Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
N Engl J Med ; 383(10): 919-930, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32877582

ABSTRACT

BACKGROUND: Sodium phenylbutyrate and taurursodiol have been found to reduce neuronal death in experimental models. The efficacy and safety of a combination of the two compounds in persons with amyotrophic lateral sclerosis (ALS) are not known. METHODS: In this multicenter, randomized, double-blind trial, we enrolled participants with definite ALS who had had an onset of symptoms within the previous 18 months. Participants were randomly assigned in a 2:1 ratio to receive sodium phenylbutyrate-taurursodiol (3 g of sodium phenylbutyrate and 1 g of taurursodiol, administered once a day for 3 weeks and then twice a day) or placebo. The primary outcome was the rate of decline in the total score on the Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised (ALSFRS-R; range, 0 to 48, with higher scores indicating better function) through 24 weeks. Secondary outcomes were the rates of decline in isometric muscle strength, plasma phosphorylated axonal neurofilament H subunit levels, and the slow vital capacity; the time to death, tracheostomy, or permanent ventilation; and the time to death, tracheostomy, permanent ventilation, or hospitalization. RESULTS: A total of 177 persons with ALS were screened for eligibility, and 137 were randomly assigned to receive sodium phenylbutyrate-taurursodiol (89 participants) or placebo (48 participants). In a modified intention-to-treat analysis, the mean rate of change in the ALSFRS-R score was -1.24 points per month with the active drug and -1.66 points per month with placebo (difference, 0.42 points per month; 95% confidence interval, 0.03 to 0.81; P = 0.03). Secondary outcomes did not differ significantly between the two groups. Adverse events with the active drug were mainly gastrointestinal. CONCLUSIONS: Sodium phenylbutyrate-taurursodiol resulted in slower functional decline than placebo as measured by the ALSFRS-R score over a period of 24 weeks. Secondary outcomes were not significantly different between the two groups. Longer and larger trials are necessary to evaluate the efficacy and safety of sodium phenylbutyrate-taurursodiol in persons with ALS. (Funded by Amylyx Pharmaceuticals and others; CENTAUR ClinicalTrials.gov number, NCT03127514.).


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Phenylbutyrates/therapeutic use , Taurochenodeoxycholic Acid/therapeutic use , Aged , Disease Progression , Double-Blind Method , Drug Combinations , Female , Humans , Intention to Treat Analysis , Male , Middle Aged , Phenylbutyrates/adverse effects , Severity of Illness Index , Taurochenodeoxycholic Acid/administration & dosage , Treatment Outcome
2.
Clin Sci (Lond) ; 136(1): 103-120, 2022 01 14.
Article in English | MEDLINE | ID: mdl-34935888

ABSTRACT

Inflammatory response and renal fibrosis are the hallmarks of chronic kidney disease (CKD). However, the specific mechanism of aldosterone-induced renal injury in the progress of CKD requires elucidation. Emerging evidence has demonstrated that absent in melanoma 2 (AIM2)-mediated inflammasome activation and endoplasmic reticulum stress (ERS) play a pivotal role in the renal fibrosis. Here, we investigated whether overexpression or deficiency of AIM2 affects ERS and fibrosis in aldosterone-infused renal injury. Interestingly, we found that AIM2 was markedly expressed in the diseased proximal tubules from human and experimental CKD. Mechanically, overactivation of AIM2 aggravated aldosterone-induced ERS and fibrotic changes in vitro while knockdown of AIM2 blunted these effects in vivo and in vitro. By contrast, AIM2 deficiency ameliorated renal structure and function deterioration, decreased proteinuria levels and lowered systolic blood pressure in vivo; silencing of AIM2 blocked inflammasome-mediated signaling pathway, relieved ERS and fibrotic changes in vivo. Furthermore, mineralocorticoid receptor (MR) antagonist eplerenone and ERS inhibitor tauroursodeoxycholic acid (TUDCA) had nephroprotective effects on the basis of AIM2 overactivation in vitro, while they failed to produce a more remarkable renoprotective effect on the treatment of AIM2 silence in vitro. Notably, the combination of TUDCA with AIM2 knockdown significantly reduced proteinuria levels in vivo. Additionally, immunofluorescence assay identified that apoptosis-associated speck-like protein (ASC) recruitment and Gasdermin-D (GSDMD) cleavage respectively occurred in the glomeruli and tubules in vivo. These findings establish a crucial role for AIM2 inflammasome in aldosterone-induced renal injury, which may provide a novel therapeutic target for the pathogenesis of CKD.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Inflammasomes , Acute Kidney Injury/chemically induced , Aldosterone/administration & dosage , Animals , DNA-Binding Proteins/genetics , Fibrosis/drug therapy , Male , Mice, Inbred C57BL , Renal Insufficiency, Chronic , Taurochenodeoxycholic Acid/administration & dosage
3.
Can J Physiol Pharmacol ; 100(3): 234-239, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34587465

ABSTRACT

Persistent hyperglycemia in type 1 diabetes triggers numerous signaling pathways, which may prove deleterious to the endothelium. As hyperglycemia damages the endothelial layer via multiple signaling pathways, including enhanced oxidative stress, downregulation of angiotensin-converting enzyme 2 signaling, and exacerbation of endoplasmic reticulum (ER) stress, it becomes difficult to prevent injury using monotherapy. Thus, the present study was conceived to evaluate the combined effect of ER stress inhibition along with angiotensin-converting enzyme 2 activation, two major contributors to hyperglycemia-induced endothelial dysfunction, in preventing endothelial dysfunction associated with type 1 diabetes. Streptozotocin-induced diabetic animals were treated with either diminazene aceturate (5 mg·kg-1 per day, p.o.) or tauroursodeoxycholic acid, sodium salt (200 mg·kg-1 per day i.p.), or both for 4 weeks. Endothelial dysfunction was evaluated using vasoreactivity assay, where acetylcholine-induced relaxation was assessed in phenylephrine pre-contracted rings. Combination therapy significantly improved vascular relaxation when compared with diabetic control as well as monotherapy. Restoration of nitrite levels along with prevention of collagen led to improved vasodilatation. Moreover, there was an overall reduction in aortic oxidative stress. We conclude that by simultaneously inhibiting ER stress and activating angiotensin-converting enzyme 2 deleterious effects of hyperglycemia on endothelium were significantly alleviated. This could serve as a novel strategy for the prevention of endothelial dysfunction.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diminazene/analogs & derivatives , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/drug effects , Taurochenodeoxycholic Acid/administration & dosage , Animals , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/physiopathology , Diminazene/administration & dosage , Diminazene/pharmacology , Drug Therapy, Combination , Endothelium, Vascular/physiopathology , Male , Oxidative Stress/drug effects , Rats, Wistar , Streptozocin , Taurochenodeoxycholic Acid/pharmacology
4.
Biochem Biophys Res Commun ; 570: 96-102, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34274852

ABSTRACT

Glucocorticoids are known to induce skeletal muscle atrophy by suppressing protein synthesis and promoting protein degradation. Tauroursodeoxycholic acid (TUDCA) has beneficial effects in several diseases, such as hepatobiliary disorders, hindlimb ischemia and glucocorticoid-induced osteoporosis. However, the effects of TUDCA on glucocorticoid -induced skeletal muscle atrophy remains unknown. Therefore, in the present research, we explored the effects of TUDCA on dexamethasone (DEX)-induced loss and the potential mechanisms involved. We found TUDCA alleviated DEX-induced muscle wasting in C2C12 myotubes, identified by improved myotube differentiation index and expression of myogenin and MHC. And it showed that TUDCA activated the Akt/mTOR/S6K signaling pathway and inhibited FoxO3a transcriptional activity to decreased expression of MuRF1 and Atrogin-1, while blocking Akt by MK2206 blocked these effects of TUDCA on myotubes. Besides, TUDCA also attenuated DEX-induced apoptosis of myotubes. Furthermore, TUDCA was administrated to the mouse model of DEX-induced skeletal muscle atrophy. The results showed that TUDCA improved DEX-induced skeletal muscle atrophy and weakness (identified by increased grip strength and prolonged running exhaustive time) in mice by suppression of apoptosis, reduction of protein degradation and promotion of protein synthesis. Taken together, our research proved for the first time that TUDCA protected against DEX-induced skeletal muscle atrophy not only by improving myogenic differentiation and protein synthesis, but also through decreasing protein degradation and apoptosis of skeletal muscle.


Subject(s)
Muscle, Skeletal/pathology , Muscular Atrophy/drug therapy , Taurochenodeoxycholic Acid/administration & dosage , Taurochenodeoxycholic Acid/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Dexamethasone , Enzyme Activation/drug effects , Male , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/drug effects , Muscular Atrophy/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Taurochenodeoxycholic Acid/pharmacology
5.
Am J Physiol Regul Integr Comp Physiol ; 319(6): R666-R672, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33074709

ABSTRACT

Endoplasmic reticulum stress contributes to ischemia-reperfusion (I/R) injury in rodent and cell models. However, the contribution of endoplasmic reticulum stress in the pathogenesis of endothelial I/R injury in humans is unknown. We tested the hypothesis that compared with placebo, inhibition of endoplasmic reticulum stress via ingestion of tauroursodeoxycholic acid would prevent the attenuation of endothelium-dependent vasodilation following I/R injury. Twelve young adults (6 women) were studied following ingestion of a placebo or 1,500 mg tauroursodeoxycholic acid (TUDCA). Endothelium-dependent vasodilation was assessed via brachial artery flow-mediated dilation (duplex ultrasonography) before and after I/R injury, which was induced by 20 min of arm ischemia followed by 20 min of reperfusion. Endothelium-independent vasodilation (glyceryl trinitrate-mediated vasodilation) was also assessed after I/R injury. Compared with placebo, TUDCA ingestion increased circulating plasma concentrations by 145 ± 90 ng/ml and increased concentrations of the taurine unconjugated form, ursodeoxycholic acid, by 560 ± 156 ng/ml (both P < 0.01). Ischemia-reperfusion injury attenuated endothelium-dependent vasodilation, an effect that did not differ between placebo (pre-I/R, 5.0 ± 2.1% vs. post-I/R, 3.5 ± 2.2%) and TUDCA (pre-I/R, 5.6 ± 2.1% vs. post-I/R, 3.9 ± 2.1%; P = 0.8) conditions. Similarly, endothelium-independent vasodilation did not differ between conditions (placebo, 19.6 ± 4.8% vs. TUDCA, 19.7 ± 6.1%; P = 0.9). Taken together, endoplasmic reticulum stress does not appear to contribute to endothelial I/R injury in healthy young adults.


Subject(s)
Brachial Artery/physiopathology , Endoplasmic Reticulum Stress , Endothelium, Vascular/physiopathology , Reperfusion Injury/physiopathology , Upper Extremity/blood supply , Vasodilation , Adult , Brachial Artery/drug effects , Brachial Artery/metabolism , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Male , Random Allocation , Reperfusion Injury/blood , Single-Blind Method , Taurochenodeoxycholic Acid/administration & dosage , Taurochenodeoxycholic Acid/blood , Vasodilation/drug effects , Young Adult
6.
Int J Mol Sci ; 21(12)2020 Jun 16.
Article in English | MEDLINE | ID: mdl-32560070

ABSTRACT

Tauroursodeoxycholic acid (TUDCA) is a US FDA-approved hydrophilic bile acid for the treatment of chronic cholestatic liver disease. In the present study, we investigate the effects of TUDCA on the proliferation and differentiation of osteoblasts and its therapeutic effect on a mice model of osteoporosis. Following treatment with different concentrations of TUDCA, cell viability, differentiation, and mineralization were measured. Three-month-old female C57BL/6 mice were randomly divided into three groups (n = 8 mice per group): (i) normal mice as the control group, (ii) ovariectomy (OVX) group (receiving phosphate-buffered saline (PBS) treatment every other day for 4 weeks), and (iii) OVX group with TUDCA (receiving TUDCA treatment every other day for 4 weeks starting 6 weeks after OVX). At 11 weeks post-surgery, serum levels of procollagen type I N-terminal propeptides (PINP) and type I collagen crosslinked C-telopeptides (CTX) were measured, and all mice were sacrificed to examine the distal femur by micro-computed tomography (CT) scans and histology. TUDCA (100 nM, 1 µM) significantly increased the proliferation and viability of osteoblasts and osteoblast differentiation and mineralization when used in vitro. Furthermore, TUDCA neutralized the detrimental effects of methylprednisolone (methylprednisolone-induced osteoblast apoptosis). In the TUDCA treatment group the PINP level was higher and the CTX level was lower, but these levels were not significantly different compared to the PBS treatment group. Micro-CT and histology showed that the TUDCA treatment group preserved more trabecular structures in the distal femur compared to the PBS treatment group. In addition, the TUDCA treatment group increased the percentage bone volume with respect to the total bone volume, bone mineral density, and mice distal femur trabeculae compared with the PBS treatment group. Taken together, our findings suggest that TUDCA may provide a favorable effect on bones and could be used for the prevention and treatment of osteoporosis.


Subject(s)
Osteoporosis/drug therapy , Ovariectomy/adverse effects , Peptide Fragments/metabolism , Procollagen/metabolism , Taurochenodeoxycholic Acid/administration & dosage , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Methylprednisolone/adverse effects , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoporosis/etiology , Osteoporosis/metabolism , Random Allocation , Taurochenodeoxycholic Acid/pharmacology , Treatment Outcome
7.
Cardiovasc Drugs Ther ; 33(1): 25-33, 2019 02.
Article in English | MEDLINE | ID: mdl-30663009

ABSTRACT

PURPOSE: The role of endoplasmic reticulum (ER) stress in cardiovascular disease is now recognized. Tauroursodeoxycholic acid (TUDCA) is known to have cardiovascular protective effects by decreasing ER stress. This study aimed to assess the ability of TUDCA to decrease ER stress, inhibit dedifferentiation of vascular smooth muscle cells (VSMCs), and reduce in-stent restenosis. METHODS: The effect of TUDCA on dedifferentiation of VSMCs and ER stress was investigated in vitro using wound-healing assays, MTT assays, and western blotting. For in vivo studies, 18 rabbits were fed an atherogenic diet to induce atheroma formation. Bare metal stents (BMS), BMS+TUDCA or Firebird stents were implanted in the left common carotid artery. Rabbits were euthanized after 28 days and processed for scanning electron microscope (SEM), histological examination (HE), and immunohistochemistry. RESULTS: In vitro TUDCA (10-1000 µmol/L) treatment significantly inhibited platelet-derived growth factor (PDGF)-BB-induced proliferation and migration in VSMCs in a concentration-dependent manner and decreased ER stress markers (IRE1, XBP1, KLF4, and GRP78). In vivo, we confirmed no significant difference in neointimal coverage on three stents surfaces; neointimal was significantly lower with BMS+TUDCA (1.6 ± 0.2 mm2) compared with Firebird (1.90 ± 0.1 mm2) and BMS (2.3 ± 0.1 mm2). Percent stenosis was lowest for BMS+TUDCA, then Firebird, and was significantly higher with BMS (28 ± 4%, 35 ± 7%, 40 ± 1%; respectively; P < 0.001). TUDCA treatment decreased ER stress in the BMS+TUDCA group compared with BMS. CONCLUSIONS: TUDCA inhibited dedifferentiation of VSMCs by decreasing ER stress and reduced in-stent restenosis, possibly through downregulation of the IRE1/XBP1 signaling pathway.


Subject(s)
Carotid Artery Diseases/surgery , Cell Dedifferentiation/drug effects , Drug-Eluting Stents , Endoplasmic Reticulum Stress/drug effects , Endovascular Procedures/instrumentation , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Taurochenodeoxycholic Acid/pharmacology , Administration, Oral , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endovascular Procedures/adverse effects , Kruppel-Like Factor 4 , Male , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima , Protein Serine-Threonine Kinases/metabolism , Rabbits , Rats, Sprague-Dawley , Recurrence , Signal Transduction/drug effects , Taurochenodeoxycholic Acid/administration & dosage , X-Box Binding Protein 1/metabolism
8.
J Dairy Sci ; 102(8): 7359-7370, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31155263

ABSTRACT

Disruption of endoplasmic reticulum (ER) homeostasis, often termed ER stress, is intrinsically linked with perturbation of lipid metabolism in humans and mice. Whether ER homeostasis is affected in cows experiencing fatty liver is unknown. The aim of this study was to investigate the potential role of ER stress in hepatic lipid accumulation in calf hepatocytes and ER stress status in dairy cows with severe fatty liver. In vitro experiments were conducted in which hepatocytes were isolated from calves and treated with different concentrations of fatty acids, tauroursodeoxycholic acid (TUDCA; a canonical inhibitor of ER stress), or both. The increase in phosphorylation level of protein kinase RNA-like ER kinase (PERK) and inositol requiring protein-1α (IRE1α) proteins, and the cleavage of activating transcription factor-6 (ATF6) protein in response to increasing doses of fatty acids (which were reversed by TUDCA treatment) in primary hepatocytes underscored a mechanistic link between fatty acids and ER stress. In addition, fatty acid treatment increased the abundance of sterol regulatory element-binding protein 1c, acetyl-CoA carboxylase-α, fatty acid synthase, and diacylglycerol acyltransferase 1, and lipid accumulation in calf primary hepatocytes, whereas inhibition of ER stress by incubating with TUDCA significantly weakened these effects. Overall, results in vitro indicate that inhibition of ER stress in calf hepatocytes alleviates fatty acid-induced lipid accumulation by downregulating the expression of lipogenic genes. In vivo experiments, liver and blood samples were collected from cows diagnosed as healthy (n = 15) or with severe fatty liver (n = 15). The phosphorylation level of PERK and IRE1α, the cleavage of ATF6 protein, and the abundance of several unfolded protein response genes (78 kDa glucose-regulated protein, AMP-dependent transcription factor 4, and spliced X-box binding protein 1) were greater in liver of cows with severe fatty liver. The present in vivo study confirms the occurrence of ER stress in dairy cows with severe fatty liver. Considering the causative role of fatty acid-induced ER stress in hepatic lipid accumulation in calf hepatocytes, the existence of ER stress in the liver of severe fatty liver cows may presage its participation in fatty liver progression in dairy cows. However, the mechanistic relationship between ER stress and fatty liver in dairy cows remain to be determined.


Subject(s)
Cattle Diseases/physiopathology , Endoplasmic Reticulum Stress/drug effects , Fatty Acids/administration & dosage , Fatty Liver/veterinary , Lipid Metabolism/drug effects , Liver/metabolism , Activating Transcription Factor 6/metabolism , Animals , Cattle , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/metabolism , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipogenesis/genetics , Liver/drug effects , Mice , Phosphorylation , Taurochenodeoxycholic Acid/administration & dosage , Unfolded Protein Response/genetics , eIF-2 Kinase/metabolism
9.
Appl Environ Microbiol ; 83(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28115375

ABSTRACT

The promising results seen in studies of secondary bile acids in experimental colitis suggest that they may represent an attractive and safe class of drugs for the treatment of inflammatory bowel diseases (IBD). However, the exact mechanism by which bile acid therapy confers protection from colitogenesis is currently unknown. Since the gut microbiota plays a crucial role in the pathogenesis of IBD, and exogenous bile acid administration may affect the community structure of the microbiota, we examined the impact of the secondary bile acid ursodeoxycholic acid (UDCA) and its taurine or glycine conjugates on the fecal microbial community structure during experimental colitis. Daily oral administration of UDCA, tauroursodeoxycholic acid (TUDCA), or glycoursodeoxycholic acid (GUDCA) equally lowered the severity of dextran sodium sulfate-induced colitis in mice, as evidenced by reduced body weight loss, colonic shortening, and expression of inflammatory cytokines. Illumina sequencing demonstrated that bile acid therapy during colitis did not restore fecal bacterial richness and diversity. However, bile acid therapy normalized the colitis-associated increased ratio of Firmicutes to Bacteroidetes Interestingly, administration of bile acids prevented the loss of Clostridium cluster XIVa and increased the abundance of Akkermansia muciniphila, bacterial species known to be particularly decreased in IBD patients. We conclude that UDCA, which is an FDA-approved drug for cholestatic liver disorders, could be an attractive treatment option to reduce dysbiosis and ameliorate inflammation in human IBD.IMPORTANCE Secondary bile acids are emerging as attractive candidates for the treatment of inflammatory bowel disease. Although bile acids may affect the intestinal microbial community structure, which significantly contributes to the course of these inflammatory disorders, the impact of bile acid therapy on the fecal microbiota during colitis has not yet been considered. Here, we studied the alterations in the fecal microbial abundance in colitic mice following the administration of secondary bile acids. Our results show that secondary bile acids reduce the severity of colitis and ameliorate colitis-associated fecal dysbiosis at the phylum level. This study indicates that secondary bile acids might act as a safe and effective drug for inflammatory bowel disease.


Subject(s)
Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/drug therapy , Taurochenodeoxycholic Acid/therapeutic use , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/therapeutic use , Animals , Bacteroides/drug effects , Colon/microbiology , Dextran Sulfate/administration & dosage , Disease Models, Animal , Feces/microbiology , Firmicutes/drug effects , Humans , Mice , Taurine/chemistry , Taurochenodeoxycholic Acid/administration & dosage , Ursodeoxycholic Acid/administration & dosage , Ursodeoxycholic Acid/chemistry
10.
Clin Sci (Lond) ; 130(18): 1665-75, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27382986

ABSTRACT

Transthyretin (TTR)-related amyloidoses are diseases characterized by extracellular deposition of amyloid fibrils and aggregates in tissues composed of insoluble misfolded TTR that becomes toxic. Previous studies have demonstrated the ability of small compounds in preventing and reversing TTR V30M deposition in transgenic mice gastrointestinal (GI) tract as well as lowering biomarkers associated with cellular stress and apoptotic mechanisms. In the present study we aimed to study TTR V30M aggregates effect in autophagy, a cellular mechanism crucial for cell survival that has been implicated in the development of several neurodegenerative diseases. We were able to demonstrate in cell culture that TTR V30M aggregates cause a partial impairment of the autophagic machinery as shown by p62 accumulation, whereas early steps of the autophagic flux remain unaffected as shown by autophagosome number evaluation and LC3 turnover assay. Our studies performed in TTR V30M transgenic animals demonstrated that tauroursodeoxycholic acid (TUDCA) and curcumin effectively reverse p62 accumulation in the GI tract pointing to the ability of both compounds to modulate autophagy additionally to mitigate apoptosis. Overall, our in vitro and in vivo studies establish an association between TTR V30M aggregates and autophagy impairment and suggest the use of autophagy modulators as an additional and alternative therapeutic approach for the treatment of TTR V30M-related amyloidosis.


Subject(s)
Amyloid Neuropathies, Familial/physiopathology , Autophagy/drug effects , Curcumin/administration & dosage , Prealbumin/chemistry , Prealbumin/metabolism , Taurochenodeoxycholic Acid/administration & dosage , Amyloid Neuropathies, Familial/drug therapy , Amyloid Neuropathies, Familial/metabolism , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Mutation, Missense , Prealbumin/genetics , Protein Aggregates/drug effects
11.
Clin Sci (Lond) ; 130(21): 1881-8, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27503949

ABSTRACT

Postprandial hyperglycaemia leads to a transient impairment in endothelial function; however, the mechanisms remain largely unknown. Previous work in cell culture models demonstrate that high glucose results in endoplasmic reticulum (ER) stress and, in animal studies, ER stress has been implicated as a cause of endothelial dysfunction. In the present study, we tested the hypothesis that acute oral administration of tauroursodeoxycholic acid (TUDCA, 1500 mg), a chemical chaperone known to alleviate ER stress, would prevent hyperglycaemia-induced endothelial dysfunction. In 12 young healthy subjects (seven men, five women), brachial artery flow-mediated dilation (FMD) was assessed at baseline, and at 60 and 120 min after an oral glucose challenge. Subjects were tested on two separate visits in a single-blind randomized cross-over design: after oral ingestion of TUDCA or placebo capsules. FMD was reduced from baseline during hyperglycaemia under the placebo condition (-32% at 60 min and -28% at 120 min post oral glucose load; P<0.05 from baseline) but not under the TUDCA condition (-4% at 60 min and +0.3% at 120 min post oral glucose load; P>0.05 from baseline). Postprandial plasma glucose and insulin were not altered by TUDCA ingestion. Plasma oxidative stress markers 3-nitrotyrosine and thiobarbituric acid reactive substance (TBARS) remained unaltered throughout the oral glucose challenge in both conditions. These results suggest that hyperglycaemia-induced endothelial dysfunction can be mitigated by oral administration of TUDCA, thus supporting the hypothesis that ER stress may contribute to endothelial dysfunction during postprandial hyperglycaemia.


Subject(s)
Blood Glucose/metabolism , Cardiovascular Diseases/prevention & control , Endothelium, Vascular/drug effects , Hyperglycemia/complications , Hyperglycemia/metabolism , Taurochenodeoxycholic Acid/administration & dosage , Adult , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Insulin/metabolism , Male , Oxidative Stress/drug effects , Postprandial Period , Thiobarbituric Acid Reactive Substances/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Young Adult
12.
Eur J Neurol ; 23(1): 45-52, 2016 01.
Article in English | MEDLINE | ID: mdl-25664595

ABSTRACT

BACKGROUND AND PURPOSE: Tauroursodeoxycholic acid (TUDCA) is a hydrophilic bile acid that is produced in the liver and used for treatment of chronic cholestatic liver diseases. Experimental studies suggest that TUDCA may have cytoprotective and anti-apoptotic action, with potential neuroprotective activity. A proof of principle approach was adopted to provide preliminary data regarding the efficacy and tolerability of TUDCA in a series of patients with amyotrophic lateral sclerosis (ALS). METHODS: As a proof of principle, using a double-blind placebo controlled design, 34 ALS patients under treatment with riluzole who were randomized to placebo or TUDCA (1 g twice daily for 54 weeks) were evaluated after a lead-in period of 3 months. The patients were examined every 6 weeks. The primary outcome was the proportion of responders [those subjects with improvement of at least 15% in the Amyotrophic Lateral Sclerosis Functional Rating Scale Revised (ALSFRS-R) slope during the treatment period compared to the lead-in phase]. Secondary outcomes included between-treatment comparison of ALSFRS-R at study end, comparison of the linear regression slopes for ALSFFRS-R mean scores and the occurrence of adverse events. RESULTS: Tauroursodeoxycholic acid was well tolerated; there were no between-group differences for adverse events. The proportion of responders was higher under TUDCA (87%) than under placebo (P = 0.021; 43%). At study end baseline-adjusted ALSFRS-R was significantly higher (P = 0.007) in TUDCA than in placebo groups. Comparison of the slopes of regression analysis showed slower progression in the TUDCA than in the placebo group (P < 0.01). CONCLUSIONS: This pilot study provides preliminary clinical data indicating that TUDCA is safe and may be effective in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Neuroprotective Agents/pharmacology , Outcome Assessment, Health Care , Riluzole/therapeutic use , Taurochenodeoxycholic Acid/pharmacology , Adult , Aged , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Pilot Projects , Taurochenodeoxycholic Acid/administration & dosage , Taurochenodeoxycholic Acid/adverse effects
13.
Adv Exp Med Biol ; 854: 431-6, 2016.
Article in English | MEDLINE | ID: mdl-26427442

ABSTRACT

We explored the potential protective effects of tauroursodeoxycholic acid (TUDCA) on cone photoreceptor survival in a model of rapid retinal degeneration, the ß-Pde6 (rd1) (rd1) mouse model. We injected two strains of rd1 mice (B6.C3-Pde6b (rd1) Hps4(le)/J and C57BL/6J-Pde6b (rd1-2)/J mice) daily from postnatal day (P) 6 to P21 with TUDCA or vehicle. At P21, retinal function was evaluated with light-adapted electroretinography (ERG) and retinal structure was observed with plastic or frozen sections. TUDCA treatment partially preserved function and structure in B6.C3-Pde6b (rd1) Hps4(le)/J mice but only partially preserved structure in C57BL/6J-Pde6b (rd1-2)/J mice. Our results suggest a possible intervention for patients undergoing rapid retinal degeneration.


Subject(s)
Protective Agents/pharmacology , Retina/drug effects , Retinitis Pigmentosa/prevention & control , Taurochenodeoxycholic Acid/pharmacology , Animals , Cell Count , Disease Models, Animal , Electroretinography , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Protective Agents/administration & dosage , Retina/pathology , Retina/physiopathology , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/physiopathology , Species Specificity , Taurochenodeoxycholic Acid/administration & dosage
14.
Am J Physiol Regul Integr Comp Physiol ; 308(5): R370-8, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25519733

ABSTRACT

Endoplasmic reticulum (ER) stress was previously reported to contribute to neurogenic hypertension while neuronal angiotensin-converting enzyme type 2 (ACE2) overexpression blunts the disease. To assess which brain regions are important for ACE2 beneficial effects and the contribution of ER stress to neurogenic hypertension, we first used transgenic mice harboring a floxed neuronal hACE2 transgene (SL) and tested the impact of hACE2 knockdown in the subfornical organ (SFO) and paraventricular nucleus (PVN) on deoxycorticosterone acetate (DOCA)-salt hypertension. SL and nontransgenic (NT) mice underwent DOCA-salt or sham treatment while infected with an adenoassociated virus (AAV) encoding Cre recombinase (AAV-Cre) or a control virus (AAV-green fluorescent protein) to the SFO or PVN. DOCA-salt-induced hypertension was reduced in SL mice, with hACE2 overexpression in the brain. This reduction was only partially blunted by knockdown of hACE2 in the SFO or PVN, suggesting that both regions are involved but not essential for ACE2 regulation of blood pressure (BP). DOCA-salt treatment did not increase the protein levels of ER stress and autophagy markers in NT mice, despite a significant increase in BP. In addition, these markers were not affected by hACE2 overexpression in the brain, despite a significant reduction of hypertension in SL mice. To further assess the role of ER stress in neurogenic hypertension, NT mice were infused intracerebroventricularlly with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor, during DOCA-salt treatment. However, TUDCA infusion failed to blunt the development of hypertension in NT mice. Our data suggest that brain ER stress does not contribute to DOCA-salt hypertension and that ACE2 blunts neurogenic hypertension independently of ER stress.


Subject(s)
Brain/enzymology , Desoxycorticosterone Acetate , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/enzymology , Hypertension/prevention & control , Peptidyl-Dipeptidase A/metabolism , Sodium Chloride, Dietary , Angiotensin-Converting Enzyme 2 , Animals , Biomarkers/metabolism , Blood Pressure , Brain/drug effects , Brain/physiopathology , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Knockdown Techniques , Humans , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Infusions, Intraventricular , Mice, Inbred C57BL , Mice, Transgenic , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiopathology , Peptidyl-Dipeptidase A/genetics , Subfornical Organ/enzymology , Subfornical Organ/physiopathology , Taurochenodeoxycholic Acid/administration & dosage , Time Factors , Up-Regulation
15.
Am J Physiol Gastrointest Liver Physiol ; 305(4): G286-94, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23764895

ABSTRACT

The bile salt export pump (BSEP), encoded by the abcb11 gene, is the major canalicular transporter of bile acids from the hepatocyte. BSEP malfunction in humans causes bile acid retention and progressive liver injury, ultimately leading to end-stage liver failure. The natural, hydrophilic, bile acid ursodeoxycholic acid (UDCA) is efficacious in the treatment of cholestatic conditions, such as primary biliary cirrhosis and cholestasis of pregnancy. The beneficial effects of UDCA include promoting bile flow, reducing hepatic inflammation, preventing apoptosis, and maintaining mitochondrial integrity in hepatocytes. However, the role of BSEP in mediating UDCA efficacy is not known. Here, we used abcb11 knockout mice (abcb11-/-) to test the effects of acute and chronic UDCA administration on biliary secretion, bile acid composition, liver histology, and liver gene expression. Acutely infused UDCA, or its taurine conjugate (TUDC), was taken up by the liver but retained, with negligible biliary output, in abcb11-/- mice. Feeding UDCA to abcb11-/- mice led to weight loss, retention of bile acids, elevated liver enzymes, and histological damage to the liver. Semiquantitative RT-PCR showed that genes encoding Mdr1a and Mdr1b (canalicular) as well as Mrp4 (basolateral) transporters were upregulated in abcb11-/- mice. We concluded that infusion of UDCA and TUDC failed to induce bile flow in abcb11-/- mice. UDCA fed to abcb11-/- mice caused liver damage and the appearance of biliary tetra- and penta-hydroxy bile acids. Supplementation with UDCA in the absence of Bsep caused adverse effects in abcb11-/- mice.


Subject(s)
ATP-Binding Cassette Transporters/deficiency , Bile Canaliculi/drug effects , Cholestasis/metabolism , Diet , Liver/drug effects , Taurochenodeoxycholic Acid/toxicity , Ursodeoxycholic Acid/toxicity , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Animals , Bile Canaliculi/metabolism , Bile Canaliculi/pathology , Biological Transport , Cholestasis/genetics , Cholestasis/pathology , Disease Models, Animal , Gene Expression Regulation , Infusions, Intravenous , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , RNA, Messenger/metabolism , Taurochenodeoxycholic Acid/administration & dosage , Taurochenodeoxycholic Acid/metabolism , Time Factors , Ursodeoxycholic Acid/administration & dosage , Ursodeoxycholic Acid/metabolism , ATP-Binding Cassette Sub-Family B Member 4
16.
Bull Acad Natl Med ; 196(7): 1333-45; discussion 1345-7, 2012 Oct.
Article in French | MEDLINE | ID: mdl-23815018

ABSTRACT

Patients with familial amyloidpolyneuropathies (FAP) require multidisciplinary neurologic and cardiologic management, including specific treatments to control the progression of systemic amyloidogenesis, symptomatic treatment of peripheral and autonomic neuropathies, and management of severe organ involvement (heart, eyes, kidneys). The first-line specific treatment of choice for met30 TTR-FAP is liver transplantation (LT) which suppresses the main source of mutant TTR, halts the progression of neuropathy in 70% of cases, and doubles the median survival time. Dual kidney-liver or heart-liver transplantation may be appropriate for patients with severe renal or cardiac failure. Tafamidis (Vyndaqel(R), Pfizer), a novel stabilizer of tetrameric TTR, has shown short-term effectiveness in slowing the progression of peripheral neuropathy in very early stages of met30 TTR-FAP This drug should thus be proposed for stage 1 symptomatic polyneuropathy. Other innovative medicines (RNA interference, antisense oligonucleotides) have been developed to block hepatic production of both mutant and wildtype TTR (noxious in late-onset forms of NAH after age 50 years), and to remove amyloid deposits (monoclonal anti-SAP). Clinical trials should first include patients with late-onset FAP or non-met30 TTR-FAP who are less responsive to LT7 and patients in whom Vyndaqel(R) is ineffective or inappropriate. Initial and periodic cardiac assessment is necessary, as cardiac impairment is inevitable and largely responsible for mortality. Symptomatic treatment is crucial to improve these patients' quality of life. Familial screening for carriers of the TTR gene mutation and regular clinical examination are essential to detect disease onset and to start specific therapy in a timely manner.


Subject(s)
Amyloid Neuropathies, Familial/therapy , Amyloid Neuropathies, Familial/drug therapy , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/surgery , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Benzoxazoles/therapeutic use , Clinical Trials as Topic , Diflunisal/therapeutic use , Disease Progression , Doxycycline/administration & dosage , Doxycycline/therapeutic use , Drug Therapy, Combination , Genetic Therapy , Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/surgery , Heart Transplantation , Humans , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery , Kidney Failure, Chronic/therapy , Kidney Transplantation , Liver Transplantation , Myocardium/pathology , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , RNA Interference , Renal Dialysis , Serum Amyloid P-Component/immunology , Taurochenodeoxycholic Acid/administration & dosage , Taurochenodeoxycholic Acid/therapeutic use
17.
J Biomater Appl ; 36(3): 552-561, 2021 09.
Article in English | MEDLINE | ID: mdl-34162236

ABSTRACT

Despite advances in the pathogenesis of Tauroursodeoxycholic acid (TUDCA) on bone, the understanding of the effects and mechanisms of bone osseointegration in TUDCA-associated Hydroxyapatite (HA)-coated titanium implants remains poor. Therefore, the present work was aimed to evaluate the effect of local administration with TUDCA on HA-coated titanium implants osseointegration in ovariectomized(OVX) rats and further investigation of the possible mechanism. Twelve weeks after bilateral ovariectomy, all animals were randomly divided into three groups: sham operation(Sham) group, OVX group and TUDCA group, and all the rats from Sham group and OVX group received HA implants and animals belonging to group TUDCA received TUDCA-HA implants until death at 12 weeks. The bilateral femurs of rats were harvested for evaluation. TUDCA increased new bone formation around the surface of titanium rods and push-out force other than group OVX. Histology, Micro-CT and biochemical analysis results showed systemic TUDCA showed positive effects than OVX group on bone formation in osteopenic rats, with beneficial effect on via activation OPG/RANKL pathway and BMP-2/Smad1 pathway and microarchitecture as well as by reducing protein expression of TNF-α and IFN-γ. The present study suggests that local use of TUDCA may bring benefits to the osseointegration of HA-coated titanium implants in patients with osteoporosis, and this effect may be related to the inhibition of inflammatory reaction and promotion of osteogenesis.


Subject(s)
Coated Materials, Biocompatible/chemistry , Durapatite/chemistry , Osseointegration/drug effects , Taurochenodeoxycholic Acid/pharmacology , Titanium/chemistry , Animals , Female , Ovariectomy , Prostheses and Implants , Rats , Rats, Sprague-Dawley , Taurochenodeoxycholic Acid/administration & dosage
18.
Life Sci ; 285: 120020, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34624320

ABSTRACT

AIMS: The bile acid (BA), tauroursodeoxycholic acid (TUDCA) regulates glucose homeostasis; however, it is not clear whether its effects on insulin signaling are due to its direct interaction with the insulin receptor (IR) or through activation of the G-coupled BA receptor, TGR5. We, herein, investigated whether the actions of TUDCA on glucose homeostasis occur via IR or TGR5 activation. MAIN METHODS: Glucose homeostasis was evaluated in high-fat diet (HFD)-obese or control (CTL) mice, after 30 days or one intraperitoneal (ip) injection of 300 mg/kg TUDCA, respectively. Molecular docking was performed to investigate the potential binding of TUDCA on the IR and TGR5. KEY FINDINGS: After 30 days of TUDCA treatment, HFD mice exhibited improvements in glucose tolerance and insulin sensitivity, which were abolished when these rodents received the IR antagonist, S961. Molecular docking experiments showed that TUDCA demonstrates high binding affinity for TGR5 and IR and strongly interacts with the insulin binding sites 1 and 2 of the IR. Consistent with this potential agonist activity of TUDCA on IR, CTL mice displayed increased hepatic phosphorylation of AKT after an ip injection of TUDCA. This effect was not associated with altered glycemia in CTL mice and was dependent on IR activation, as S961 prevented hepatic AKT activation by TUDCA. Furthermore, TUDCA activated the hepatic protein kinase A (PKA) and cAMP response element-binding protein (CREB) pathway in CTL mice, even after the administration of S961. SIGNIFICANCE: We provide novel evidence that TUDCA may be an agonist of the IR, in turn activating AKT and contributing, at least in part, to its beneficial effects upon glucose homeostasis.


Subject(s)
Glucose/metabolism , Receptor, Insulin/agonists , Taurochenodeoxycholic Acid/pharmacology , Animals , Binding Sites , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose Tolerance Test , Homeostasis/drug effects , Male , Mice , Molecular Docking Simulation , Obesity/metabolism , Protein Binding , Receptor, Insulin/chemistry , Receptors, G-Protein-Coupled/metabolism , Taurochenodeoxycholic Acid/administration & dosage
19.
J Prev Alzheimers Dis ; 8(4): 483-494, 2021.
Article in English | MEDLINE | ID: mdl-34585224

ABSTRACT

BACKGROUND: Obesity and diabetes are well-established risk factors of Alzheimer's disease (AD). In the brains of patients with AD and model mice, diabetes-related factors have been implicated in the pathological changes of AD. However, the molecular mechanistic link between the peripheral metabolic state and AD pathophysiology have remained elusive. Endoplasmic reticulum (ER) stress is known as one of the major contributors to the metabolic abnormalities in obesity and diabetes. Interventions aimed at reducing ER stress have been shown to improve the systemic metabolic abnormalities, although their effects on the AD pathology have not been extensively studied. OBJECTIVES: We examined whether interventions targeting ER stress attenuate the obesity/diabetes-induced Aß accumulation in brains. We also aimed to determine whether ER stress that took place in the peripheral tissues or central nervous system was more important in the Aß neuropathology. Furthermore, we explored if age-related metabolic abnormalities and Aß accumulation could be suppressed by reducing ER stress. METHODS: APP transgenic mice (A7-Tg), which exhibit Aß accumulation in the brain, were used as a model of AD to analyze parameters of peripheral metabolic state, ER stress, and Aß pathology in the brain. Intraperitoneal or intracerebroventricular administration of taurodeoxycholic acid (TUDCA), a chemical chaperone, was performed in high-fat diet (HFD)-fed A7-Tg mice for ~1 month, followed by analyses at 9 months of age. Mice fed a normal diet were treated with TUDCA by drinking water for 4 months and intraperitoneally for 1 month in parallel, and analyzed at 15 months of age. RESULTS: Intraperitoneal administration of TUDCA suppressed ER stress in the peripheral tissues and ameliorated the HFD-induced obesity and insulin resistance. Concomitantly, Aß levels in the brain were significantly reduced. In contrast, intracerebroventricular administration of TUDCA had no effect on the Aß levels. Peripheral administration of TUDCA was also effective against the age-related obesity and insulin resistance, and markedly reduced amyloid accumulation. CONCLUSIONS: Interventions that target peripheral ER stress might be beneficial therapeutic and prevention strategies against brain Aß pathology associated with metabolic overload and aging.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Antiviral Agents/administration & dosage , Endoplasmic Reticulum Stress/drug effects , Taurochenodeoxycholic Acid/administration & dosage , Alzheimer Disease/prevention & control , Animals , Antiviral Agents/pharmacology , Brain/metabolism , Diet , Disease Models, Animal , Humans , Infusions, Intraventricular , Injections, Intraperitoneal , Mice , Mice, Transgenic , Taurochenodeoxycholic Acid/pharmacology
20.
Mol Cell Endocrinol ; 521: 111116, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33321116

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder and the major cause of dementia. According to predictions of the World Health Organization, more than 150 million people worldwide will suffer from dementia by 2050. An increasing number of studies have associated AD with type 2 diabetes mellitus (T2DM), since most of the features found in T2DM are also observed in AD, such as insulin resistance and glucose intolerance. In this sense, some bile acids have emerged as new therapeutic targets to treat AD and metabolic disorders. The taurine conjugated bile acid, tauroursodeoxycholic (TUDCA), reduces amyloid oligomer accumulation and improves cognition in APP/PS1 mice model of AD, and also improves glucose-insulin homeostasis in obese and type 2 diabetic mice. Herein, we investigated the effect of TUDCA upon glucose metabolism in streptozotocin-induced AD mice model (Stz). The Stz mice that received 300 mg/kg TUDCA during 10 days (Stz + TUDCA), showed improvement in glucose tolerance and insulin sensitivity, reduced fasted and fed glycemia, increased islet mass and ß-cell area, as well as increased glucose-stimulated insulin secretion, compared with Stz mice that received only PBS. Stz + TUDCA mice also displayed lower neuroinflammation, reduced protein content of amyloid oligomer in the hippocampus, improved memory test and increased protein content of insulin receptor ß-subunit in the hippocampus. In conclusion, TUDCA treatment enhanced glucose homeostasis in the streptozotocin-induced Alzheimer's disease mice model, pointing this bile acid as a good strategy to counteract glucose homeostasis disturbance in AD pathology.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Bile Acids and Salts/metabolism , Blood Glucose/drug effects , Hippocampus/drug effects , Insulin-Secreting Cells/drug effects , Taurochenodeoxycholic Acid/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Glucose/metabolism , Glucose/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/drug therapy , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Memory and Learning Tests , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Streptozocin/toxicity , Taurochenodeoxycholic Acid/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL