/microbiology , Colon/drug effects , Colon/microbiology , Colon/pathology , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Inflammatory Bowel Diseases/microbiology , Mice, Inbred C57BL
12.
Sci Rep ; 11(1): 571, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436961

ABSTRACT

Damage to the small intestine caused by non-steroidal anti-inflammatory drugs (NSAIDs) occurs more frequently than in the upper gastrointestinal tract, is more difficult to diagnose and no effective treatments exist. Hence, we investigated whether probiotics can control the onset of this severe condition in a murine model of intestinal inflammation induced by the NSAID, indomethacin. Probiotic supplementation to mice reduce the body weight loss, anemia, shortening of the small intestine, cell infiltration into the intestinal tissue and the loss of Paneth and Goblet cells associated with intestinal inflammation. Furthermore, a high antimicrobial activity in the intestinal fluids of mice fed with probiotics compared to animals on a conventional diet was elicited against several pathogens. Interestingly, probiotics dampened the oxidative stress and several local and systemic markers of an inflammatory process, as well as increased the secretion of IL-10 by regulatory T cells. Even more importantly, probiotics induced important changes in the large intestine microbiota characterized by an increase in anaerobes and lactobacilli, and a significant decrease in total enterobacteria. We conclude that oral probiotic supplementation in NSAID-induced inflammation increases intestinal antimicrobial activity and reinforces the intestinal epithelial barrier in order to avoid pathogens and commensal invasion and maintain intestinal homeostasis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Dietary Supplements , Gastrointestinal Microbiome , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/microbiology , Lactobacillus , Probiotics/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Disease Models, Animal , Goblet Cells/pathology , Indomethacin/administration & dosage , Indomethacin/adverse effects , Inflammation , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/prevention & control , Interleukin-10/metabolism , Intestines/cytology , Intestines/pathology , Mice, Inbred BALB C , Oxidative Stress , T-Lymphocytes, Regulatory/metabolism
13.
Front Immunol ; 12: 791565, 2021.
Article in English | MEDLINE | ID: mdl-35069573

ABSTRACT

Intestinal diseases, such as inflammatory bowel diseases (IBDs) and colorectal cancer (CRC) generally characterized by clinical symptoms, including malabsorption, intestinal dysfunction, injury, and microbiome imbalance, as well as certain secondary intestinal disease complications, continue to be serious public health problems worldwide. The role of vitamin K (VK) on intestinal health has drawn growing interest in recent years. In addition to its role in blood coagulation and bone health, several investigations continue to explore the role of VK as an emerging novel biological compound with the potential function of improving intestinal health. This study aims to present a thorough review on the bacterial sources, intestinal absorption, uptake of VK, and VK deficiency in patients with intestinal diseases, with emphasis on the effect of VK supplementation on immunity, anti-inflammation, intestinal microbes and its metabolites, antioxidation, and coagulation, and promoting epithelial development. Besides, VK-dependent proteins (VKDPs) are another crucial mechanism for VK to exert a gastroprotection role for their functions of anti-inflammation, immunomodulation, and anti-tumorigenesis. In summary, published studies preliminarily show that VK presents a beneficial effect on intestinal health and may be used as a therapeutic drug to prevent/treat intestinal diseases, but the specific mechanism of VK in intestinal health has yet to be elucidated.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome/immunology , Inflammatory Bowel Diseases , Intestinal Mucosa , Vitamin K Deficiency , Vitamin K , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/therapy , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Vitamin K/immunology , Vitamin K/therapeutic use , Vitamin K Deficiency/immunology , Vitamin K Deficiency/microbiology
14.
Int J Mol Sci ; 23(1)2021 Dec 24.
Article in English | MEDLINE | ID: mdl-35008631

ABSTRACT

Gastrointestinal health is influenced by the functional genes and metabolites generated by the human microbiome. As the volume of current biomedical and translational research indicates, the importance and impact of this ecosystem of microorganisms, especially those comprising the gut microbiome on human health, has become increasingly apparent. Changes to the gut microbiome are associated with inflammatory bowel disease (IBD), which is characterized by persistent intestinal inflammation. Furthermore, the lifetime dietary choices of their host may positively or negatively affect both the gut microbiome and its impact on IBD. As such, "anti-inflammatory" dietary supplements, their impact, and mechanisms in restoring gut microbiota homeostasis during IBD is an area of intensive research. Dietary supplementation may represent an important adjuvant treatment avenue for limiting intestinal inflammation in IBD. Overall, this review addresses the development of the gut microbiome, the significance of the gut microbiome in IBD, and the use of dietary supplements such as vitamin D, fish oil, and resveratrol in the mitigation of IBD-associated gut dysbiosis and intestinal inflammation.


Subject(s)
Dietary Supplements , Fish Oils/therapeutic use , Gastrointestinal Microbiome , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/microbiology , Resveratrol/therapeutic use , Vitamin D/therapeutic use , Animals , Fish Oils/pharmacology , Gastrointestinal Microbiome/drug effects , Humans , Inflammatory Bowel Diseases/pathology , Resveratrol/pharmacology , Vitamin D/pharmacology
15.
Pharmacol Res ; 161: 105135, 2020 11.
Article in English | MEDLINE | ID: mdl-32814166

ABSTRACT

The intestinal epithelial layer serves as a physical and functional barrier between the microbe-rich lumen and immunologically active submucosa; it prevents systemic translocation of microbial pyrogenic products (e.g. endotoxin) that elicits immune activation upon translocation to the systemic circulation. Loss of barrier function has been associated with chronic 'low-grade' systemic inflammation which underlies pathogenesis of numerous no-communicable chronic inflammatory disease. Thus, targeting gut barrier dysfunction is an effective strategy for the prevention and/or treatment of chronic disease. This review intends to emphasize on the beneficial effects of herbal formulations, phytochemicals and traditional phytomedicines in attenuating intestinal barrier dysfunction. It also aims to provide a comprehensive understanding of intestinal-level events leading to a 'leaky-gut' and systemic complications mediated by endotoxemia. Additionally, a variety of detectable markers and diagnostic criteria utilized to evaluate barrier improving capacities of experimental therapeutics has been discussed. Collectively, this review provides rationale for targeting gut barrier dysfunction by phytotherapies for treating chronic diseases that are associated with endotoxemia-induced systemic inflammation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Endotoxemia/drug therapy , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/drug effects , Phytotherapy , Plant Extracts/therapeutic use , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/isolation & purification , Bacteria/immunology , Bacteria/metabolism , Chronic Disease , Dysbiosis , Endotoxemia/metabolism , Endotoxemia/microbiology , Endotoxemia/pathology , Endotoxins/metabolism , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/isolation & purification , Gastrointestinal Microbiome , Host-Pathogen Interactions , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Permeability , Plant Extracts/adverse effects , Plant Extracts/isolation & purification
16.
Cell Host Microbe ; 28(2): 273-284.e6, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32668218

ABSTRACT

The clinical spectra of irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD) intersect to form a scantily defined overlap syndrome, termed pre-IBD. We show that increased Enterobacteriaceae and reduced Clostridia abundance distinguish the fecal microbiota of pre-IBD patients from IBS patients. A history of antibiotics in individuals consuming a high-fat diet was associated with the greatest risk for pre-IBD. Exposing mice to these risk factors resulted in conditions resembling pre-IBD and impaired mitochondrial bioenergetics in the colonic epithelium, which triggered dysbiosis. Restoring mitochondrial bioenergetics in the colonic epithelium with 5-amino salicylic acid, a PPAR-γ (peroxisome proliferator-activated receptor gamma) agonist that stimulates mitochondrial activity, ameliorated pre-IBD symptoms. As with patients, mice with pre-IBD exhibited notable expansions of Enterobacteriaceae that exacerbated low-grade mucosal inflammation, suggesting that remediating dysbiosis can alleviate inflammation. Thus, environmental risk factors cooperate to impair epithelial mitochondrial bioenergetics, thereby triggering microbiota disruptions that exacerbate inflammation and distinguish pre-IBD from IBS.


Subject(s)
Anti-Bacterial Agents/adverse effects , Diet, High-Fat/adverse effects , Dysbiosis/pathology , Energy Metabolism/physiology , Inflammatory Bowel Diseases/microbiology , Irritable Bowel Syndrome/microbiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Dysbiosis/chemically induced , Enterobacteriaceae/growth & development , Gastrointestinal Microbiome , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Leukocyte L1 Antigen Complex/metabolism , Mesalamine/therapeutic use , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , PPAR gamma/agonists
17.
Inflamm Bowel Dis ; 26(9): 1315-1318, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32483628

ABSTRACT

The use of biological agents for the treatment of chronic inflammatory conditions such as inflammatory bowel diseases (IBD) has been on the rise.1,2 Current biological therapies include antitumor necrosis factor-α (anti-TNF-α), anti-interleukin-12/23, and anti-integrin agents. Before initiation of biological drugs, screening for Mycobacterium tuberculosis infection is required to avoid reactivation or worsening of disease after immunosuppression. It has been shown that anti-TNF-α treated patients have a 14-fold increased risk of tuberculosis (TB) infection/reactivation compared with healthy controls.3 The methods for screening for TB have evolved over time and vary from region to region.


Subject(s)
Inflammatory Bowel Diseases/microbiology , Interferon-gamma Release Tests , Latent Tuberculosis/diagnosis , Mass Screening/methods , Tuberculin Test , Adult , Biological Therapy/adverse effects , Biological Therapy/standards , Female , Gastroenterology/standards , Humans , Inflammatory Bowel Diseases/drug therapy , Latent Tuberculosis/microbiology , Male , Mass Screening/standards , Mycobacterium tuberculosis , Practice Guidelines as Topic
18.
Neurogastroenterol Motil ; 32(10): e13914, 2020 10.
Article in English | MEDLINE | ID: mdl-32476236

ABSTRACT

BACKGROUND: Butyrate has shown anti-inflammatory and regenerative properties, providing symptomatic relief when orally supplemented in patients suffering from various colonic diseases. We investigated the effect of a colonic-delivery formulation of butyrate on the fecal microbiota of patients with inflammatory bowel diseases (IBDs). METHODS: In this double-blind, placebo-controlled, pilot study, 49 IBD patients (n = 19 Crohn's disease, CD and n = 30 ulcerative colitis, UC) were randomized to oral administration of microencapsulated-sodium-butyrate (BLM) or placebo for 2 months, in addition to conventional therapy. Eighteen healthy volunteers (HVs) were recruited to provide a healthy microbiota model of the local people. Fecal microbiota from stool samples was assessed by 16S sequencing. Clinical disease activity and quality of life (QoL) were evaluated before and after treatment. KEY RESULTS: At baseline, HVs showed a different microbiota composition compared with IBD patients. Sodium-butyrate altered the gut microbiota of IBD patients by increasing bacteria able to produce SCFA in UC patients (Lachnospiraceae spp.) and the butyrogenic colonic bacteria in CD patients (Butyricicoccus). In UC patients, QoL was positively affected by treatment. CONCLUSIONS AND INFERENCES: Sodium-butyrate supplementation increases the growth of bacteria able to produce SCFA with potentially anti-inflammatory action. The clinical impact of this finding requires further investigation.


Subject(s)
Butyric Acid/administration & dosage , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/drug therapy , Administration, Oral , Adult , Aged , Capsules , Double-Blind Method , Female , Histamine Antagonists/administration & dosage , Humans , Inflammatory Bowel Diseases/microbiology , Male , Middle Aged , Pilot Projects , Young Adult
19.
Curr Drug Targets ; 21(14): 1417-1427, 2020.
Article in English | MEDLINE | ID: mdl-32364071

ABSTRACT

The pathogenesis of inflammatory bowel disease (IBD) remains unknown. However, there is growing evidence that the increase in the overall incidence of IBD relates to the improvement of sanitary and hygienic conditions of the society leading to lower exposure to both bacterial and parasitic infections. IBD is incurable and characterized by alternating periods of exacerbation and remission of symptoms. Therefore, the main goal of treatment strategies in IBD patients is the most effective maintenance of clinical and endoscopic remission, which does allow patients to function normally for a significant part of life. Taking into account the evidence from different areas, there is a strong rationale supporting the concept that bacteria are important in gut inflammation and that probiotic bacteria may modulate the host-microbe interaction in a way that is directly beneficial to IBD patients along with nutritional support. In this review, we focus on the potential role of gastrointestinal microbiota in the pathogenesis of IBD and the possible value of probiotics, prebiotics, and symbiotics as well as nutritional support in the treatment of IBD.


Subject(s)
Inflammatory Bowel Diseases/therapy , Nutrition Therapy , Probiotics/therapeutic use , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/microbiology
20.
Molecules ; 25(10)2020 May 14.
Article in English | MEDLINE | ID: mdl-32422882

ABSTRACT

Vitamin D is widely known to regulate bone health, but there is increasing evidence that it may also ameliorate colitis through inflammation, cell proliferation and apoptosis, and the microbiota. The purpose of this review is to systematically examine the mechanisms by which vitamin D reduces colitis. PubMed and Web of Science were searched for articles published between 2008 and 2019 using key words such as "vitamin D," "colitis," "inflammatory bowel disease," "inflammation," "apoptosis," "cell proliferation," and "gut bacteria". Retrieved articles were further narrowed and it was determined whether their title and abstracts contained terminology pertaining to vitamin D in relation to colitis in human clinical trials, animal studies, and cell culture/biopsy studies, as well as selecting the best match sorting option in relation to the research question. In total, 30 studies met the established criteria. Studies consistently reported results showing that vitamin D supplementation can downregulate inflammatory pathways of COX-2, TNF-α, NF-κB, and MAPK, modify cell kinetics, and alter gut microbiome, all of which contribute to an improved state of colitis. Although vitamin D and vitamin D analogs have demonstrated positive effects against colitis, more randomized, controlled human clinical trials are needed to determine the value of vitamin D as a therapeutic agent in the treatment of colitis.


Subject(s)
Colitis/diet therapy , Dietary Supplements , Gene Expression Regulation/drug effects , Inflammatory Bowel Diseases/diet therapy , Vitamin D/administration & dosage , Animals , Clinical Trials as Topic , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Colon/drug effects , Colon/microbiology , Colon/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Inflammation , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL