Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
1.
Sci Rep ; 11(1): 17788, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493738

ABSTRACT

Bile acid profiles are altered in obese individuals with asthma. Thus, we sought to better understand how obesity-related systemic changes contribute to lung pathophysiology. We also test the therapeutic potential of nitro-oleic acid (NO2-OA), a regulator of metabolic and inflammatory signaling pathways, to mitigate allergen and obesity-induced lung function decline in a murine model of asthma. Bile acids were measured in the plasma of healthy subjects and individuals with asthma and serum and lung tissue of mice with and without allergic airway disease (AAD). Lung function, indices of inflammation and hepatic bile acid enzyme expression were measured in obese mice with house dust mite-induced AAD treated with vehicle or NO2-OA. Serum levels of glycocholic acid and glycoursodeoxycholic acid clinically correlate with body mass index and airway hyperreactivity whereas murine levels of ß-muricholic acid and tauro-ß-muricholic acid were significantly increased and positively correlated with impaired lung function in obese mice with AAD. NO2-OA reduced murine bile acid levels by modulating hepatic expression of bile acid synthesis enzymes, with a concomitant reduction in small airway resistance and tissue elastance. Bile acids correlate to body mass index and lung function decline and the signaling actions of nitroalkenes can limit AAD by modulating bile acid metabolism, revealing a potential pharmacologic approach to improving the current standard of care.


Subject(s)
Asthma/metabolism , Asthma/physiopathology , Bile Acids and Salts/metabolism , Fatty Acids/physiology , Lung/physiopathology , Nitro Compounds/therapeutic use , Obesity/metabolism , Oleic Acids/therapeutic use , Adolescent , Adult , Animals , Anti-Asthmatic Agents/therapeutic use , Antigens, Dermatophagoides/toxicity , Asthma/drug therapy , Asthma/etiology , Diet, High-Fat/adverse effects , Drug Evaluation, Preclinical , Fatty Acids/chemistry , Female , Forced Expiratory Volume , Glycocholic Acid/blood , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/complications , Obesity/physiopathology , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/metabolism , Thinness , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/blood , Vital Capacity , Young Adult
2.
Clin Sci (Lond) ; 135(14): 1689-1706, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34236076

ABSTRACT

Recent studies reveal that bile acid metabolite composition and its metabolism are changed in metabolic disorders, such as obesity, type 2 diabetes and metabolic associated fatty liver disease (MAFLD), yet its role and the mechanism remain largely unknown. In the present study, metabolomic analysis of 163 serum and stool samples of our metabolic disease cohort was performed, and we identified glycoursodeoxycholic acid (GUDCA), glycine-conjugated bile acid produced from intestinal bacteria, was decreased in both serum and stool samples from patients with hyperglycemia. RNA-sequencing and quantitative PCR results indicated that GUDCA alleviated endoplasmic reticulum (ER) stress in livers of high fat diet (HFD)-fed mice without alteration of liver metabolism. In vitro, GUDCA reduced palmitic acid induced-ER stress and -apoptosis, as well as stabilized calcium homeostasis. In vivo, GUDCA exerted effects on amelioration of HFD-induced insulin resistance and hepatic steatosis. In parallel, ER stress and apoptosis were decreased in GUDCA-treated mice as compared with vehicle-treated mice in liver. These findings demonstrate that reduced GUDCA is an indicator of hyperglycemia. Supplementation of GUDCA could be an option for the treatment of diet-induced metabolic disorders, including insulin resistance and hepatic steatosis, with inhibiting ER stress.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Lipid Metabolism/drug effects , Metabolic Diseases/drug therapy , Obesity/metabolism , Ursodeoxycholic Acid/analogs & derivatives , Animals , Diet, High-Fat/methods , Endoplasmic Reticulum Stress/physiology , Fatty Liver/drug therapy , Fatty Liver/metabolism , Female , Humans , Lipid Metabolism/physiology , Liver/drug effects , Liver/metabolism , Male , Metabolic Diseases/metabolism , Middle Aged , Ursodeoxycholic Acid/pharmacology
3.
J Hepatol ; 75(5): 1164-1176, 2021 11.
Article in English | MEDLINE | ID: mdl-34242699

ABSTRACT

BACKGROUND & AIMS: 24-Norursodeoxycholic acid (NorUDCA) is a novel therapeutic bile acid used to treat immune-mediated cholestatic liver diseases, such as primary sclerosing cholangitis (PSC), where dysregulated T cells including CD8+ T cells contribute to hepatobiliary immunopathology. We hypothesized that NorUDCA may directly modulate CD8+ T cell function thus contributing to its therapeutic efficacy. METHODS: NorUDCA's immunomodulatory effects were first studied in Mdr2-/- mice, as a cholestatic model of PSC. To differentiate NorUDCA's immunomodulatory effects on CD8+ T cell function from its anticholestatic actions, we also used a non-cholestatic model of hepatic injury induced by an excessive CD8+ T cell immune response upon acute non-cytolytic lymphocytic choriomeningitis virus (LCMV) infection. Studies included molecular and biochemical approaches, flow cytometry and metabolic assays in murine CD8+ T cells in vitro. Mass spectrometry was used to identify potential CD8+ T cell targets modulated by NorUDCA. The signaling effects of NorUDCA observed in murine cells were validated in circulating T cells from patients with PSC. RESULTS: NorUDCA demonstrated immunomodulatory effects by reducing hepatic innate and adaptive immune cells, including CD8+ T cells in the Mdr2-/- model. In the non-cholestatic model of CD8+ T cell-driven immunopathology induced by acute LCMV infection, NorUDCA ameliorated hepatic injury and systemic inflammation. Mechanistically, NorUDCA demonstrated strong immunomodulatory efficacy in CD8+ T cells affecting lymphoblastogenesis, expansion, glycolysis and mTORC1 signaling. Mass spectrometry identified that NorUDCA regulates CD8+ T cells by targeting mTORC1. NorUDCA's impact on mTORC1 signaling was further confirmed in circulating PSC CD8+ T cells. CONCLUSIONS: NorUDCA has a direct modulatory impact on CD8+ T cells and attenuates excessive CD8+ T cell-driven hepatic immunopathology. These findings are relevant for treatment of immune-mediated liver diseases such as PSC. LAY SUMMARY: Elucidating the mechanisms by which 24-norursodeoxycholic acid (NorUDCA) works for the treatment of immune-mediated liver diseases, such as primary sclerosing cholangitis, is of considerable clinical interest. Herein, we uncovered an unrecognized property of NorUDCA in the immunometabolic regulation of CD8+ T cells, which has therapeutic relevance for immune-mediated liver diseases, including PSC.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Inflammation/drug therapy , Liver/drug effects , Ursodeoxycholic Acid/analogs & derivatives , Animals , CD8-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Inflammation/physiopathology , Liver/physiopathology , Mice , Mice, Inbred C57BL , Ursodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/therapeutic use
4.
J Clin Invest ; 131(9)2021 05 03.
Article in English | MEDLINE | ID: mdl-33938457

ABSTRACT

Intestinal farnesoid X receptor (FXR) signaling is involved in the development of obesity, fatty liver disease, and type 2 diabetes. However, the role of intestinal FXR in atherosclerosis and its potential as a target for clinical treatment have not been explored. The serum levels of fibroblast growth factor 19 (FGF19), which is encoded by an FXR target gene, were much higher in patients with hypercholesterolemia than in control subjects and were positively related to circulating ceramide levels, indicating a link between intestinal FXR, ceramide metabolism, and atherosclerosis. Among ApoE-/- mice fed a high-cholesterol diet (HCD), intestinal FXR deficiency (in FxrΔIE ApoE-/- mice) or direct FXR inhibition (via treatment with the FXR antagonist glycoursodeoxycholic acid [GUDCA]) decreased atherosclerosis and reduced the levels of circulating ceramides and cholesterol. Sphingomyelin phosphodiesterase 3 (SMPD3), which is involved in ceramide synthesis in the intestine, was identified as an FXR target gene. SMPD3 overexpression or C16:0 ceramide supplementation eliminated the improvements in atherosclerosis in FxrΔIE ApoE-/- mice. Administration of GUDCA or GW4869, an SMPD3 inhibitor, elicited therapeutic effects on established atherosclerosis in ApoE-/- mice by decreasing circulating ceramide levels. This study identified an intestinal FXR/SMPD3 axis that is a potential target for atherosclerosis therapy.


Subject(s)
Atherosclerosis , Ceramides/biosynthesis , Intestinal Mucosa/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Ursodeoxycholic Acid/analogs & derivatives , Animals , Atherosclerosis/chemically induced , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Ceramides/genetics , Diet, High-Fat/adverse effects , Female , Humans , Male , Mice , Mice, Knockout, ApoE , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Sphingomyelin Phosphodiesterase/genetics , Ursodeoxycholic Acid/pharmacology
5.
Biomolecules ; 11(5)2021 04 23.
Article in English | MEDLINE | ID: mdl-33922434

ABSTRACT

We previously demonstrated that the bile acid taurocholic acid (TCA) inhibits features of age-related macular degeneration (AMD) in vitro. The purpose of this study was to determine if the glycine-conjugated bile acids glycocholic acid (GCA), glycodeoxycholic acid (GDCA), and glycoursodeoxycholic acid (GUDCA) can protect retinal pigment epithelial (RPE) cells against oxidative damage and inhibit vascular endothelial growth factor (VEGF)-induced angiogenesis in choroidal endothelial cells (CECs). Paraquat was used to induce oxidative stress and disrupt tight junctions in HRPEpiC primary human RPE cells. Tight junctions were assessed via transepithelial electrical resistance and ZO-1 immunofluorescence. GCA and GUDCA protected RPE tight junctions against oxidative damage at concentrations of 100-500 µM, and GDCA protected tight junctions at 10-500 µM. Angiogenesis was induced with VEGF in RF/6A macaque CECs and evaluated with cell proliferation, cell migration, and tube formation assays. GCA inhibited VEGF-induced CEC migration at 50-500 µM and tube formation at 10-500 µM. GUDCA inhibited VEGF-induced CEC migration at 100-500 µM and tube formation at 50-500 µM. GDCA had no effect on VEGF-induced angiogenesis. None of the three bile acids significantly inhibited VEGF-induced CEC proliferation. These results suggest glycine-conjugated bile acids may be protective against both atrophic and neovascular AMD.


Subject(s)
Bile Acids and Salts/metabolism , Neovascularization, Pathologic/prevention & control , Retinal Pigment Epithelium/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cell Culture Techniques , Cell Movement/drug effects , Cell Proliferation/drug effects , Choroid/metabolism , Endothelial Cells/metabolism , Glycine/metabolism , Glycocholic Acid/pharmacology , Glycodeoxycholic Acid/pharmacology , Humans , Macaca mulatta , Neovascularization, Pathologic/metabolism , Oxidative Stress/drug effects , Tight Junctions/metabolism , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Wet Macular Degeneration/metabolism
6.
J Am Heart Assoc ; 10(7): e019820, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33787322

ABSTRACT

Background Although glycoursodeoxycholic acid (GUDCA) has been associated with the improvement of metabolic disorders, its effect on atherosclerosis remains elusive. This study aimed to investigate the role of GUDCA in the development of atherosclerosis and its potential mechanisms. Methods and Results Human THP-1 macrophages were used to investigate the effect of GUDCA on oxidized low-density lipoprotein-induced foam cell formation in vitro. We found that GUDCA downregulated scavenger receptor A1 mRNA expression, reduced oxidized low-density lipoprotein uptake, and inhibited macrophage foam cell formation. In an in vivo study, apolipoprotein E-deficient mice were fed a Western diet for 10 weeks to induce atherosclerosis, and then were gavaged once daily with or without GUDCA for 18 weeks. Parameters of systemic metabolism and atherosclerosis were detected. We found that GUDCA improved cholesterol homeostasis and protected against atherosclerosis progression as evidenced by reduced plaque area along with lipid deposition, ameliorated local chronic inflammation, and elevated plaque stability. In addition, 16S rDNA sequencing showed that GUDCA administration partially normalized the Western diet-associated gut microbiota dysbiosis. Interestingly, the changes of bacterial genera (Alloprevotella, Parabacteroides, Turicibacter, and Alistipes) modulated by GUDCA were correlated with the plaque area in mice aortas. Conclusions Our study for the first time indicates that GUDCA attenuates the development of atherosclerosis, probably attributable to the inhibition of foam cell formation, maintenance of cholesterol homeostasis, and modulation of gut microbiota.


Subject(s)
Atherosclerosis/drug therapy , Gastrointestinal Microbiome/physiology , Gene Expression Regulation , RNA, Messenger/genetics , Scavenger Receptors, Class A/genetics , Ursodeoxycholic Acid/analogs & derivatives , Animals , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Down-Regulation/drug effects , Female , Foam Cells/drug effects , Foam Cells/metabolism , Foam Cells/pathology , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Scavenger Receptors, Class A/biosynthesis , Ursodeoxycholic Acid/pharmacology
7.
Expert Rev Gastroenterol Hepatol ; 14(10): 985-998, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32674617

ABSTRACT

INTRODUCTION: Primary biliary cholangitis (PBC) is a chronic cholestatic liver disease which on progression causes cirrhosis; various studies also suggested that several diseases can co-exist in patients. In existing depiction of disease PBC, apart from entire use of ursodeoxycholic acid (UDCA), several patients need to step forward to liver-transplantation or death due to resistance or non-responder with UDCA monotherapy. AREAS COVERED: To overcome this non-respondent treatment, novel bile acid semi-synthetic analogs have been identified which shows their potency against for farnesoid X receptor and transmembrane G protein-coupled receptor-5 which are identified as target for many developing analogs which have desirable pharmacokinetic profiles. EXPERT OPINION: A range of studies suggests that adding semisynthetic analogs in therapeutic regime improves liver biochemistries in patients with suboptimal response to UDCA. Thus, the aspire of this review is to abridge and compare therapeutic value and current markets affirm of various bile acids semi-synthetic analogs which certainly are having promising effects in PBC monotherapy or in pooled treatment with UDCA for PBC.


Subject(s)
Cholagogues and Choleretics/therapeutic use , Liver Cirrhosis, Biliary/drug therapy , Ursodeoxycholic Acid/therapeutic use , Animals , Bile Acids and Salts/therapeutic use , Chenodeoxycholic Acid/analogs & derivatives , Chenodeoxycholic Acid/therapeutic use , Cholic Acids/therapeutic use , Humans , Liver Cirrhosis, Biliary/etiology , Nitrates/therapeutic use , Ursodeoxycholic Acid/analogs & derivatives
8.
FASEB J ; 34(5): 6198-6214, 2020 05.
Article in English | MEDLINE | ID: mdl-32162746

ABSTRACT

Mitochondrial dysfunction is the leading cause of reactive oxygen species (ROS) burst and apoptosis in hepatic ischemia/reperfusion (I/R) injury. Ursodeoxycholyl lysophosphatidylethanolamide (UDCA-LPE) is a hepatotargeted agent that exerts hepatoprotective roles by regulating lipid metabolism. Our previous studies have shown that UDCA-LPE improves hepatic I/R injury by inhibiting apoptosis and inflammation. However, the role of UDCA-LPE in lipid metabolism and mitochondrial function in hepatic I/R remains unknown. In the present study, we investigated the role of UDCA-LPE in hepatic I/R by focusing on the interface of phospholipid metabolism and mitochondrial homeostasis. Livers from 28-week-old mice, primary hepatocytes and HepG2 cells were subjected to in vivo and in vitro I/R, respectively. Analyses of oxidative stress, imaging, ATP generation, genetics, and lipidomics showed that I/R was associated with mitochondrial dysfunction and a reduction in phospholipids. UDCA-LPE alleviated mitochondria-dependent oxidative stress and apoptosis and prevented the decrease of phospholipid levels. Our study found that cytosolic phospholipase A2 (cPLA2 ), a phospholipase that is activated during I/R, hydrolyzed mitochondrial membrane phospholipids and led to mitochondria-mediated oxidative stress and apoptosis. UDCA-LPE inhibited the interaction between cPLA2 and mitochondria and reduced phospholipid hydrolysis-mediated injury. UDCA-LPE might regulate the crosstalk between the phospholipid metabolism and the mitochondria, restore mitochondrial function and ameliorate I/R injury.


Subject(s)
Lipid Metabolism Disorders/prevention & control , Liver Diseases/prevention & control , Lysophospholipids/pharmacology , Mitochondria/drug effects , Oxidative Stress/drug effects , Phospholipids/metabolism , Reperfusion Injury/complications , Ursodeoxycholic Acid/analogs & derivatives , Animals , Apoptosis , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lipid Metabolism Disorders/etiology , Lipid Metabolism Disorders/metabolism , Liver Diseases/etiology , Liver Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Protective Agents/pharmacology , Reactive Oxygen Species , Ursodeoxycholic Acid/pharmacology
9.
Int J Mol Sci ; 20(22)2019 Nov 11.
Article in English | MEDLINE | ID: mdl-31717968

ABSTRACT

The bile acid-phospholipid conjugate ursodeoxycholyl-lysophosphatidylethanolamide (UDCA-LPE) was shown to have anti-inflammatory, antisteatotic, and antifibrotic properties, rendering it as a drug targeting non-alcoholic steatohepatitis (NASH). On a molecular level, it disrupted the heterotetrameric fatty acid uptake complex localized in detergent-resistant membrane domains of the plasma membrane (DRM-PM). However, its mode of action was unclear. Methodologically, UDCA-LPE was incubated with the liver tumor cell line HepG2 as well as their isolated DRM-PM and all other cellular membranes (non-DRM). The membrane cholesterol and phospholipids were quantified as well as the DRM-PM protein composition by Western blotting. The results show a loss of DRM-PM by UDCA-LPE (50 µM) with a 63.13 ± 7.14% reduction of phospholipids and an 81.94 ± 8.30% reduction of cholesterol in relation to mg total protein. The ratio of phospholipids to cholesterol changed from 2:1 to 4:1, resembling those of non-DRM fractions. Among the members of the fatty acid uptake complex, the calcium-independent membrane phospholipase A2 (iPLA2ß) abandoned DRM-PM most rapidly. As a consequence, the other members of this transport system disappeared as well as the DRM-PM anchored fibrosis regulating proteins integrin ß-1 and lysophospholipid receptor 1 (LPAR-1). It is concluded that UDCA-LPE executes its action by iPLA2ß removal from DRM-PM and consequent dissolution of the raft lipid platform.


Subject(s)
Cholesterol/metabolism , Lysophospholipids/pharmacology , Membrane Microdomains/drug effects , Phospholipases A2/metabolism , Ursodeoxycholic Acid/analogs & derivatives , Hep G2 Cells , Humans , Integrin beta1/metabolism , Membrane Microdomains/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Ursodeoxycholic Acid/pharmacology
10.
World J Gastroenterol ; 25(33): 4835-4849, 2019 Sep 07.
Article in English | MEDLINE | ID: mdl-31543677

ABSTRACT

Liver fibrosis is the common pathological basis of all chronic liver diseases, and is the necessary stage for the progression of chronic liver disease to cirrhosis. As one of pathogenic factors, inflammation plays a predominant role in liver fibrosis via communication and interaction between inflammatory cells, cytokines, and the related signaling pathways. Damaged hepatocytes induce an increase in pro-inflammatory factors, thereby inducing the development of inflammation. In addition, it has been reported that inflammatory response related signaling pathway is the main signal transduction pathway for the development of liver fibrosis. The crosstalk regulatory network leads to hepatic stellate cell activation and proinflammatory cytokine production, which in turn initiate the fibrotic response. Compared with the past, the research on the pathogenesis of liver fibrosis has been greatly developed. However, the liver fibrosis mechanism is complex and many pathways involved need to be further studied. This review mainly focuses on the crosstalk regulatory network among inflammatory cells, cytokines, and the related signaling pathways in the pathogenesis of chronic inflammatory liver diseases. Moreover, we also summarize the recent studies on the mechanisms underlying liver fibrosis and clinical efforts on the targeted therapies against the fibrotic response.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/immunology , Liver Cirrhosis/immunology , Liver/pathology , Signal Transduction/immunology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Disease Models, Animal , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/metabolism , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/metabolism , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Inflammation/drug therapy , Inflammation/pathology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Liver/cytology , Liver/immunology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Signal Transduction/drug effects , Sulfoxides , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/therapeutic use
11.
Pharmacol Ther ; 203: 107396, 2019 11.
Article in English | MEDLINE | ID: mdl-31356908

ABSTRACT

Ursodeoxycholic acid (UDCA) is a secondary bile acid issued from the transformation of (cheno)deoxycholic acid by intestinal bacteria, acting as a key regulator of the intestinal barrier integrity and essential for lipid metabolism. UDCA is also a long-established drug, largely used for the dissolution of cholesterol gallstones, the treatment of primary biliary cholangitis and other hepatobiliary disorders. The history of UDCA is briefly retraced here as well as its multifactorial mechanism of action, based on its anti-inflammatory, antioxidant and cytoprotective activities. The present review is centred around the anticancer properties of UDCA and synthetic antitumor derivatives designed over the past 20 years. Paradoxically, depending on the conditions, UDCA exhibits both pro- and anti-apoptotic properties toward different cell types. In particular, the UDCA drug can protect epithelial cells from damages and apoptosis while inducing inhibition of proliferation and apoptotic and/or autophagic death of cancer cells. The effects of UDCA on cancer cell migration, cancer stem cells and drug-induced dysbiosis are also evoked. The drug has revealed modest activities against colon and gastric cancers but may be useful to improve treatments of hepatocellular carcinoma, notably in combination with other drugs such as sorafenib. UDCA can also protect from damages induced by cancer chemotherapeutic agents. The potential of UDCA in cancer, as a chemo-protecting or chemotherapeutic agent, is highlighted here as well as the design of tumour-active derivatives, including UDCA-drug conjugates. A repurposing of UDCA in oncology should be further considered.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/prevention & control , Ursodeoxycholic Acid/therapeutic use , Animals , Humans , Ursodeoxycholic Acid/analogs & derivatives
12.
Lancet Gastroenterol Hepatol ; 4(10): 781-793, 2019 10.
Article in English | MEDLINE | ID: mdl-31345778

ABSTRACT

BACKGROUND: Norursodeoxycholic acid is an orally administered side chain-shortened homologue of ursodeoxycholic acid that undergoes hepatic enrichment with hepatoprotective, anti-inflammatory, and antifibrotic activity. We assessed the efficacy of two doses of norursodeoxycholic acid versus placebo for the treatment of non-alcoholic fatty liver disease. METHODS: We did a multicentre, double-blind, placebo-controlled, randomised, phase 2 dose-finding clinical trial in tertiary referral hospitals and medical centres in Austria (n=6) and Germany (n=23) for patients with non-alcoholic fatty liver disease with or without diabetes. Patients with a clinical diagnosis of non-alcoholic fatty liver disease and serum alanine aminotransferase (ALT) concentrations of more than 0·8 times the upper limit of normal were randomly assigned (1:1:1) using a computer-generated central randomisation. Patients were randomly assigned to receive either norursodeoxycholic acid capsules at 500 mg per day or 1500 mg per day, or placebo, for 12 weeks with a subsequent 4-week follow-up period. All individuals involved in the trial were masked to treatment allocation. The primary efficacy endpoint was the mean relative percentage change in ALT concentrations between baseline and end of treatment assessed in the intention-to-treat population. This trial is registered with EudraCT, number 2013-004605-38. FINDINGS: Between March 30, 2015, and Sept 20, 2016, of 198 individuals included in the analysis, 67 patients were randomly assigned to receive 500 mg norursodeoxycholic acid, 67 to 1500 mg norursodeoxycholic acid, and 64 to placebo. A dose-dependent reduction in serum ALT between baseline and end of treatment was observed with norursodeoxycholic acid versus placebo, with a significant effect in the 1500 mg group (mean change -27·8%, 95% repeated CI -34·7 to -14·4; p<0·0001). Serious adverse events (n=6) and treatment-emergent adverse events (n=314) were reported in a similar proportion of patients across groups. 112 treatment-emergent adverse events occurred in the 1500 mg group, 99 in the 500 mg group, and 103 in the placebo group. The most frequent adverse events were headache, gastrointestinal disorders, and infections (eg, diarrhoea, abdominal pain, or nasopharyngitis). INTERPRETATION: Norursodeoxycholic acid at 1500 mg resulted in a significant reduction of serum ALT within 12 weeks of treatment when compared with placebo. Norursodeoxycholic acid was safe and well tolerated encouraging further studies. FUNDING: Dr Falk Pharma GmbH.


Subject(s)
Cholagogues and Choleretics/administration & dosage , Non-alcoholic Fatty Liver Disease/drug therapy , Ursodeoxycholic Acid/analogs & derivatives , Adult , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Blood Glucose/metabolism , Cholagogues and Choleretics/adverse effects , Cholagogues and Choleretics/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Lipids/blood , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Treatment Outcome , Ursodeoxycholic Acid/administration & dosage , Ursodeoxycholic Acid/adverse effects , Ursodeoxycholic Acid/therapeutic use
13.
Chem Biol Interact ; 310: 108745, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31299240

ABSTRACT

Ursodeoxycholic acid (UDCA) is a major effective constituent of bear bile powder, which is widely used as function food in China and is documented in the Chinese pharmacopoeia as a traditional Chinese medicine. UDCA has been developed as the only accepted therapy by the US FDA for primary biliary cholangitis. Recently, the US FDA granted accelerated approval to obeticholic acid (OCA), a semisynthetic bile acid derivative from chenodeoxycholic acid, for primary biliary cholangitis. However, some perplexing toxicities of UDCA have been reported in the clinic. The present work aimed to investigate the difference between UDCA and OCA in regard to potential metabolic activation through acyl glucuronidation and hepatic accumulation of consequent acyl glucuronides. Our results demonstrated that the metabolic fates of UDCA and OCA were similar. Both UDCA and OCA were predominantly metabolically activated by conjugation to the acyl glucuronide in human liver microsomes. UGT1A3 played a predominant role in the carboxyl glucuronidation of both UDCA and OCA, while UGT2B7 played a major role in their hydroxyl glucuronidation. Further uptake studies revealed that OATP1B1- and 1B3-transfected cells could selectively uptake UDCA acyl glucuronide, but not UDCA, OCA, and OCA acyl glucuronide. In summary, the liver disposition of OCA is different from that of UDCA due to hepatic uptake, and liver accumulation of UDCA acyl glucuronide might be related to the perplexing toxicities of UDCA.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Glucuronides/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , Microsomes, Liver/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Ursodeoxycholic Acid/metabolism , Animals , Biological Transport , Chenodeoxycholic Acid/metabolism , Humans , Medicine, Chinese Traditional , Ursidae , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/toxicity
14.
Handb Exp Pharmacol ; 256: 237-264, 2019.
Article in English | MEDLINE | ID: mdl-31236688

ABSTRACT

Bile acids (BAs) are key molecules in generating bile flow, which is an essential function of the liver. In the last decades, there have been great advances in the understanding of BA physiology, and new insights have emerged regarding the role of BAs in determining cell damage and death in several liver diseases. This new knowledge has helped to better delineate the pathophysiology of cholestasis and the adaptive responses of hepatocytes to cholestatic liver injury as well as of the mechanisms of injury of biliary epithelia. In this context, therapeutic approaches for liver diseases using hydrophilic BA (i.e., ursodeoxycholic acid, tauroursodeoxycholic, and, more recently, norursodeoxycholic acid), have been revamped. In the present review, we summarize current experimental and clinical data regarding these BAs and its role in the treatment of certain liver diseases.


Subject(s)
Liver Diseases/drug therapy , Taurochenodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/pharmacology , Bile Acids and Salts , Cholestasis , Humans , Liver
15.
Chem Phys Lipids ; 223: 104778, 2019 09.
Article in English | MEDLINE | ID: mdl-31173728

ABSTRACT

Bile salts (BS) form hydrophobic Small's primary micelles at concentrations above the critical micelle concentration (CMC), while at concentrations above 3CMC they form secondary micelles (by the association of primary micelles via H-bonds). In this paper the self-associations of the anions of isohenodeoxycholic acid (3-epimer of henodeoxycholic acid, ICD) and the anions of isoursodeoxycholic acid (3-epimer of ursodeoxycholic acid, IUD) are examined, since the thermodynamic parameters of their self-association have not yet been published. Forming of IUD aggregates with two or three building units is slightly more favorable via α sides of steroid skeletons, regarding hydrophobicity, while regarding steric repulsive interactions it is more favorable to associate via ß sides. Due to this, IUD in the vicinity of the CMC can form primary micelles by association of IUD particles both from the convex side and from the concave side of the steroid ring system. Therefore, IUD is significantly more prone to initial micellization than bile salt derivatives whose steroidal skeletons contain equatorially oriented OH groups.


Subject(s)
Deoxycholic Acid/chemistry , Sodium/chemistry , Ursodeoxycholic Acid/analogs & derivatives , Water/chemistry , Deoxycholic Acid/analogs & derivatives , Molecular Conformation , Thermodynamics , Ursodeoxycholic Acid/chemistry
16.
Nat Med ; 24(12): 1919-1929, 2018 12.
Article in English | MEDLINE | ID: mdl-30397356

ABSTRACT

The anti-hyperglycemic effect of metformin is believed to be caused by its direct action on signaling processes in hepatocytes, leading to lower hepatic gluconeogenesis. Recently, metformin was reported to alter the gut microbiota community in humans, suggesting that the hyperglycemia-lowering action of the drug could be the result of modulating the population of gut microbiota. However, the critical microbial signaling metabolites and the host targets associated with the metabolic benefits of metformin remained elusive. Here, we performed metagenomic and metabolomic analysis of samples from individuals with newly diagnosed type 2 diabetes (T2D) naively treated with metformin for 3 d, which revealed that Bacteroides fragilis was decreased and the bile acid glycoursodeoxycholic acid (GUDCA) was increased in the gut. These changes were accompanied by inhibition of intestinal farnesoid X receptor (FXR) signaling. We further found that high-fat-diet (HFD)-fed mice colonized with B. fragilis were predisposed to more severe glucose intolerance, and the metabolic benefits of metformin treatment on glucose intolerance were abrogated. GUDCA was further identified as an intestinal FXR antagonist that improved various metabolic endpoints in mice with established obesity. Thus, we conclude that metformin acts in part through a B. fragilis-GUDCA-intestinal FXR axis to improve metabolic dysfunction, including hyperglycemia.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Microbiome/drug effects , Metformin/administration & dosage , Obesity/drug therapy , Receptors, Cytoplasmic and Nuclear/genetics , Bacteroides/drug effects , Bacteroides/pathogenicity , Bile Acids and Salts/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/genetics , Gene Expression Regulation, Bacterial/drug effects , Glucose Intolerance/drug therapy , Glucose Intolerance/genetics , Glucose Intolerance/microbiology , Humans , Hyperglycemia/drug therapy , Hyperglycemia/genetics , Hyperglycemia/microbiology , Hyperglycemia/pathology , Metabolome/drug effects , Metabolome/genetics , Metagenomics/methods , Obesity/genetics , Obesity/microbiology , Obesity/pathology , Ursodeoxycholic Acid/analogs & derivatives
17.
Int J Mol Sci ; 19(10)2018 Oct 20.
Article in English | MEDLINE | ID: mdl-30347788

ABSTRACT

Ursodeoxycholyl lysophosphatidylethanolamide (UDCA-LPE) is a synthetic bile acid-phospholipid conjugate with profound hepatoprotective and anti-fibrogenic functions in vitro and in vivo. Herein, we aimed to demonstrate the inhibitory effects of UDCA-LPE on pro-fibrogenic integrin signalling. UDCA-LPE treatment of human embryonic liver cell line CL48 and primary human hepatic stellate cells induced a non-classical internalization of integrin ß1 resulting in dephosphorylation and inhibition of SRC and focal adhesion kinase (FAK). Signalling analyses suggested that UDCA-LPE may act as a heterobivalent ligand for integrins and lysophospholipid receptor1 (LPAR1) and co-immunoprecipitation demonstrated the bridging effect of UDCA-LPE on integrin ß1 and LPAR1. The disruption of either the UDCA-moiety binding to integrins by RGD-containing peptide GRGDSP or the LPE-moiety binding to LPAR1 by LPAR1 antagonist Ki16425 reversed inhibitory functions of UDCA-LPE. The lack of inhibitory functions of UDCA-PE and UDCA-LPE derivatives (14:0 and 12:0, LPE-moiety containing shorter fatty acid chain) as well as the consistency of the translocation of UDCA-LPE and integrins, which co-fractionated with LPE but not UDCA, suggested that the observed UDCA-LPE-induced translocation of integrins was mediated by LPE endocytic transport pathway.


Subject(s)
Endocytosis , Integrin beta1/metabolism , Lysophospholipids/pharmacology , Membrane Microdomains/metabolism , Ursodeoxycholic Acid/analogs & derivatives , Cell Line , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Ursodeoxycholic Acid/pharmacology
18.
PLoS One ; 13(8): e0200897, 2018.
Article in English | MEDLINE | ID: mdl-30067827

ABSTRACT

Alpha-1 Antitrypsin (α1AT) Deficiency is a genetic disease in which accumulation of α1AT mutant Z (α1ATZ) protein in the ER of hepatocytes causes chronic liver injury, liver fibrosis, and hepatocellular carcinoma. No effective medical therapy is currently available for the disease. We previously found that norUDCA improves the α1AT deficiency associated liver disease by promoting autophagic degradation of α1ATZ protein in liver in a mouse model of the disease. The current study unravels the novel underlying cellular mechanism by which norUDCA modulates autophagy. HTOZ cells, modified from HeLa Tet-Off cells by transfection with the resulting pTRE1-ATZ plasmid and expressing mutant Z proteins, were studied in these experiments. The role of norUDCA in inducing autophagy, autophagy-mediated degradation of α1ATZ and the role of AMPK in norUDCA-induced autophagy were examined in the current report. NorUDCA promoted disposal of α1ATZ via autophagy-mediated degradation of α1ATZ in HTOZ cells. Activation of AMPK was required for norUDCA-induced autophagy and α1ATZ degradation. Moreover, mTOR/ULK1 was involved in norUDCA-induced AMPK activation and autophagy in HTOZ cells. Our results provide novel mechanistic insights into the therapeutic action of norUDCA in promoting the clearance of α1ATZ in vitro and suggest a novel therapeutic approach for the treatment of α1ATZ deficiency disease and its associated liver diseases.


Subject(s)
Autophagy , Ursodeoxycholic Acid/analogs & derivatives , alpha 1-Antitrypsin/metabolism , Adenylate Kinase/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , Disease Models, Animal , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Liver/metabolism , Liver Diseases/drug therapy , Liver Diseases/metabolism , Mice , Mutant Proteins/metabolism , Proteolysis , RNA, Messenger/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Ursodeoxycholic Acid/metabolism , Ursodeoxycholic Acid/pharmacology
19.
PLoS One ; 13(5): e0197836, 2018.
Article in English | MEDLINE | ID: mdl-29795632

ABSTRACT

AIM: Endotoxin-mediated liver inflammation is a key component of many acute and chronic liver diseases contributing to liver damage, fibrosis and eventually organ failure. Here, we investigated ursodeoxycholyl lysophosphatidylethanolamide (UDCA-LPE), a synthetic bile acid-phospholipid conjugate regarding its anti-inflammatory and anti-fibrogenic properties. METHODS: Anti-inflammatory properties of UDCA-LPE were evaluated in a mouse model of D-galactosamine/lipopolysaccharide (GalN/LPS)-induced acute liver injury, LPS treated RAW264.7 macrophages and murine primary Kupffer cells. Furthermore, anti-inflammatory and anti-fibrotic effects of UDCA-LPE were studied on primary hepatic stellate cells (HSC) incubated with supernatant from LPS±UDCA-LPE treated RAW264.7 cells. RESULTS: UDCA-LPE ameliorated LPS-induced increase of IL-6, TNF-α, TGF-ß, NOX-2 in the GalN/LPS model by up to 80.2% for IL-6. Similarly, UDCA-LPE markedly decreased the expression of inflammatory cytokines IL-6, TNF-α and TGF-ß as well as the chemokines MCP1 and RANTES in LPS-stimulated RAW 264.7 cells. Anti-inflammatory effects were also observed in primary murine Kupffer cells. Mechanistic evaluation revealed a reversion of LPS-activated pro-inflammatory TLR4 pathway by UDCA-LPE. Moreover, UDCA-LPE inhibited iNOS and NOX-2 expression while activating eNOS via phosphorylation of AKT and pERK1/2 in RAW264.7 cells. HSC treated with conditioned medium from LPS±UDCA-LPE RAW264.7 cells showed lower fibrogenic activation due to less SMAD2/3 phosphorylation, reduced expression of profibrogenic CTGF and reduced pro-inflammatory chemokine expression. CONCLUSION: In the setting of endotoxin-mediated liver inflammation, UDCA-LPE exerts profound anti-inflammatory and anti-fibrotic effect implying a promising potential for the drug candidate as an experimental approach for the treatment of acute and chronic liver diseases.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Endotoxins/toxicity , Inflammation/drug therapy , Lipopolysaccharides/toxicity , Lysophospholipids/pharmacology , Ursodeoxycholic Acid/analogs & derivatives , Animals , Anti-Inflammatory Agents , Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Inflammation/chemically induced , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism , Ursodeoxycholic Acid/pharmacology
20.
Prog Retin Eye Res ; 65: 50-76, 2018 07.
Article in English | MEDLINE | ID: mdl-29481975

ABSTRACT

Diseases that affect the eye, including photoreceptor degeneration, diabetic retinopathy, and glaucoma, affect 11.8 million people in the US, resulting in vision loss and blindness. Loss of sight affects patient quality of life and puts an economic burden both on individuals and the greater healthcare system. Despite the urgent need for treatments, few effective options currently exist in the clinic. Here, we review research on promising neuroprotective strategies that promote neuronal survival with the potential to protect against vision loss and retinal cell death. Due to the large number of neuroprotective strategies, we restricted our review to approaches that we had direct experience with in the laboratory. We focus on drugs that target survival pathways, including bile acids like UDCA and TUDCA, steroid hormones like progesterone, therapies that target retinal dopamine, and neurotrophic factors. In addition, we review rehabilitative methods that increase endogenous repair mechanisms, including exercise and electrical stimulation therapies. For each approach, we provide background on the neuroprotective strategy, including history of use in other diseases; describe potential mechanisms of action; review the body of research performed in the retina thus far, both in animals and in humans; and discuss considerations when translating each treatment to the clinic and to the retina, including which therapies show the most promise for each retinal disease. Despite the high incidence of retinal diseases and the complexity of mechanisms involved, several promising neuroprotective treatments provide hope to prevent blindness. We discuss attractive candidates here with the goal of furthering retinal research in critical areas to rapidly translate neuroprotective strategies into the clinic.


Subject(s)
Neuroprotective Agents , Retinal Diseases/therapy , Animals , Cell Death/drug effects , Electric Stimulation Therapy , Exercise Therapy , Humans , Nerve Growth Factors/pharmacology , Nerve Growth Factors/therapeutic use , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Retina/drug effects , Taurochenodeoxycholic Acid/pharmacology , Taurochenodeoxycholic Acid/therapeutic use , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/therapeutic use , Vision Disorders/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...