Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.093
Filter
1.
J Neuroimmunol ; 395: 578422, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39178494

ABSTRACT

Intestinal microbes play a crucial role in gut health and the immune-mediated central nervous system through the "gut-brain" axis. However, probiotic safety and efficacy in Neuromyelitis optica spectrum disorder (NMOSD) are not well-explored. A pilot clinic trial for NMOSD with probiotic intervention revealed alterations in the microbiota (increased Anaerostipes, Bacteroides; decreased Granulicatella, Streptococcus, Rothia). Metabolite analysis showed elevated 2-methylbutyric and isobutyric acids, reduced lithocholic acid (LCA), and glycodeoxycholic acid (GDCA). Immune markers Interleukin (IL-7), vascular endothelial growth factor (VEGF-A), and B lymphocyte chemoattractant (BLC) decreased, while plasma cells and transitional B cells increased post-probiotics, suggesting potential immunomodulatory effects on NMOSD.


Subject(s)
B-Lymphocytes , Cell Differentiation , Lithocholic Acid , Neuromyelitis Optica , Probiotics , Humans , Neuromyelitis Optica/immunology , Female , Cell Differentiation/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Middle Aged , Male , Adult , Pilot Projects , Gastrointestinal Microbiome/drug effects
2.
Sci Rep ; 14(1): 18705, 2024 08 12.
Article in English | MEDLINE | ID: mdl-39134638

ABSTRACT

The incidence of duodenal tumors (DTs) is increasing. However, the mechanisms underlying its development remain unclear. Environmental factors, including the microbiome and bile acids (BAs), are believed to influence tumor development. Therefore, we conducted a single-center, prospective, observational study to investigate the potential differences between patients with DTs and healthy controls (HCs) based on these factors. In addition, the BAs in the duodenal fluid were measured using liquid chromatography-tandem mass spectrometry. We recruited 41 patients and performed 16S rRNA-seq. There was no difference in the observed ASVs or PCoA plot of Bray-Curtis dissimilarity between the DTs and HCs. The lithocholic acid concentration was significantly lower in the DT group than in the control group. The ratio of CDCA to LCA was significantly higher in patients with DTs. No significant differences in microbiota were observed between DTs and HCs. In patients with DTs, the lithocholic acid concentration in duodenal was significantly lower than in HCs.


Subject(s)
Bile Acids and Salts , Duodenal Neoplasms , Duodenum , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Male , Bile Acids and Salts/metabolism , Female , Prospective Studies , Middle Aged , Duodenal Neoplasms/microbiology , Duodenal Neoplasms/metabolism , Duodenum/metabolism , Duodenum/microbiology , Aged , RNA, Ribosomal, 16S/genetics , Adult , Lithocholic Acid/metabolism , Microbiota , Case-Control Studies
3.
Structure ; 32(9): 1465-1476.e3, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38971159

ABSTRACT

OleP is a bacterial cytochrome P450 involved in oleandomycin biosynthesis as it catalyzes regioselective epoxidation on macrolide intermediates. OleP has recently been reported to convert lithocholic acid (LCA) into murideoxycholic acid through a highly regioselective reaction and to unspecifically hydroxylate testosterone (TES). Since LCA and TES mainly differ by the substituent group at the C17, here we used X-ray crystallography, equilibrium binding assays, and molecular dynamics simulations to investigate the molecular basis of the diverse reactivity observed with the two steroids. We found that the differences in the structure of TES and LCA affect the capability of these molecules to directly form hydrogen bonds with N-terminal residues of OleP internal helix I. The establishment of these contacts, by promoting the bending of helix I, fosters an efficient trigger of the open-to-closed structural transition that occurs upon substrate binding to OleP and contributes to the selectivity of the subsequent monooxygenation reaction.


Subject(s)
Bacterial Proteins , Cytochrome P-450 Enzyme System , Hydrogen Bonding , Molecular Dynamics Simulation , Protein Binding , Testosterone , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/chemistry , Crystallography, X-Ray , Substrate Specificity , Testosterone/metabolism , Testosterone/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Lithocholic Acid/chemistry , Lithocholic Acid/metabolism , Binding Sites , Hydroxylation
4.
Nat Commun ; 15(1): 5697, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972900

ABSTRACT

Climate and environmental changes threaten human mental health, but the impacts of specific environmental conditions on neuropsychiatric disorders remain largely unclear. Here, we show the impact of a humid heat environment on the brain and the gut microbiota using a conditioned housing male mouse model. We demonstrate that a humid heat environment can cause anxiety-like behaviour in male mice. Microbial 16 S rRNA sequencing analysis reveals that a humid heat environment caused gut microbiota dysbiosis (e.g., decreased abundance of Lactobacillus murinus), and metabolomics reveals an increase in serum levels of secondary bile acids (e.g., lithocholic acid). Moreover, increased neuroinflammation is indicated by the elevated expression of proinflammatory cytokines in the serum and cortex, activated PI3K/AKT/NF-κB signalling and a microglial response in the cortex. Strikingly, transplantation of the microbiota from mice reared in a humid heat environment readily recapitulates these abnormalities in germ-free mice, and these abnormalities are markedly reversed by Lactobacillus murinus administration. Human samples collected during the humid heat season also show a decrease in Lactobacillus murinus abundance and an increase in the serum lithocholic acid concentration. In conclusion, gut microbiota dysbiosis induced by a humid heat environment drives the progression of anxiety disorders by impairing bile acid metabolism and enhancing neuroinflammation, and probiotic administration is a potential therapeutic strategy for these disorders.


Subject(s)
Anxiety , Bile Acids and Salts , Dysbiosis , Gastrointestinal Microbiome , Hot Temperature , Animals , Male , Mice , Bile Acids and Salts/metabolism , Humans , Dysbiosis/microbiology , Anxiety/microbiology , Mice, Inbred C57BL , Humidity , Lithocholic Acid/metabolism , Lactobacillus , Brain/metabolism , NF-kappa B/metabolism , RNA, Ribosomal, 16S/genetics , Disease Models, Animal , Anxiety Disorders/metabolism , Anxiety Disorders/microbiology , Anxiety Disorders/etiology , Signal Transduction , Cytokines/metabolism
5.
Int J Pharm ; 662: 124503, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39043284

ABSTRACT

In the study, we aimed to investigate the activity of nanoformulations containing 5-fluorouracil and polymer-magnetic hybrids bearing membrane-penetrating and ligand-receptor-recognizing agents against colorectal cancer cells. The formation and characterization of iron oxide particles covered with polymeric shells comprising lithocholic acid and folic acid moieties are presented. The efficiency of nanoformulations combined by the simple mixing of low doses of 5-fluorouracil with the obtained hybrids was demonstrated against DLD-1 and HT-29 colon cancer cells. The most pronounced cytotoxic potential against HT-29 cells was observed in the cases of particles based on block and randomly arranged copolymers functionalized by FA motifs with depletion of viable cells by approximately 50 % compared to control cells and cells treated by 5-FU applied in free form. In the case of the DLD-1 cell line, the percentage of viable DLD-1 cells decreased by about 30 to 40% after treatment with the block and randomly arranged copolymer decorated by FA-moiety, when compared to 5-FU at the free form and the untreated control. The induction of apoptosis associated with PS-translocation was determined to be the main mechanism of their cytotoxic effects. Moreover, the safety profiles of the nanoformulations were established through hemolysis assay and the analysis of the viability of human colorectal fibroblasts. It was indicated that all tested nanoparticles met the compatibility requirements at the in vitro level. It should be emphasized that in many cases, there was a significant improvement in the compatibility of hybrids with the FA motif compared to non-functionalized hybrids with the addition of 5-FU. These findings suggest that the presence of FA might modulate the toxicity of chemotherapeutic agents.


Subject(s)
Apoptosis , Cell Survival , Colonic Neoplasms , Fluorouracil , Folic Acid , Lithocholic Acid , Polymers , Humans , Folic Acid/chemistry , Folic Acid/administration & dosage , Lithocholic Acid/chemistry , Fluorouracil/administration & dosage , Fluorouracil/chemistry , Colonic Neoplasms/drug therapy , Cell Survival/drug effects , HT29 Cells , Polymers/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Hemolysis/drug effects , Drug Carriers/chemistry , Magnetite Nanoparticles/chemistry
6.
Cell Host Microbe ; 32(8): 1315-1330.e5, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39043190

ABSTRACT

Bariatric surgical procedures such as sleeve gastrectomy (SG) provide effective type 2 diabetes (T2D) remission in human patients. Previous work demonstrated that gastrointestinal levels of the bacterial metabolite lithocholic acid (LCA) are decreased after SG in mice and humans. Here, we show that LCA worsens glucose tolerance and impairs whole-body metabolism. We also show that taurodeoxycholic acid (TDCA), which is the only bile acid whose concentration increases in the murine small intestine post-SG, suppresses the bacterial bile acid-inducible (bai) operon and production of LCA both in vitro and in vivo. Treatment of diet-induced obese mice with TDCA reduces LCA levels and leads to microbiome-dependent improvements in glucose handling. Moreover, TDCA abundance is decreased in small intestinal tissue from T2D patients. This work reveals that TDCA is an endogenous inhibitor of LCA production and suggests that TDCA may contribute to the glucoregulatory effects of bariatric surgery.


Subject(s)
Bariatric Surgery , Bile Acids and Salts , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Intestine, Small , Mice, Inbred C57BL , Obesity , Gastrointestinal Microbiome/drug effects , Animals , Mice , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Bile Acids and Salts/metabolism , Intestine, Small/metabolism , Intestine, Small/microbiology , Obesity/surgery , Obesity/metabolism , Obesity/microbiology , Male , Lithocholic Acid/metabolism , Glucose/metabolism
7.
Obes Surg ; 34(9): 3420-3433, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39042309

ABSTRACT

INTRODUCTION: Excess body fat elevates colorectal cancer risk. While bariatric surgery (BRS) induces significant weight loss, its effects on the fecal stream and colon biology are poorly understood. Specifically, limited data exist on the impact of bariatric surgery (BRS) on fecal secondary bile acids (BA), including lithocholic acid (LCA), a putative promotor of colorectal carcinogenesis. METHODS: This cross-sectional case-control study included 44 patients with obesity; 15 pre-BRS (controls) vs. 29 at a median of 24.1 months post-BRS. We examined the fecal concentrations of 11 BA by liquid chromatography and gene abundance of BA-metabolizing bacterial enzymes through fecal metagenomic sequencing. Differences were quantified using non-parametric tests for BA levels and linear discriminant analysis (LDA) effect size (LEfSe) for genes encoding BA-metabolizing enzymes. RESULTS: Total fecal secondary BA concentrations trended towards lower levels post- vs. pre-BRS controls (p = 0.07). Individually, fecal LCA concentrations were significantly lower post- vs. pre-BRS (8477.0 vs. 11,914.0 uM/mg, p < 0.008). Consistent with this finding, fecal bacterial genes encoding BA-metabolizing enzymes, specifically 3-betahydroxycholanate-3-dehydrogenase (EC 1.1.1.391) and 3-alpha-hydroxycholanate dehydrogenase (EC 1.1.1.52), were also lower post- vs. pre-BRS controls (LDA of - 3.32 and - 2.64, respectively, adjusted p < 0.0001). Post-BRS fecal BA concentrations showed significant inverse correlations with weight loss, a healthy diet quality, and increased physical activity. CONCLUSIONS: Concentrations of LCA, a secondary BA, and bacterial genes needed for BA metabolism are lower post-BRS. These changes can impact health and modulate the colorectal cancer cascade. Further research is warranted to examine how surgical alterations and the associated dietary changes impact bile acid metabolism.


Subject(s)
Bariatric Surgery , Bile Acids and Salts , Feces , Obesity, Morbid , Humans , Feces/microbiology , Pilot Projects , Male , Female , Cross-Sectional Studies , Case-Control Studies , Middle Aged , Bile Acids and Salts/metabolism , Adult , Obesity, Morbid/surgery , Obesity, Morbid/microbiology , Gastrointestinal Microbiome/physiology , Weight Loss , Lithocholic Acid/metabolism
8.
Mol Pharm ; 21(7): 3566-3576, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38899552

ABSTRACT

Oxidative stress is pivotal in retinal disease progression, causing dysfunction in various retinal components. An effective antioxidant, such as probucol (PB), is vital to counteract oxidative stress and emerges as a potential candidate for treating retinal degeneration. However, the challenges associated with delivering lipophilic drugs such as PB to the posterior segment of the eye, specifically targeting photoreceptor cells, necessitate innovative solutions. This study uses formulation-based spray dry encapsulation technology to develop polymer-based PB-lithocholic acid (LCA) nanoparticles and assesses their efficacy in the 661W photoreceptor-like cell line. Incorporating LCA enhances nanoparticles' biological efficacy without compromising PB stability. In vitro studies demonstrate that PB-LCA nanoparticles prevent reactive oxygen species (ROS)-induced oxidative stress by improving cellular viability through the nuclear erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. These findings propose PB-LCA nanoparticles as a promising therapeutic strategy for oxidative stress-induced retinopathies.


Subject(s)
Antioxidants , Lithocholic Acid , Nanoparticles , Oxidative Stress , Polymers , Probucol , Reactive Oxygen Species , Probucol/pharmacology , Probucol/administration & dosage , Probucol/chemistry , Oxidative Stress/drug effects , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , Lithocholic Acid/chemistry , Lithocholic Acid/pharmacology , Animals , Polymers/chemistry , Cell Line , Antioxidants/pharmacology , Antioxidants/chemistry , NF-E2-Related Factor 2/metabolism , Cell Survival/drug effects , Mice , Heme Oxygenase-1/metabolism , Humans
9.
ACS Chem Biol ; 19(7): 1440-1446, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38901034

ABSTRACT

Peptide-bile acid hybrids offer promising drug candidates due to enhanced pharmacological properties, such as improved protease resistance and oral bioavailability. However, it remains unknown whether bile acids can be incorporated into peptide chains by the ribosome to produce a peptide-bile acid hybrid macrocyclic peptide library for target-based de novo screening. In this study, we achieved the ribosomal incorporation of lithocholic acid (LCA)-d-tyrosine into peptide chains. This led to the construction of a peptide-LCA hybrid macrocyclic peptide library, which enabled the identification of peptides TP-2C-4L3 (targeting Trop2) and EP-2C-4L5 (targeting EphA2) with strong binding affinities. Notably, LCA was found to directly participate in binding to EphA2 and confer on the peptides improved stability and resistance to proteases. Cell staining experiments confirmed the high specificity of the peptides for targeting Trop2 and EphA2. This study highlights the benefits of LCA in peptides and paves the way for de novo discovery of stable peptide-LCA hybrid drugs.


Subject(s)
Lithocholic Acid , Peptide Library , Peptides , Ribosomes , Lithocholic Acid/chemistry , Lithocholic Acid/analogs & derivatives , Lithocholic Acid/metabolism , Ribosomes/metabolism , Humans , Peptides/chemistry , Peptides/metabolism , Receptor, EphA2/metabolism , Receptor, EphA2/chemistry , Drug Discovery , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism
10.
Nanoscale ; 16(21): 10350-10365, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38739006

ABSTRACT

Tumour cells secrete various proangiogenic factors like VEGF, PDGF, and EGF that result in the formation of highly vascularized tumours with an immunosuppressive tumour microenvironment. As tumour growth and metastasis are highly dependent on angiogenesis, targeting tumour vasculature along with rapidly dividing tumour cells is a potential approach for cancer treatment. Here, we specifically engineered sub-100 sized nanomicelles (DTX-CA4 NMs) targeting proliferation and angiogenesis using an esterase-sensitive phosphocholine-tethered docetaxel conjugate of lithocholic acid (LCA) (PC-LCA-DTX) and a poly(ethylene glycol) (PEG) derivative of an LCA-combretastatin A4 conjugate (PEG-LCA-CA4). DTX-CA4 NMs effectively inhibit the tumour growth in syngeneic (CT26) and xenograft (HCT116) colorectal cancer models, inhibit tumour recurrence, and enhance the percentage survival in comparison with individual drug-loaded NMs. DTX-CA4 NMs enhance the T cell-mediated anti-tumour immune response and DTX-CA4 NMs in combination with an immune checkpoint inhibitor, anti-PDL1 antibody, enhance the anti-tumour response. We additionally showed that DTX-CA4 NMs effectively attenuate the production of ceramide-1-phosphate, a key metabolite of the sphingolipid pathway, by downregulating the expression of ceramide kinase at both transcriptional and translational levels. Therefore, this study presents the engineering of effective DTX-CA4 NMs for targeting the tumour microenvironment that can be explored further for clinical applications.


Subject(s)
Cell Proliferation , Ceramides , Docetaxel , Micelles , Neovascularization, Pathologic , Animals , Ceramides/chemistry , Ceramides/pharmacology , Humans , Mice , Cell Proliferation/drug effects , Docetaxel/pharmacology , Docetaxel/chemistry , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Lithocholic Acid/chemistry , Lithocholic Acid/pharmacology , Polyethylene Glycols/chemistry , Cell Line, Tumor , Mice, Inbred BALB C , Stilbenes/chemistry , Stilbenes/pharmacology , HCT116 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Female , Angiogenesis
11.
Biosens Bioelectron ; 259: 116383, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38749286

ABSTRACT

Lithocholic acid (LCA), a secondary bile acid, has emerged as a potential early diagnostic biomarker for various liver diseases. In this study, we introduce a novel near-infrared (NIR) polymethine dye-based biosensor, capable of sensitive and selective detection of LCA in phosphate buffer and artificial urine (AU) solutions. The detection mechanism relies on the formation of J-aggregates resulting from the interplay of 3,3-Diethylthiatricarbocyanine iodide (DiSC2(7)) dye molecules and LCA, which induces a distinctive red shift in both absorption and fluorescence spectra. The biosensor demonstrates a detection limit for LCA of 70 µM in PBS solution (pH 7.4), while in AU solution, it responds to an LCA concentration as low as ∼60 µM. Notably, the proposed biosensor exhibits outstanding selectivity for LCA, effectively distinguishing it from common interferents such as uric acid, ascorbic acid, and glucose. This rapid, straightforward, and cost-effective spectrometer-based method underscores its potential for early diagnosis of liver diseases by monitoring LCA concentrations.


Subject(s)
Biosensing Techniques , Limit of Detection , Lithocholic Acid , Biosensing Techniques/methods , Lithocholic Acid/chemistry , Lithocholic Acid/analysis , Humans , Spectrometry, Fluorescence/methods , Fluorescent Dyes/chemistry , Carbocyanines/chemistry
12.
Mol Nutr Food Res ; 68(11): e2300910, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38794856

ABSTRACT

Gut epithelial barrier disruption is commonly observed in Western diseases like diabetes and inflammatory bowel disease (IBD). Enhanced epithelial permeability triggers inflammatory responses and gut microbiota dysbiosis. Reduced bacterial diversity in IBD affects gut microbiota metabolism, altering microbial products such as secondary bile acids (BAs), which potentially play a role in gut barrier regulation and immunity. Dietary fibers such as pectin may substitute effects of these BAs. The study examines transepithelial electrical resistance of gut epithelial T84 cells and the gene expression of tight junctions after exposure to (un)sulfated secondary BAs. This is compared to the impact of the dietary fiber pectin with different degrees of methylation (DM) and blockiness (DB), with disruption induced by calcium ionophore A23187 under both normal and hyperglycemic conditions. Unsulfated lithocholic acid (LCA) and deoxycholic acid (DCA) show a stronger rescuing effect, particularly evident under 20 mM glucose levels. DM19 with high DB (HB) and DM43HB pectin exhibit rescuing effects under both glucose conditions. Notably, DM19HB and DM43HB display higher rescue effects under 20 mM glucose compared to 5 mM glucose. The study demonstrates that specific pectins such as DM19HB and DM43HB may serve as alternatives for preventing barrier disruption in the case of disturbed DCA metabolism.


Subject(s)
Bile Acids and Salts , Hyperglycemia , Pectins , Pectins/pharmacology , Humans , Bile Acids and Salts/metabolism , Deoxycholic Acid/pharmacology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Cell Line , Tight Junctions/drug effects , Tight Junctions/metabolism , Lithocholic Acid/pharmacology , Dietary Fiber/pharmacology , Glucose/metabolism , Gastrointestinal Microbiome/drug effects , Permeability/drug effects
13.
Commun Biol ; 7(1): 465, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38632312

ABSTRACT

High temperature and humidity in the environment are known to be associated with discomfort and disease, yet the underlying mechanisms remain unclear. We observed a decrease in plasma glucagon-like peptide-1 levels in response to high-temperature and humidity conditions. Through 16S rRNA gene sequencing, alterations in the gut microbiota composition were identified following exposure to high temperature and humidity conditions. Notably, changes in the gut microbiota have been implicated in bile acid synthesis. Further analysis revealed a decrease in lithocholic acid levels in high-temperature and humidity conditions. Subsequent in vitro experiments demonstrated that lithocholic acid increases glucagon-like peptide-1 secretion in NCI-H716 cells. Proteomic analysis indicated upregulation of farnesoid X receptor expression in the ileum. In vitro experiments revealed that the combination of lithocholic acid with farnesoid X receptor inhibitors resulted in a significant increase in GLP-1 levels compared to lithocholic acid alone. In this study, we elucidate the mechanism by which reduced lithocholic acid suppresses glucagon-like peptide 1 via farnesoid X receptor activation under high-temperature and humidity condition.


Subject(s)
Gastrointestinal Microbiome , Glucagon-Like Peptide 1 , Animals , Mice , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humidity , Proteomics , RNA, Ribosomal, 16S , Temperature , Transcription Factors , Bile Acids and Salts , Lithocholic Acid
14.
Bioorg Med Chem Lett ; 105: 129760, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38641151

ABSTRACT

The naturally occurring bile acid lithocholic acid (LCA) has been a crucial core structure for many non-sugar-containing sialyltranferase (ST) inhibitors documented in literature. With the aim of elucidating the impact of the terminal carboxyl acid substituent of LCA on its ST inhibition, in this present study, we report the (bio)isosteric replacement-based design and synthesis of sulfonate and sulfate analogues of LCA. Among these compounds, the sulfate analogue SPP-002 was found to selectively inhibit N-glycan sialylation by at least an order of magnitude, indicating a substantial improvement in both potency and selectivity when compared to the unmodified parent bile acid. Molecular docking analysis supported the stronger binding of the synthetic analogue in the enzyme active site. Treatment with SPP-002 also hampered the migration, adhesion, and invasion of MDA-MB-231 cells in vitro by suppressing the expression of signaling proteins involved in the cancer metastasis-associated integrin/FAK/paxillin pathway. In totality, these findings offer not only a novel structural scaffold but also valuable insights for the future development of more potent and selective ST inhibitors with potential therapeutic effects against tumor cancer metastasis.


Subject(s)
Lithocholic Acid , Molecular Docking Simulation , Sialyltransferases , Lithocholic Acid/pharmacology , Lithocholic Acid/chemistry , Lithocholic Acid/chemical synthesis , Lithocholic Acid/analogs & derivatives , Humans , Sialyltransferases/antagonists & inhibitors , Sialyltransferases/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Structure-Activity Relationship , Sulfates/chemistry , Sulfates/pharmacology , Sulfates/chemical synthesis , Neoplasm Metastasis , Sulfonic Acids/pharmacology , Sulfonic Acids/chemistry , Sulfonic Acids/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Molecular Structure , Cell Adhesion/drug effects , Dose-Response Relationship, Drug , Paxillin/metabolism , Paxillin/antagonists & inhibitors , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Drug Discovery
15.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673867

ABSTRACT

Sialyltransferase-catalyzed membrane protein and lipid glycosylation plays a vital role as one of the most abundant post-translational modifications and diversification reactions in eukaryotes. However, aberrant sialylation has been associated with cancer malignancy and metastasis. Sialyltransferases thus represent emerging targets for the development of small molecule cancer drugs. Herein, we report the inhibitory effects of a recently discovered lithocholic acid derivative FCW393 on sialyltransferase catalytic activity, integrin sialyation, cancer-associated signal transduction, MDA-MB-231 and B16F10 cell migration and invasion, and in in vivo studies, on tumor growth, metastasis, and angiogenesis. FCW393 showed effective and selective inhibition of the sialyltransferases ST6GAL1 (IC50 = 7.8 µM) and ST3GAL3 (IC50 = 9.45 µM) relative to ST3GAL1 (IC50 > 400 µM) and ST8SIA4 (IC50 > 100 µM). FCW393 reduced integrin sialylation in breast cancer and melanoma cells dose-dependently and downregulated proteins associated with the integrin-regulated FAK/paxillin and GEF/Rho/ROCK pathways, and with the VEGF-regulated Akt/NFκB/HIF-1α pathway. FCW393 inhibited cell migration (IC50 = 2.6 µM) and invasion in in vitro experiments, and in in vivo studies of tumor-bearing mice, FCW393 reduced tumor size, angiogenesis, and metastatic potential. Based on its demonstrated selectivity, cell permeability, relatively low cytotoxicity (IC50 = 55 µM), and high efficacy, FCW393 shows promising potential as a small molecule experimental tool compound and a lead for further development of a novel cancer therapeutic.


Subject(s)
Cell Movement , Sialyltransferases , Sialyltransferases/metabolism , Sialyltransferases/antagonists & inhibitors , Humans , Animals , Mice , Cell Line, Tumor , Cell Movement/drug effects , Neoplasm Metastasis , Female , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Signal Transduction/drug effects , Cell Proliferation/drug effects , Lithocholic Acid/pharmacology
16.
Am J Transplant ; 24(7): 1132-1145, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38452932

ABSTRACT

Mycophenolate mofetil (MMF) is one of the most used immunosuppressive drugs in organ transplantation, but frequent gastrointestinal (GI) side effects through unknown mechanisms limit its clinical use. Gut microbiota and its metabolites were recently reported to play a vital role in MMF-induced GI toxicity, but the specific mechanism of how they interact with the human body is still unclear. Here, we found that secondary bile acids (BAs), as bacterial metabolites, were significantly reduced by MMF administration in the gut of mice. Microbiome data and fecal microbiota transfer model supported a microbiota-dependent effect on the reduction of secondary BAs. Supplementation of the secondary BA lithocholic acid alleviated MMF-induced weight loss, colonic inflammation, and oxidative phosphorylation damage. Genetic deletion of the vitamin D3 receptor (VDR), which serves as a primary colonic BA receptor, in colonic epithelial cells (VDRΔIEC) abolished the therapeutic effect of lithocholic acid on MMF-induced GI toxicity. Impressively, we discovered that paricalcitol, a Food and Drug Administration-approved VDR agonist that has been used in clinics for years, could effectively alleviate MMF-induced GI toxicity. Our study reveals a previously unrecognized mechanism of gut microbiota, BAs, and VDR signaling in MMF-induced GI side effects, offering potential therapeutic strategies for clinics.


Subject(s)
Bile Acids and Salts , Gastrointestinal Microbiome , Mycophenolic Acid , Receptors, Calcitriol , Animals , Mycophenolic Acid/pharmacology , Mice , Gastrointestinal Microbiome/drug effects , Receptors, Calcitriol/metabolism , Bile Acids and Salts/metabolism , Immunosuppressive Agents , Mice, Inbred C57BL , Male , Gastrointestinal Diseases/chemically induced , Lithocholic Acid , Humans
17.
J Steroid Biochem Mol Biol ; 240: 106507, 2024 06.
Article in English | MEDLINE | ID: mdl-38508471

ABSTRACT

Cytochrome P450 enzyme with 7ß-hydroxylation capacity has attracted widespread attentions due to the vital roles in the biosynthesis of ursodeoxycholic acid (UDCA), a naturally active molecule for the treatment of liver and gallbladder diseases. In this study, a novel P450 hydroxylase (P450FE) was screen out from Fusarium equiseti HG18 and identified by a combination of genome and transcriptome sequencing, as well as heterologous expression in Pichia pastoris. The biotransformation of lithocholic acid (LCA) by whole cells of recombinant Pichia pastoris further confirmed the C7ß-hydroxylation with 5.2% UDCA yield. It was firstly identified a fungal P450 enzyme from Fusarium equiseti HG18 with the capacity to catalyze the LCA oxidation producing UDCA. The integration of homology modeling and molecular docking discovered the substrate binding to active pockets, and the key amino acids in active center were validated by site-directed mutagenesis, and revealed that Q112, V362 and L363 were the pivotal residues of P450FE in regulating the activity and selectivity of 7ß-hydroxylation. Specifically, V362I mutation exhibited 2.6-fold higher levels of UDCA and higher stereospecificity than wild-type P450FE. This advance provided guidance for improving the catalytic efficiency and selectivity of P450FE in LCA hydroxylation, indicative of the great potential in green synthesis of UDCA from biologically toxic LCA.


Subject(s)
Cytochrome P-450 Enzyme System , Fusarium , Molecular Docking Simulation , Saccharomycetales , Ursodeoxycholic Acid , Fusarium/enzymology , Fusarium/genetics , Fusarium/metabolism , Ursodeoxycholic Acid/metabolism , Ursodeoxycholic Acid/chemistry , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/chemistry , Hydroxylation , Fungal Proteins/metabolism , Fungal Proteins/genetics , Fungal Proteins/chemistry , Mutagenesis, Site-Directed , Lithocholic Acid/metabolism , Lithocholic Acid/chemistry , Substrate Specificity
18.
J Agric Food Chem ; 72(10): 5452-5462, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38428036

ABSTRACT

Deoxynivalenol (DON) is a common mycotoxin that induces intestinal inflammation and oxidative damage in humans and animals. Given that lithocholic acid (LCA) has been suggested to inhibit intestinal inflammation, we aimed to investigate the protective effects of LCA on DON-exposed porcine intestinal epithelial IPI-2I cells and the underlying mechanisms. Indeed, LCA rescued DON-induced cell death in IPI-2I cells and reduced DON-stimulated inflammatory cytokine levels and oxidative stress. Importantly, the nuclear receptor PPARγ was identified as a key transcriptional factor involved in the DON-induced inflammation and oxidative stress processes in IPI-2I cells. The PPARγ function was found compromised, likely due to the hyperphosphorylation of the p38 and ERK signaling pathways. In contrast, the DON-induced inflammatory responses and oxidative stress were restrained by LCA via PPARγ-mediated reprogramming of the core inflammatory and antioxidant genes. Notably, the PPARγ-modulated transcriptional regulations could be attributed to the altered recruitments of coactivator SRC-1/3 and corepressor NCOR1/2, along with the modified histone marks H3K27ac and H3K18la. This study emphasizes the protective actions of LCA on DON-induced inflammatory damage and oxidative stress in intestinal epithelial cells via PPARγ-mediated epigenetically transcriptional reprogramming, including histone acetylation and lactylation.


Subject(s)
Lithocholic Acid , PPAR gamma , Trichothecenes , Humans , Animals , Swine , PPAR gamma/metabolism , Lithocholic Acid/adverse effects , Lithocholic Acid/metabolism , Epithelial Cells/metabolism , Oxidative Stress , Inflammation/metabolism
19.
Sci Rep ; 14(1): 6750, 2024 03 21.
Article in English | MEDLINE | ID: mdl-38514730

ABSTRACT

Signals for the maintenance of epithelial homeostasis are provided in part by commensal bacteria metabolites, that promote tissue homeostasis in the gut and remote organs as microbiota metabolites enter the bloodstream. In our study, we investigated the effects of bile acid metabolites, 3-oxolithocholic acid (3-oxoLCA), alloisolithocholic acid (AILCA) and isolithocholic acid (ILCA) produced from lithocholic acid (LCA) by microbiota, on the regulation of innate immune responses connected to the expression of host defense peptide cathelicidin in lung epithelial cells. The bile acid metabolites enhanced expression of cathelicidin at low concentrations in human bronchial epithelial cell line BCi-NS1.1 and primary bronchial/tracheal cells (HBEpC), indicating physiological relevance for modulation of innate immunity in airway epithelium by bile acid metabolites. Our study concentrated on deciphering signaling pathways regulating expression of human cathelicidin, revealing that LCA and 3-oxoLCA activate the surface G protein-coupled bile acid receptor 1 (TGR5, Takeda-G-protein-receptor-5)-extracellular signal-regulated kinase (ERK1/2) cascade, rather than the nuclear receptors, aryl hydrocarbon receptor, farnesoid X receptor and vitamin D3 receptor in bronchial epithelium. Overall, our study provides new insights into the modulation of innate immune responses by microbiota bile acid metabolites in the gut-lung axis, highlighting the differences in epithelial responses between different tissues.


Subject(s)
Bile Acids and Salts , Cathelicidins , Humans , Bile Acids and Salts/metabolism , Cathelicidins/metabolism , MAP Kinase Signaling System , Receptors, G-Protein-Coupled/metabolism , Epithelium/metabolism , Lithocholic Acid/pharmacology , Lithocholic Acid/metabolism
20.
Thromb Res ; 235: 68-74, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38306775

ABSTRACT

INTRODUCTION: The gut microbiome plays a crucial role in various diseases, and its regulation is a potential treatment option for these conditions. However, the relationship between the gut microbiome and venous thromboembolism (VTE) remains poorly explored. METHODS: In this study, we collected feces and serum samples from 8 VTE patients and 7 healthy controls. The gut microbiota and serum metabolites were analyzed using 16S rRNA gene sequencing and liquid chromatography-mass spectrometry, respectively. Additionally, a combined analysis of microbiota and metabolome was performed. RESULTS: The alpha and beta diversity between the VTE and control groups were significantly different. Patients with VTE exhibited an overgrowth of Blautia, Roseburia, Coprococcus, and Ruminococcus. Moreover, serum metabolomics analysis revealed altered levels of choline and lithocholic acid. Pathway enrichment analysis indicated a significant upregulation of bile secretion pathways. In addition, a positive correlation was observed between the levels of serum choline and lithocholic acid and the abundance of gut flora enriched in the VTE group. CONCLUSION: This study provided novel insights into the disordered gut microbiota and serum metabolome associated with VTE, suggesting potential common pathological mechanisms between VTE and arterial thrombosis. Targeted modulation of the gut microbiome may hold promise as a preventive and therapeutic approach for VTE.


Subject(s)
Gastrointestinal Microbiome , Venous Thromboembolism , Humans , Gastrointestinal Microbiome/genetics , RNA, Ribosomal, 16S/genetics , Metabolome , Choline , Lithocholic Acid
SELECTION OF CITATIONS
SEARCH DETAIL