1
|
McInturff EL, France SP, Leverett CA, Flick AC, Lindsey EA, Berritt S, Carney DW, DeForest JC, Ding HX, Fink SJ, Gibson TS, Gray K, Hubbell AK, Johnson AM, Liu Y, Mahapatra S, McAlpine IJ, Watson RB, O'Donnell CJ. Synthetic Approaches to the New Drugs Approved During 2021. J Med Chem 2023; 66:10150-10201. [PMID: 37528515 DOI: 10.1021/acs.jmedchem.3c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Each year, new drugs are introduced to the market, representing structures that have affinity for biological targets implicated in human diseases and conditions. These new chemical entities (NCEs), particularly small molecules and antibody-drug conjugates, provide insight into molecular recognition and serve as potential leads for the design of future medicines. This annual review is part of a continuing series highlighting the most likely process-scale synthetic approaches to 35 NCEs that were first approved anywhere in the world during 2021.
Collapse
Affiliation(s)
- Emma L McInturff
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Scott P France
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Carolyn A Leverett
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andrew C Flick
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Erick A Lindsey
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Simon Berritt
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Daniel W Carney
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Jacob C DeForest
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10777 Science Center Drive, San Diego, California 92121, United States
| | - Hong X Ding
- Pharmacodia (Beijing) Co. Ltd., Beijing, 100085, China
| | - Sarah J Fink
- Takeda Pharmaceuticals, 125 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Tony S Gibson
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Kaitlyn Gray
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Aran K Hubbell
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amber M Johnson
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yiyang Liu
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Subham Mahapatra
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Indrawan J McAlpine
- Genesis Therapeutics, 11568 Sorrento Valley Road, Suite 8, San Diego, California 92121, United States
| | - Rebecca B Watson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10777 Science Center Drive, San Diego, California 92121, United States
| | - Christopher J O'Donnell
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
2
|
Lin X, Zhu X, Xin Y, Zhang P, Xiao Y, He T, Guo H. Intermittent Fasting Alleviates Non-Alcoholic Steatohepatitis by Regulating Bile Acid Metabolism and Promoting Fecal Bile Acid Excretion in High-Fat and High-Cholesterol Diet Fed Mice. Mol Nutr Food Res 2023; 67:e2200595. [PMID: 37148502 DOI: 10.1002/mnfr.202200595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/15/2023] [Indexed: 05/08/2023]
Abstract
SCOPE Intermittent fasting (IF) has a protective role across a wide range of chronic disorders, including obesity, diabetes, and cardiovascular disease, but its protection against non-alcoholic steatohepatitis (NASH) is still lacking. This study seeks to investigate how IF alleviates NASH by regulating gut microbiota and bile acids (BAs) composition. METHODS AND RESULTS Male C57BL/6 mice are fed a high-fat and high-cholesterol (HFHC) diet for 16 weeks to establish a NASH model. Mice then continued HFHC feeding and are treated with or without every other day fasting for 10 weeks. Hepatic pathology is assessed using hematoxylin-eosin staining. Gut microbiota of the cecum are profiled using 16S rDNA gene sequencing and the levels of BAs in serum, colon contents, and feces are measured using ultra-performance liquid chromatography-tandem mass spectrometry. Results indicate that IF significantly decreases murine body weight, insulin resistance, hepatic steatosis, ballooning, and lobular inflammation. IF reshapes the gut microbiota, reduces the accumulation of serum BAs, and increases total colonic and fecal BAs. Moreover, IF increases the expression of cholesterol 7α-hydroxylase 1 in liver, but decreases the expressions of both farnesoid-X-receptor and fibroblast growth factor 15 in the ileum. CONCLUSION IF alleviates NASH by regulating bile acid metabolism and promoting fecal bile acid excretion.
Collapse
Affiliation(s)
- Xiaozhuan Lin
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang, 524023, China
| | - Xuan Zhu
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yan Xin
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang, 524023, China
| | - Peiwen Zhang
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang, 524023, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Yunjun Xiao
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Taiping He
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang, 524023, China
| | - Honghui Guo
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang, 524023, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| |
Collapse
|
3
|
Sugahara S, Haga H, Ikeda C, Makino N, Matsuda A, Kakizaki Y, Hoshikawa K, Katsumi T, Ishizawa T, Kobayashi T, Maki K, Suzuki F, Murakami R, Sato H, Ueno Y. Role of Bile-Derived Extracellular Vesicles in Hepatocellular Proliferation after Partial Hepatectomy in Rats. Int J Mol Sci 2023; 24:ijms24119230. [PMID: 37298180 DOI: 10.3390/ijms24119230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Although liver regeneration has been extensively studied, the effects of bile-derived extracellular vesicles (bile EVs) on hepatocytes has not been elucidated. We examined the influence of bile EVs, collected from a rat model of 70% partial hepatectomy (PH), on hepatocytes. We produced bile-duct-cannulated rats. Bile was collected over time through an extracorporeal bile duct cannulation tube. Bile EVs were extracted via size exclusion chromatography. The number of EVs released into the bile per liver weight 12 h after PH significantly increased. Bile EVs collected 12 and 24 h post-PH, and after sham surgery (PH12-EVs, PH24-EVs, sham-EVs) were added to the rat hepatocyte cell line, and 24 h later, RNA was extracted and transcriptome analysis performed. The analysis revealed that more upregulated/downregulated genes were observed in the group with PH24-EVs. Moreover, the gene ontology (GO) analysis focusing on the cell cycle revealed an upregulation of 28 types of genes in the PH-24 group, including genes that promote cell cycle progression, compared to the sham group. PH24-EVs induced hepatocyte proliferation in a dose-dependent manner in vitro, whereas sham-Evs showed no significant difference compared to the controls. This study revealed that post-PH bile Evs promote the proliferation of the hepatocytes, and genes promoting cell cycles are upregulated in hepatocytes.
Collapse
Affiliation(s)
- Shinpei Sugahara
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Hiroaki Haga
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Chisaki Ikeda
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Naohiko Makino
- Yamagata University Health Administration Center, 1-4-12 Kojirakawa-Machi, Yamagata 990-8560, Japan
| | - Akiko Matsuda
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Yasuharu Kakizaki
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Kyoko Hoshikawa
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Tomohiro Katsumi
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Tetsuya Ishizawa
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Toshikazu Kobayashi
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Keita Maki
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Fumiya Suzuki
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Ryoko Murakami
- Genomic Information Analysis Unit, Department of Genomic Cohort Research, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Hidenori Sato
- Genomic Information Analysis Unit, Department of Genomic Cohort Research, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| | - Yoshiyuki Ueno
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-8595, Japan
| |
Collapse
|
4
|
Zhang S, Chen A, Jiang L, Liu X, Chai L. Copper-mediated shifts in transcriptomic responses of intestines in Bufo gargarizans tadpoles to lead stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:50144-50161. [PMID: 36790706 DOI: 10.1007/s11356-023-25801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/04/2023] [Indexed: 04/16/2023]
Abstract
The differential transcriptomic responses of intestines in Bufo gargarizans tadpoles to Pb alone or in the presence of Cu were evaluated. Tadpoles were exposed to 30 μg/L Pb individually and in combination with Cu at 16 or 64 μg/L from Gosner stage (Gs) 26 to Gs 38. After de novo assembly, 105,107 unigenes were generated. Compared to the control group, 7387, 6937, and 11139 differentially expressed genes (DEGs) were identified in the treatment of Pb + Cu0, Pb + Cu16, and Pb + Cu64, respectively. In addition, functional annotation and enrichment analysis of DEGs revealed substantial transcriptional reprogramming of diverse molecular and biological pathways were induced in all heavy metal treatments. The relative expression levels of genes associated with intestinal epithelial barrier and bile acids (BAs) metabolism, such as mucin2, claudin5, ZO-1, Asbt, and Ost-β, were validated by qPCR. This study demonstrated that Pb exposure induced transcriptional responses in tadpoles, and the responses could be modulated by Cu.
Collapse
Affiliation(s)
- Siliang Zhang
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Aixia Chen
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Ling Jiang
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Xiaoli Liu
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China.
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China.
| |
Collapse
|
5
|
Cai J, Rimal B, Jiang C, Chiang JYL, Patterson AD. Bile acid metabolism and signaling, the microbiota, and metabolic disease. Pharmacol Ther 2022; 237:108238. [PMID: 35792223 DOI: 10.1016/j.pharmthera.2022.108238] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022]
Abstract
The diversity, composition, and function of the bacterial community inhabiting the human gastrointestinal tract contributes to host health through its role in producing energy or signaling molecules that regulate metabolic and immunologic functions. Bile acids are potent metabolic and immune signaling molecules synthesized from cholesterol in the liver and then transported to the intestine where they can undergo metabolism by gut bacteria. The combination of host- and microbiota-derived enzymatic activities contribute to the composition of the bile acid pool and thus there can be great diversity in bile acid composition that depends in part on the differences in the gut bacteria species. Bile acids can profoundly impact host metabolic and immunological functions by activating different bile acid receptors to regulate signaling pathways that control a broad range of complex symbiotic metabolic networks, including glucose, lipid, steroid and xenobiotic metabolism, and modulation of energy homeostasis. Disruption of bile acid signaling due to perturbation of the gut microbiota or dysregulation of the gut microbiota-host interaction is associated with the pathogenesis and progression of metabolic disorders. The metabolic and immunological roles of bile acids in human health have led to novel therapeutic approaches to manipulate the bile acid pool size, composition, and function by targeting one or multiple components of the microbiota-bile acid-bile acid receptor axis.
Collapse
Affiliation(s)
- Jingwei Cai
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Bipin Rimal
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
| | - John Y L Chiang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
6
|
Bile Acid-Drug Interaction via Organic Anion-Transporting Polypeptide 4C1 Is a Potential Mechanism of Altered Pharmacokinetics of Renally Excreted Drugs. Int J Mol Sci 2022; 23:ijms23158508. [PMID: 35955643 PMCID: PMC9369231 DOI: 10.3390/ijms23158508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Patients with liver diseases not only experience the adverse effects of liver-metabolized drugs, but also the unexpected adverse effects of renally excreted drugs. Bile acids alter the expression of renal drug transporters, however, the direct effects of bile acids on drug transport remain unknown. Renal drug transporter organic anion-transporting polypeptide 4C1 (OATP4C1) was reported to be inhibited by chenodeoxycholic acid. Therefore, we predicted that the inhibition of OATP4C1-mediated transport by bile acids might be a potential mechanism for the altered pharmacokinetics of renally excreted drugs. We screened 45 types of bile acids and calculated the IC50, Ki values, and bile acid−drug interaction (BDI) indices of bile acids whose inhibitory effect on OATP4C1 was >50%. From the screening results, lithocholic acid (LCA), glycine-conjugated lithocholic acid (GLCA), and taurine-conjugated lithocholic acid (TLCA) were newly identified as inhibitors of OATP4C1. Since the BDI index of LCA was 0.278, LCA is likely to inhibit OATP4C1-mediated transport in clinical settings. Our findings suggest that dose adjustment of renally excreted drugs may be required in patients with renal failure as well as in patients with hepatic failure. We believe that our findings provide essential information for drug development and safe drug treatment in clinics.
Collapse
|
7
|
Li S, Qu X, Zhang L, Wang N, Chen M, Zhao X, Wang J, Lv H, Qi Y, Zhang L, Liu J, Shi Y. Serum Total Bile Acids in Relation to Gastrointestinal Cancer Risk: A Retrospective Study. Front Oncol 2022; 12:859716. [PMID: 35756666 PMCID: PMC9213662 DOI: 10.3389/fonc.2022.859716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background Bile acids (BAs) have been proposed to promote gastrointestinal cells carcinogenesis. However, studies on serum total bile acid (TBA) levels and gastrointestinal cancers (GICs) risk are rare. Methods We conducted a retrospective case-control study from 2015 to 2019 at the First Affiliated Hospital of Air Force Military Medical University, in which 4,256 GICs cases and 1,333 controls were recruited. Patients' demographic, clinical and laboratory data were collected. The odds ratios (ORs) with 95% confidence intervals (CIs) were estimated using binary logistic regression models. Results Positive associations were observed between serum TBA levels and risks of esophageal cancer (EC), gastric cancer (GC) and colorectal cancer (CRC). Overall, ORs of EC, GC and CRC risk rose with the TBA levels increasing. After adjustment for potential confounders, the OR of TBA-positive for EC risk was 4.89 (95% CI: 3.20-7.49), followed by GC (OR: 3.92, 95% CI: 2.53-6.08), and CRC (OR: 3.32, 95% CI: 2.04-5.11). Patients aged 60 years or older have a higher risk of GICs, especially for EC patients. Males are associated with a higher risk of GC, while females are associated with a higher risk of CRC. Preoperative serum TBA positive and negative was significantly different in the presence or absence of hematogenous metastasis among EC patients (P=0.014), and lymph node metastasis among GC patients (P=0.018). Conclusions This retrospective study showed positive associations between serum TBA level and GICs risk, and a higher serum TBA level constitutes a risk factor for GICs.
Collapse
Affiliation(s)
- Songbo Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiaodong Qu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Luyao Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Na Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Min Chen
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xingyu Zhao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jie Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China.,School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Huanhuan Lv
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China.,School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Ying Qi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China.,School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Lifeng Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Junye Liu
- Department of Radiation Protective Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
8
|
Modulation of the Bile Acid Enterohepatic Cycle by Intestinal Microbiota Alleviates Alcohol Liver Disease. Cells 2022; 11:cells11060968. [PMID: 35326419 PMCID: PMC8946080 DOI: 10.3390/cells11060968] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Reshaping the intestinal microbiota by the ingestion of fiber, such as pectin, improves alcohol-induced liver lesions in mice by modulating bacterial metabolites, including indoles, as well as bile acids (BAs). In this context, we aimed to elucidate how oral supplementation of pectin affects BA metabolism in alcohol-challenged mice receiving feces from patients with alcoholic hepatitis. Pectin reduced alcohol liver disease. This beneficial effect correlated with lower BA levels in the plasma and liver but higher levels in the caecum, suggesting that pectin stimulated BA excretion. Pectin modified the overall BA composition, favoring an augmentation in the proportion of hydrophilic forms in the liver, plasma, and gut. This effect was linked to an imbalance between hydrophobic and hydrophilic (less toxic) BAs in the gut. Pectin induced the enrichment of intestinal bacteria harboring genes that encode BA-metabolizing enzymes. The modulation of BA content by pectin inhibited farnesoid X receptor signaling in the ileum and the subsequent upregulation of Cyp7a1 in the liver. Despite an increase in BA synthesis, pectin reduced BA serum levels by promoting their intestinal excretion. In conclusion, pectin alleviates alcohol liver disease by modifying the BA cycle through effects on the intestinal microbiota and enhanced BA excretion.
Collapse
|
9
|
Kuosmanen RT, Truong K, Rissanen KT, Sievänen EI. The Effect of the Side Chain on Gelation Properties of Bile Acid Alkyl Amides. ChemistryOpen 2021; 10:1150-1157. [PMID: 34806846 PMCID: PMC8607806 DOI: 10.1002/open.202100245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/02/2021] [Indexed: 11/06/2022] Open
Abstract
Six bile acid alkyl amide derivatives were studied with respect to their gelation properties. The derivatives were composed of three different bile acids with hexyl or cyclohexyl side chains. The gelation behaviour of all six compounds were studied for 36 solvents with varying polarities. Gelation was observed mainly in aromatic solvents, which is characteristic for bile-acid-based low molecular weight gelators. Out of 108 bile acid-solvent combinations, a total of 44 gel systems were formed, 28 of which from lithocholic acid derivatives, only two from deoxycholic acid derivatives, and 14 from cholic acid derivatives. The majority of the gel systems were formed from bile acids with hexyl side chains, contrary to the cyclohexyl group, which seems to be a poor gelation moiety. These results indicate that the spatial demand of the side chain is the key feature for the gelation properties of the bile acid amides.
Collapse
Affiliation(s)
- Riikka T. Kuosmanen
- Department of ChemistryUniversity of JyvaskylaP.O. Box 3540014JyväskyläFinland
| | - Khai‐Nghi Truong
- Department of ChemistryUniversity of JyvaskylaP.O. Box 3540014JyväskyläFinland
| | - Kari T. Rissanen
- Department of ChemistryUniversity of JyvaskylaP.O. Box 3540014JyväskyläFinland
| | - Elina I. Sievänen
- Department of ChemistryUniversity of JyvaskylaP.O. Box 3540014JyväskyläFinland
| |
Collapse
|
10
|
Jazaeri F, Sheibani M, Nezamoleslami S, Moezi L, Dehpour AR. Current Models for Predicting Drug-induced Cholestasis: The Role of Hepatobiliary Transport System. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:1-21. [PMID: 34567142 PMCID: PMC8457732 DOI: 10.22037/ijpr.2020.113362.14254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Drug-induced cholestasis is the main type of liver disorder accompanied by high morbidity and mortality. Evidence for the role of hepatobiliary pumps in the cholestasis patho-mechanism is constantly increasing. Recognition of the interactions of chemical agents with these transporters at the initial phases of drug discovery can help develop new drug candidates with low cholestasis potential. This review delivers an outline of the role of these transport proteins in bile creation. It addresses the pathophysiological mechanism for drug-induced cholestasis. In-vitro models, including cell-based and membrane-based approaches and In-vivo models such as genetic knockout animals, are considered. The benefits and restrictions of each model are discussed in this review. Current understandings into the cellular and molecular process that control the activity of hepatobiliary pumps have directed to a better understanding of the pathophysiology of drug-induced cholestasis. A combination of in-vitro monitoring for transport interaction, in-silico predicting systems, and consideration of and metabolic and physicochemical properties must cause more effective monitoring of possible liver problems.
Collapse
Affiliation(s)
- Farahnaz Jazaeri
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,F. J. and M. Sh. contributed equally to this work
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,F. J. and M. Sh. contributed equally to this work
| | - Sadaf Nezamoleslami
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Moezi
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad-Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Tessitore M, Sorrentino E, Schiano Di Cola G, Colucci A, Vajro P, Mandato C. Malnutrition in Pediatric Chronic Cholestatic Disease: An Up-to-Date Overview. Nutrients 2021; 13:2785. [PMID: 34444944 PMCID: PMC8400766 DOI: 10.3390/nu13082785] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/16/2022] Open
Abstract
Despite recent advances, the causes of and effective therapies for pediatric chronic cholestatic diseases remain elusive, and many patients progress to liver failure and need liver transplantation. Malnutrition is a common complication in these patients and is a well-recognized, tremendous challenge for the clinician. We undertook a narrative review of both recent and relevant older literature, published during the last 20 years, for studies linking nutrition to pediatric chronic cholestasis. The collected data confirm that malnutrition and failure to thrive are associated with increased risks of morbidity and mortality, and they also affect the outcomes of liver transplantation, including long-term survival. Malnutrition in children with chronic liver disease is multifactorial and with multiple potential nutritional deficiencies. To improve life expectancy and the quality of life, patients require careful assessments and appropriate management of their nutritional statuses by multidisciplinary teams, which can identify and/or prevent specific deficiencies and initiate appropriate interventions. Solutions available for the clinical management of these children in general, as well as those directed to specific etiologies, are summarized. We particularly focus on fat-soluble vitamin deficiency and malnutrition due to fat malabsorption. Supplemental feeding, including medium-chain triglycerides, essential fatty acids, branched-chain amino acids, and the extra calories needed to overcome the consequences of anorexia and high energy requirements, is reviewed. Future studies should address the need for further improving commercially available and nutritionally complete infant milk formulae for the dietary management of this fragile category of patients. The aid of a specialist dietitian, educational training regarding nutritional guidelines for stakeholders, and improving family nutritional health literacy appear essential.
Collapse
Affiliation(s)
- Maria Tessitore
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Chair of Pediatrics and Residency Program of Pediatrics, Via S. Allende, University of Salerno, 84081 Baronissi, SA, Italy; (M.T.); (E.S.); (G.S.D.C.); (A.C.); (P.V.)
| | - Eduardo Sorrentino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Chair of Pediatrics and Residency Program of Pediatrics, Via S. Allende, University of Salerno, 84081 Baronissi, SA, Italy; (M.T.); (E.S.); (G.S.D.C.); (A.C.); (P.V.)
| | - Giuseppe Schiano Di Cola
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Chair of Pediatrics and Residency Program of Pediatrics, Via S. Allende, University of Salerno, 84081 Baronissi, SA, Italy; (M.T.); (E.S.); (G.S.D.C.); (A.C.); (P.V.)
| | - Angelo Colucci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Chair of Pediatrics and Residency Program of Pediatrics, Via S. Allende, University of Salerno, 84081 Baronissi, SA, Italy; (M.T.); (E.S.); (G.S.D.C.); (A.C.); (P.V.)
| | - Pietro Vajro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Chair of Pediatrics and Residency Program of Pediatrics, Via S. Allende, University of Salerno, 84081 Baronissi, SA, Italy; (M.T.); (E.S.); (G.S.D.C.); (A.C.); (P.V.)
| | - Claudia Mandato
- Department of Pediatrics, Santobono-Pausilipon Children’s Hospital Via M. Fiore, 80129 Naples, Italy
| |
Collapse
|
12
|
Zhang Z, Du Z, Liu Q, Wu T, Tang Q, Zhang J, Huang C, Huang Y, Li R, Li Y, Zhao Y, Zhang G, Zhou J, Huang H, Fang Z, He J. Glucagon-like peptide 1 analogue prevents cholesterol gallstone formation by modulating intestinal farnesoid X receptor activity. Metabolism 2021; 118:154728. [PMID: 33581130 DOI: 10.1016/j.metabol.2021.154728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/13/2021] [Accepted: 02/07/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Cholesterol gallstone disease (CGD) is a common gastrointestinal disease. Liraglutide, an analogue of glucagon-like peptide 1, has been approved to treat type 2 diabetes. Clinical studies have suggested a potential role of liraglutide in CGD. METHODS Mice were subcutaneously injected with liraglutide, then fed a lithogenic diet. Bile duct cannulation was performed to collect bile output in mice. Intestinal-specific ablation or pharmacological inhibition of farnesoid X receptor (FXR) was used to study its functions in CGD. RESULTS Liraglutide could protect mice against CGD. Liraglutide treatment increased the biliary concentration of cholesterol, phospholipids and bile acids and thereby decreased the cholesterol saturation index. The resistance to CGD conferred by liraglutide is likely a result of increased bile acid synthesis and efficient bile acid transport. The expression of a key bile acid synthetic enzyme, Cyp7a1, was significantly increased in liraglutide-treated mice. The increased expression of Cyp7a1 resulted from a relieved suppression signal of Fgf15 from the ileum. Mechanistically, liraglutide treatment altered bile acid composition and suppressed FXR activity in the ileum. Genetic ablation or pharmacological inhibition of FXR in the intestine protected mice against CGD. More importantly, intestinal FXR was required for liraglutide-mediated regulation of hepatic expression of Cyp7a1. CONCLUSION Liraglutide improved CGD by increasing bile acid secretion and decreasing cholesterol saturation index. Liraglutide attenuates the negative feedback inhibition of bile acids through inhibiting intestinal FXR activity. Our results suggest that liraglutide may represent a novel way for treating or preventing cholesterol gallstones in individuals with high risk of CGD.
Collapse
Affiliation(s)
- Zijing Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Zuo Du
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Tong Wu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Qin Tang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jinhang Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Cuiyuan Huang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Ya Huang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Rui Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Yanping Li
- Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Yingnan Zhao
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Guorong Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jian Zhou
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Hui Huang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - ZhongZe Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China.
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China; Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| |
Collapse
|
13
|
Phillips CL, Welch BA, Garrett MR, Grayson BE. Regional heterogeneity in rat Peyer's patches through whole transcriptome analysis. Exp Biol Med (Maywood) 2021; 246:513-522. [PMID: 33236653 PMCID: PMC7934146 DOI: 10.1177/1535370220973014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/20/2020] [Indexed: 11/17/2022] Open
Abstract
Peyer's patches are gut-associated lymphoid tissue located throughout the intestinal wall. Peyer's patches consist of highly organized ovoid-shaped follicles, classified as non-encapsulated lymphatic tissues, populated with B cells, T cells, macrophages, and dendritic cells and function as an organism's intestinal surveillance. Limited work compares the gene profiles of Peyer's patches derived from different intestinal regions. In the current study, we first performed whole transcriptome analysis using RNAseq to compare duodenal and ileal Peyer's patches obtained from the small intestine of Long Evans rats. Of the 12,300 genes that were highly expressed, 18.5% were significantly different between the duodenum and ileum. Using samples obtained from additional subjects (n = 10), we validated the novel gene expression patterns in Peyer's patches obtained from the three regions of the small intestine. Rats had a significantly reduced number of Peyer's patches in the duodenum in comparison to either the jejunum or ileum. Regional differences in structural, metabolic, and immune-related genes were validated. Genes such as alcohol dehydrogenase 1, gap junction protein beta 2, and serine peptidase inhibitor clade b, member 1a were significantly reduced in the ileum in comparison to other regions. On the other hand, genes such as complement C3d receptor type, lymphocyte cytosolic protein 1, and lysozyme C2 precursor were significantly lower in the duodenum. In summary, the gene expression pattern of Peyer's patches is influenced by intestinal location and may contribute to its role in that segment.
Collapse
Affiliation(s)
- Charles L Phillips
- Program in Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bradley A Welch
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bernadette E Grayson
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
14
|
Alqahtani MS, Kazi M, Alsenaidy MA, Ahmad MZ. Advances in Oral Drug Delivery. Front Pharmacol 2021; 12:618411. [PMID: 33679401 PMCID: PMC7933596 DOI: 10.3389/fphar.2021.618411] [Citation(s) in RCA: 356] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
The oral route is the most common route for drug administration. It is the most preferred route, due to its advantages, such as non-invasiveness, patient compliance and convenience of drug administration. Various factors govern oral drug absorption including drug solubility, mucosal permeability, and stability in the gastrointestinal tract environment. Attempts to overcome these factors have focused on understanding the physicochemical, biochemical, metabolic and biological barriers which limit the overall drug bioavailability. Different pharmaceutical technologies and drug delivery systems including nanocarriers, micelles, cyclodextrins and lipid-based carriers have been explored to enhance oral drug absorption. To this end, this review will discuss the physiological, and pharmaceutical barriers influencing drug bioavailability for the oral route of administration, as well as the conventional and novel drug delivery strategies. The challenges and development aspects of pediatric formulations will also be addressed.
Collapse
Affiliation(s)
- Mohammed S. Alqahtani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad A. Alsenaidy
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Z. Ahmad
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Antidiarrheal Effect of Sechang-Zhixie-San on Acute Diarrhea Mice and Network Pharmacology Deciphering Its Characteristics and Potential Mechanisms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8880298. [PMID: 33381214 PMCID: PMC7749774 DOI: 10.1155/2020/8880298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/05/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Sechang-Zhixie-San (SCZX) is an ancient prescription used for pediatric diarrhea by the Yi people in China, which consists of Rodgersia sambucifolia Hemsley (known as Yantuo and abbreviated as YT) and Bentonite (BN). Now, it is also a Chinese patent medicine used in the clinic to treat infantile diarrhea. Besides evaluating the antidiarrheal effect of SCZX on diarrhea mice induced by Folium Sennae, the purpose of this study is to outline the characteristics of the antidiarrheal effect and reveal the potential mechanisms of SCZX through the analysis of the mechanism and active components of YT via network pharmacology and molecular docking, combined with the research progress of BN obtained from the literature. SCZX (3.12 and 12.48 g/kg) effectively inhibited diarrhea in mice, significantly lowering the loose stool rate (LSR), loose stool level (LSL), and loose stool index (LSI). Using network pharmacology, the "herb-compound-target-pathway-pharmacological action" network was mapped to indicate the antidiarrheal mechanism of YT. And the docking results revealed that 4 components of YT including quercetin, geranyl-1-O-α-L-arabinopyranosyl-(1 ⟶ 6)-β-D-glucopyranoside, 3α-O-(E)-p-hydroxy-cinnamoyl-olean-12-en-27-oic acid, and daucosterol showed significant docking activities with STAT3, EGFR, and SLC10A2, involving 11 pathways such as Th17 cell differentiation, Jak-STAT signaling pathway, ErbB signaling pathway, and HIF-1 signaling pathway. According to our research results and literature reports, the antidiarrheal could be summarized into five aspects: inhibiting intestinal inflammation, acting as a barrier to the intestinal mucosal, regulating water and ion transport, involving the purification of intestinal microorganisms, and intestinal transmission, which might be dependent on multiple proteins and intervention in multiple pathways.
Collapse
|
16
|
Volixibat in adults with non-alcoholic steatohepatitis: 24-week interim analysis from a randomized, phase II study. J Hepatol 2020; 73:231-240. [PMID: 32234329 DOI: 10.1016/j.jhep.2020.03.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/27/2020] [Accepted: 03/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Volixibat is an inhibitor of the apical sodium-dependent bile acid transporter (ASBT) that has been hypothesized to improve non-alcoholic steatohepatitis (NASH) by blocking bile acid reuptake and stimulating hepatic bile acid production. We studied the safety, tolerability and efficacy of volixibat in patients with NASH. METHODS In this double-blind, phase II dose-finding study, adults with ≥5% steatosis and NASH without cirrhosis (N = 197) were randomized to receive volixibat (5, 10 or 20 mg) or placebo once daily for 48 weeks. The endpoints of a predefined interim analysis (n = 80), at week 24, were: ≥5% reduction in MRI-proton density fat fraction and ≥20% reduction in serum alanine aminotransferase levels. The primary endpoint was a ≥2-point reduction in non-alcoholic fatty liver disease activity score without worsening fibrosis at week 48. RESULTS Volixibat did not meet either interim endpoint; the study was terminated owing to lack of efficacy. In participants receiving any volixibat dose, mean serum 7-alpha-hydroxy-4-cholesten-3-one (C4; a biomarker of bile acid synthesis) increased from baseline to week 24 (+38.5 ng/ml [SD 53.18]), with concomitant decreases in serum total cholesterol (-14.5 mg/dl [SD 28.32]) and low-density lipoprotein cholesterol (-16.1 mg/dl [SD 25.31]). These changes were generally dose-dependent. On histological analysis, a greater proportion of participants receiving placebo (38.5%, n = 5/13) than volixibat (30.0%, n = 9/30) met the primary endpoint. Treatment-emergent adverse events (TEAEs) were mainly mild or moderate. No serious TEAEs were related to volixibat. Diarrhoea was the most common TEAE overall and the most common TEAE leading to discontinuation. CONCLUSIONS Increased serum C4 and decreased serum cholesterol levels provide evidence of target engagement. However, inhibition of ASBT by volixibat did not elicit a liver-related therapeutic benefit in adults with NASH. LAY SUMMARY A medicine called volixibat has previously been shown to reduce cholesterol levels in the blood. This study investigated whether volixibat could reduce the amount of fat in the liver and reduce liver injury in adults with an advanced form of non-alcoholic fatty liver disease. Volixibat did not reduce the amount of fat in the liver, nor did it have any other beneficial effect on liver injury. Participants in the study generally tolerated the side effects of volixibat and, as in previous studies, the main side effect was diarrhoea. These results show that volixibat is not an effective treatment for people with fatty liver disease. CLINICAL TRIAL IDENTIFIER NCT02787304.
Collapse
|
17
|
Nigam SK, Bush KT, Bhatnagar V, Poloyac SM, Momper JD. The Systems Biology of Drug Metabolizing Enzymes and Transporters: Relevance to Quantitative Systems Pharmacology. Clin Pharmacol Ther 2020; 108:40-53. [PMID: 32119114 PMCID: PMC7292762 DOI: 10.1002/cpt.1818] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
Abstract
Quantitative systems pharmacology (QSP) has emerged as a transformative science in drug discovery and development. It is now time to fully rethink the biological functions of drug metabolizing enzymes (DMEs) and transporters within the framework of QSP models. The large set of DME and transporter genes are generally considered from the perspective of the absorption, distribution, metabolism, and excretion (ADME) of drugs. However, there is a growing amount of data on the endogenous physiology of DMEs and transporters. Recent studies—including systems biology analyses of “omics” data as well as metabolomics studies—indicate that these enzymes and transporters, which are often among the most highly expressed genes in tissues like liver, kidney, and intestine, have coordinated roles in fundamental biological processes. Multispecific DMEs and transporters work together with oligospecific and monospecific ADME proteins in a large multiorgan remote sensing and signaling network. We use the Remote Sensing and Signaling Theory (RSST) to examine the roles of DMEs and transporters in intratissue, interorgan, and interorganismal communication via metabolites and signaling molecules. This RSST‐based view is applicable to bile acids, uric acid, eicosanoids, fatty acids, uremic toxins, and gut microbiome products, among other small organic molecules of physiological interest. Rooting this broader perspective of DMEs and transporters within QSP may facilitate an improved understanding of fundamental biology, physiologically based pharmacokinetics, and the prediction of drug toxicities based upon the interplay of these ADME proteins with key pathways in metabolism and signaling. The RSST‐based view should also enable more tailored pharmacotherapy in the setting of kidney disease, liver disease, metabolic syndrome, and diabetes. We further discuss the pharmaceutical and regulatory implications of this revised view through the lens of systems physiology.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics and Medicine, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Kevin T Bush
- Departments of Pediatrics and Medicine, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Vibha Bhatnagar
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeremiah D Momper
- Division of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
18
|
Impaired bile acid metabolism with defectives of mitochondrial-tRNA taurine modification and bile acid taurine conjugation in the taurine depleted cats. Sci Rep 2020; 10:4915. [PMID: 32188916 PMCID: PMC7080809 DOI: 10.1038/s41598-020-61821-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/03/2020] [Indexed: 11/28/2022] Open
Abstract
Taurine that conjugates with bile acid (BA) and mitochondrial-tRNA (mt-tRNA) is a conditional essential amino acid in humans, similarly to cats. To better understand the influence of acquired depletion of taurine on BA metabolism, the profiling of BAs and its intermediates, BA metabolism-enzyme expression, and taurine modified mt-tRNAs were evaluated in the taurine deficient diet-supplemented cats. In the taurine depleted cats, taurine-conjugated bile acids in bile and taurine-modified mt-tRNA in liver were significantly decreased, whereas unconjugated BA in serum was markedly increased. Impaired bile acid metabolism in the liver was induced accompanied with the decreases of mitochondrial cholesterol 27-hydroxylase expression and mitochondrial activity. Consequently, total bile acid concentration in bile was significantly decreased by the low activity of mitochondrial bile acid synthesis. These results implied that the insufficient dietary taurine intake causes impaired bile acid metabolism, and in turn, a risk for the various diseases similar to the mitochondrial diseases would be enhanced.
Collapse
|
19
|
Choi JU, Maharjan R, Pangeni R, Jha SK, Lee NK, Kweon S, Lee HK, Chang KY, Choi YK, Park JW, Byun Y. Modulating tumor immunity by metronomic dosing of oxaliplatin incorporated in multiple oral nanoemulsion. J Control Release 2020; 322:13-30. [PMID: 32169534 DOI: 10.1016/j.jconrel.2020.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 01/17/2023]
Abstract
In this study, a system for oral delivery of oxaliplatin (OXA) was prepared for metronomic chemotherapy to enhance antitumor efficacy and modulate tumor immunity. OXA was complexed with Nα-deoxycholyl-l-lysyl-methylester (DCK) (OXA/DCK) and formulated as a nanoemulsion (OXA/DCK-NE). OXA/DCK-NE showed 3.35-fold increased permeability across a Caco-2 cell monolayer, resulting in 1.73-fold higher oral bioavailability than free OXA. In addition, treatment of the B16F10.OVA cell line with OXA/DCK-NE resulted in successful upregulation of immunogenic cell death (ICD) markers both in vitro and in vivo. In a B16F10.OVA tumor-bearing mouse model, treatment with OXA/DCK-NE substantially impeded tumor growth by 63.9 ± 13.3% compared to the control group, which was also greater than the intravenous (IV) OXA group. Moreover, treatment with a combination of oral OXA/DCK-NE and anti-programmed cell death protein-1 (αPD-1) antibody resulted in 78.3 ± 9.67% greater inhibition compared to controls. More important, OXA/DCK-NE alone had immunomodulatory effects, such as enhancement of tumor antigen uptake, activation of dendritic cells in tumor-draining lymph nodes, and augmentation of both the population and function of immune effector cells in tumor tissue as well as in the spleen; no such effects were seen in the OXA IV group. These observations provide a rationale for combining oral metronomic OXA with immunotherapy to elicit synergistic antitumor effects.
Collapse
Affiliation(s)
- Jeong Uk Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ruby Maharjan
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Rudra Pangeni
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam 58554, Republic of Korea
| | - Saurav Kumar Jha
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam 58554, Republic of Korea
| | - Na Kyeong Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seho Kweon
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ha Kyeong Lee
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | | | | | - Jin Woo Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam 58554, Republic of Korea.
| | - Youngro Byun
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
20
|
Abstract
Congenital diarrheal disorders are heterogeneous conditions characterized by diarrhea with onset in the first years of life. They range from simple temporary conditions, such as cow's milk protein intolerance to irreversible complications, such as microvillous inclusion disease with significant morbidity and mortality. Advances in genomic medicine have improved our understanding of these disorders, leading to an ever-increasing list of identified causative genes. The diagnostic approach to these conditions consists of establishing the presence of diarrhea by detailed review of the history, followed by characterizing the composition of the diarrhea, the response to fasting, and with further specialized testing.
Collapse
Affiliation(s)
- Abdul Aziz Elkadri
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.
| |
Collapse
|
21
|
Wang L, Zhou Y, Wang X, Zhang G, Guo B, Hou X, Ran J, Zhang Q, Li C, Zhao X, Geng Y, Feng S. Mechanism of Asbt ( Slc10a2)-related bile acid malabsorption in diarrhea after pelvic radiation. Int J Radiat Biol 2020; 96:510-519. [PMID: 31900034 DOI: 10.1080/09553002.2020.1707324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background: Radiation is a mode of treatment for many pelvic malignancies, most of which originate in the gynecologic, gastrointestinal, and genitourinary systems. However, the healthy gut is unavoidably included in the irradiation volume, resulting in undesirable results that manifest as radiation-induced diarrhea (RID), which is the most common side effect of radiation therapy and significantly affects the patients' quality of life. This study aimed to investigate the potential mechanism of diarrhea after pelvic radiotherapy in rats based on the effect of radiation on bile acid homeostasis and sodium-dependent bile acid transporter (Asbt).Methods: In this experimental study, male Sprague-Dawley rats were divided into the following groups - pelvic irradiation, cholestyramine-concurrent radiation, and control groups. The rats in the pelvic irradiation group were irradiated in the pelvic region with 2 Gy per day for five consecutive days. The total bile acid (TBA) levels in the ileum, colon, and feces were measured using automatic biochemical analyzer, and the levels of individual bile acids were evaluated by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). The mRNA and protein expression of Asbt in ileum were assessed by qRT-PCR and Western blot assay. The rats in the cholestyramine-concurrent radiation group were administered with cholestyramine, a bile acid-chelating resin, and concurrent radiation for 5 days. The body weight of rats was monitored daily, and the degree of diarrhea was scored.Results: Diarrhea was observed at 2 and 3 days post-pelvic radiation. The TBA levels were significantly decreased at 4 and 5 days post-radiation in the ileum (p < .01, p < .01) and increased at 4 and 5 days post-radiation in the colon (p < .05, p < .05). The fecal excretions of TBA were significantly increased at 3, 4, and 5 days post-radiation (p < .05). The levels of individual bile acids were significantly decreased in the ileum and increased in the colon and feces, post-radiation. The mRNA and protein expression of Asbt in the ileum gradually decreased with increasing days of pelvic radiation and significantly decreased at 3 and 5 days post-radiation, respectively. Furthermore, a significant decrease in body weight was observed post-pelvic radiation, and cholestyramine administration did not reverse the weight loss. However, the incidence of RID was decreased after administration of cholestyramine.Conclusions: Bile acid malabsorption is partially responsible for RID post-pelvic radiation in rats, and the potential mechanism is related to the downregulation of the ileal Asbt.
Collapse
Affiliation(s)
- Lina Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Radiation Therapy, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yan Zhou
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaohu Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Gansu Provincial Cancer Hospital, Lanzhou, China.,Lanzhou Heavy Ion Hospital, Lanzhou, China
| | - Guangwen Zhang
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Bin Guo
- Department of Radiation Therapy, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoming Hou
- Department of Radiation Therapy, The First Hospital of Lanzhou University, Lanzhou, China
| | - Juntao Ran
- Department of Radiation Therapy, The First Hospital of Lanzhou University, Lanzhou, China
| | | | - Chengcheng Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xueshan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yichao Geng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Shuangwu Feng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
22
|
Brønden A, Knop FK. Gluco-Metabolic Effects of Pharmacotherapy-Induced Modulation of Bile Acid Physiology. J Clin Endocrinol Metab 2020; 105:5601203. [PMID: 31630179 DOI: 10.1210/clinem/dgz025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/04/2019] [Accepted: 10/04/2019] [Indexed: 02/08/2023]
Abstract
CONTEXT The discovery and characterization of the bile acid specific receptors farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5) have facilitated a wealth of research focusing on the link between bile acid physiology and glucose metabolism. Modulation of FXR and TGR5 activation have been demonstrated to affect the secretion of glucagon-like peptide 1, insulin, and glucagon as well as energy expenditure and gut microbiota composition, with potential beneficial effects on glucose metabolism. EVIDENCE ACQUISITION A search strategy based on literature searches in on PubMed with various combinations of the key words FXR, TGR5, agonist, apical sodium-dependent bile acid transporter (ASBT), bile acid sequestrant, metformin, and glucose metabolism has been applied to obtain material for the present review. Furthermore, manual searches including scanning of reference lists in relevant papers and conference proceedings have been performed. EVIDENCE SYNTHESIS This review provides an outline of the link between bile acid and glucose metabolism, with a special focus on the gluco-metabolic impact of treatment modalities with modulating effects on bile acid physiology; including FXR agonists, TGR5 agonists, ASBT inhibitors, bile acid sequestrants, and metformin. CONCLUSIONS Any potential beneficial gluco-metabolic effects of FXR agonists remain to be established, whereas the clinical relevance of TGR5-based treatment modalities seems limited because of substantial safety concerns of TGR5 agonists observed in animal models. The glucose-lowering effects of ASBT inhibitors, bile acid sequestrants, and metformin are at least partly mediated by modulation of bile acid circulation, which might allow an optimization of these bile acid-modulating treatment modalities. (J Clin Endocrinol Metab XX: 00-00, 2019).
Collapse
Affiliation(s)
- Andreas Brønden
- Center for Clinical M etabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Filip K Knop
- Center for Clinical M etabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Steno Diabetes Copenhagen, DK-2820 Gentofte, Denmark
| |
Collapse
|
23
|
Effects of Microcystin-LR on the Microstructure and Inflammation-Related Factors of Jejunum in Mice. Toxins (Basel) 2019; 11:toxins11090482. [PMID: 31438657 PMCID: PMC6783826 DOI: 10.3390/toxins11090482] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/12/2019] [Accepted: 08/17/2019] [Indexed: 12/16/2022] Open
Abstract
The increasing cyanobacterial blooms have recently been considered a severe environmental problem. Microcystin-leucine arginine (MC-LR) is one of the secondary products of cyanobacteria metabolism and most harmful cyanotoxins found in water bodies. Studies show MC-LR negatively affects various human organs when exposed to it. The phenotype of the jejunal chronic toxicity induced by MC-LR has not been well described. The aim of this paper was to investigate the effects of MC-LR on the jejunal microstructure and expression level of inflammatory-related factors in jejunum. Mice were treated with different doses (1, 30, 60, 90 and 120 μg/L) of MC-LR for six months. The microstructure and mRNA expression levels of inflammation-related factors in jejunum were analyzed. Results showed that the microstructure of the jejunum was destroyed and expression levels of inflammation-related factors interleukin (IL)-1β, interleukin (IL)-8, tumor necrosis factor alpha, transforming growth factor-β1 and interleukin (IL)-10 were altered at different MC-LR concentrations. To the best of our knowledge, this is the first study that mice were exposed to a high dose of MC-LR for six months. Our data demonstrated MC-LR had the potential to cause intestinal toxicity by destroying the microstructure of the jejunum and inducing an inflammatory response in mice, which provided new insight into understanding the prevention and diagnosis of the intestinal diseases caused by MC-LR.
Collapse
|
24
|
Pangeni R, Jha SK, Maharjan R, Choi JU, Chang KY, Choi YK, Byun Y, Park JW. Intestinal transport mechanism and in vivo anticancer efficacy of a solid oral formulation incorporating an ion-pairing complex of pemetrexed with deoxycholic acid derivative. Int J Nanomedicine 2019; 14:6339-6356. [PMID: 31496690 PMCID: PMC6690926 DOI: 10.2147/ijn.s209722] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 07/27/2019] [Indexed: 12/20/2022] Open
Abstract
Objective The rational combination of immunotherapy with standard chemotherapy shows synergistic clinical activities in cancer treatment. In the present study, an oral powder formulation of pemetrexed (PMX) was developed to enhance intestinal membrane permeability and investigate its application in metronomic chemotherapy in combination with immunotherapy. Methods PMX was ionically complexed with a bile acid derivative (Nα-deoxycholyl-l-lysyl-methylester; DCK) as a permeation enhancer and mixed with dispersing agents, such as poloxamer 188 (P188) and Labrasol, to form an amorphous oral powder formulation of PMX/DCK (PMX/DCK-OP). Results The apparent permeability (Papp) of PMX/DCK-OP across a Caco-2 cell monolayer was 2.46- and 8.26-fold greater than that of PMX/DCK and free PMX, respectively, which may have been due to the specific interaction of DCK with bile acid transporters, as well as the alteration of membrane fluidity due to Labrasol and P188. Furthermore, inhibition of bile acid transporters by actinomycin D in Caco-2 cell monolayers decreased the Papp of PMX/DCK-OP by 75.4%, suggesting a predominant role of bile acid transporters in the intestinal absorption of PMX/DCK-OP. In addition, caveola/lipid raft-dependent endocytosis, macropinocytosis, passive diffusion, and paracellular transport mechanisms significantly influenced the permeation of PMX/DCK-OP through the intestinal membrane. Therefore, the oral bioavailability of PMX/DCK-OP in rats was 19.8%±6.93%, which was 294% higher than that of oral PMX. Moreover, an in vivo anticancer efficacy study in B16F10 cell-bearing mice treated with a combination of oral PMX/DCK-OP and intraperitoneal anti-PD1 exhibited significant suppression of tumor growth, and the tumor volume was maximally inhibited by 2.03- and 3.16-fold compared to the oral PMX/DCK-OP and control groups, respectively. Conclusion These findings indicated the therapeutic potential of a combination of low-dose oral chemotherapy and immunotherapy for synergistic anticancer efficacy.
Collapse
Affiliation(s)
- Rudra Pangeni
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Saurav Kumar Jha
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Ruby Maharjan
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong Uk Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | | | | | - Youngro Byun
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin Woo Park
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| |
Collapse
|
25
|
Chothe PP, Czuba LC, Ayewoh EN, Swaan PW. Tyrosine Phosphorylation Regulates Plasma Membrane Expression and Stability of the Human Bile Acid Transporter ASBT (SLC10A2). Mol Pharm 2019; 16:3569-3576. [DOI: 10.1021/acs.molpharmaceut.9b00426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Paresh P. Chothe
- Department of Pharmaceutical Sciences, University of Maryland, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Lindsay C. Czuba
- Department of Pharmaceutical Sciences, University of Maryland, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Ebehiremen N. Ayewoh
- Department of Pharmaceutical Sciences, University of Maryland, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Peter W. Swaan
- Department of Pharmaceutical Sciences, University of Maryland, 20 Penn Street, Baltimore, Maryland 21201, United States
| |
Collapse
|
26
|
New Insights in Genetic Cholestasis: From Molecular Mechanisms to Clinical Implications. Can J Gastroenterol Hepatol 2018; 2018:2313675. [PMID: 30148122 PMCID: PMC6083523 DOI: 10.1155/2018/2313675] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 02/06/2023] Open
Abstract
Cholestasis is characterised by impaired bile secretion and accumulation of bile salts in the organism. Hereditary cholestasis is a heterogeneous group of rare autosomal recessive liver disorders, which are characterised by intrahepatic cholestasis, pruritus, and jaundice and caused by defects in genes related to the secretion and transport of bile salts and lipids. Phenotypic manifestation is highly variable, ranging from progressive familial intrahepatic cholestasis (PFIC)-with onset in early infancy and progression to end-stage liver disease-to a milder intermittent mostly nonprogressive form known as benign recurrent intrahepatic cholestasis (BRIC). Cases have been reported of initially benign episodic cholestasis that subsequently transitions to a persistent progressive form of the disease. Therefore, BRIC and PFIC seem to represent two extremes of a continuous spectrum of phenotypes that comprise one disease. Thus far, five representatives of PFIC (named PFIC1-5) caused by pathogenic mutations present in both alleles of ATP8B1, ABCB11, ABCB4, TJP2, and NR1H4 have been described. In addition to familial intrahepatic cholestasis, partial defects in ATP8B1, ABCB11, and ABCB4 predispose patients to drug-induced cholestasis and intrahepatic cholestasis in pregnancy. This review summarises the current knowledge of the clinical manifestations, genetics, and molecular mechanisms of these diseases and briefly outlines the therapeutic options, both conservative and invasive, with an outlook for future personalised therapeutic strategies.
Collapse
|
27
|
Peroxisome Proliferator-Activated Receptor- γ Prevents Cholesterol Gallstone Formation in C57bl Mice by Regulating Bile Acid Synthesis and Enterohepatic Circulation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7475626. [PMID: 30105244 PMCID: PMC6076980 DOI: 10.1155/2018/7475626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/13/2018] [Accepted: 06/27/2018] [Indexed: 12/28/2022]
Abstract
To investigate the role of the peroxisome proliferator-activated receptor-γ (PPARγ) in the progression of cholesterol gallstone disease (CGD), C57bl/6J mice were randomized to the following groups (n=7/group): L (lithogenic diet, LGD), LM (LGD+pioglitazone), CM (chow diet+pioglitazone), and NC (normal control, chow diet). Gallbladder stones were observed by microscopy. Histological gallbladder changes were assessed. Bile acids (BA) and cholesterol were measured in the serum, bile, and feces. Proteins and mRNA expression of genes involved in BA metabolism and enterohepatic circulation were assessed by western blotting and real-time RT-PCR. PPARγ activation was performed in LO2 cell by lentivirus transfection and in Caco2 cell by PPARγ agonist treatment. Downregulation of farnesoid X receptor (FXR) by small interference RNA (siRNA) was performed in L02 cells and Caco2 cells, respectively. Results showed that pharmacological activation of PPARγ by pioglitazone prevents cholesterol gallstone formation by increasing biliary BA synthesis and enterohepatic circulation. Activated PPARγ induced the expression of genes involved in enterohepatic circulation and bile acid synthesis (like PCG1α, BSEP, MRP2, MRP3, MRP4, NTCP, CYP7A1, CYP27A1, ASBT, OSTα, and OSTβ). Downregulation of FXR repressed expression of partial genes involved in BA enterohepatic circulation. These findings suggest a new function of PPARγ in preventing CGD by handling BA synthesis and transport through a FXR dependent or independent pathway.
Collapse
|
28
|
Zhang K, Liu YL, Zhang Y, Zhang J, Deng Z, Wu X, Yin Y. Dynamic oral administration of uridine affects the diurnal rhythm of bile acid and cholesterol metabolism-related genes in mice. BIOL RHYTHM RES 2018. [DOI: 10.1080/09291016.2018.1474844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
| | - Yi-lin Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
| | - Yumei Zhang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
| | - Juan Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xin Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
| | - Yulong Yin
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
29
|
Human and rat precision-cut intestinal slices as ex vivo models to study bile acid uptake by the apical sodium-dependent bile acid transporter. Eur J Pharm Sci 2018; 121:65-73. [PMID: 29751102 DOI: 10.1016/j.ejps.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 03/06/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
|
30
|
Palmer M, Jennings L, Silberg DG, Bliss C, Martin P. A randomised, double-blind, placebo-controlled phase 1 study of the safety, tolerability and pharmacodynamics of volixibat in overweight and obese but otherwise healthy adults: implications for treatment of non-alcoholic steatohepatitis. BMC Pharmacol Toxicol 2018; 19:10. [PMID: 29548345 PMCID: PMC5857122 DOI: 10.1186/s40360-018-0200-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 03/07/2018] [Indexed: 12/13/2022] Open
Abstract
Background Accumulation of toxic free cholesterol in hepatocytes may cause hepatic inflammation and fibrosis. Volixibat inhibits bile acid reuptake via the apical sodium bile acid transporter located on the luminal surface of the ileum. The resulting increase in bile acid synthesis from cholesterol could be beneficial in patients with non-alcoholic steatohepatitis. This adaptive dose-finding study investigated the safety, tolerability, pharmacodynamics, and pharmacokinetics of volixibat. Methods Overweight and obese adults were randomised 3:1 to double-blind volixibat or placebo, respectively, for 12 days. Volixibat was initiated at a once-daily dose of 20 mg, 40 mg or 80 mg. Based on the assessment of predefined safety events, volixibat dosing was either escalated or reduced. Other dose regimens (titrations and twice-daily dosing) were also evaluated. Assessments included safety, tolerability, stool hardness, faecal bile acid (FBA) excretion, and serum levels of 7α-hydroxy-4-cholesten-3-one (C4) and lipids. Results All 84 randomised participants (volixibat, 63; placebo, 21) completed the study, with no serious adverse events at doses of up to 80 mg per day (maximum assessed dose). The median number of daily bowel evacuations increased from 1 (range 0–4) to 2 (0–8) during volixibat treatment, and stool was looser with volixibat than placebo. Volixibat was minimally absorbed; serum levels were rarely quantifiable at any dose or sampling time point, thereby precluding pharmacokinetic analyses. Mean daily FBA excretion was 930.61 μmol (standard deviation [SD] 468.965) with volixibat and 224.75 μmol (195.403) with placebo; effects were maximal at volixibat doses ≥20 mg/day. Mean serum C4 concentrations at day 12 were 98.767 ng/mL (standard deviation, 61.5841) with volixibat and 16.497 ng/mL (12.9150) with placebo. Total and low-density lipoprotein cholesterol levels decreased in the volixibat group, with median changes of − 0.70 mmol/L (range − 2.8 to 0.4) and − 0.6990 mmol/L (− 3.341 to 0.570), respectively. Conclusions This study indicates that maximal inhibition of bile acid reabsorption, as assessed by FBA excretion, occurs at volixibat doses of ≥20 mg/day in obese and overweight adults, without appreciable change in gastrointestinal tolerability. These findings guided dose selection for an ongoing phase 2 study in patients with non-alcoholic steatohepatitis. Trial registration ClinicalTrials.gov identifier: NCT02287779 (registration first received 6 November 2014). Electronic supplementary material The online version of this article (10.1186/s40360-018-0200-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa Palmer
- Global Development Lead Hepatology, Shire, 300 Shire Way, Lexington, MA, 02421, USA.
| | - Lee Jennings
- Global Development Lead Hepatology, Shire, 300 Shire Way, Lexington, MA, 02421, USA
| | - Debra G Silberg
- Shire International GmbH, Zahlerweg 10, 6301, Zug, Switzerland
| | - Caleb Bliss
- Global Development Lead Hepatology, Shire, 300 Shire Way, Lexington, MA, 02421, USA
| | - Patrick Martin
- Global Development Lead Hepatology, Shire, 300 Shire Way, Lexington, MA, 02421, USA
| |
Collapse
|
31
|
Tiessen RG, Kennedy CA, Keller BT, Levin N, Acevedo L, Gedulin B, van Vliet AA, Dorenbaum A, Palmer M. Safety, tolerability and pharmacodynamics of apical sodium-dependent bile acid transporter inhibition with volixibat in healthy adults and patients with type 2 diabetes mellitus: a randomised placebo-controlled trial. BMC Gastroenterol 2018; 18:3. [PMID: 29304731 PMCID: PMC5756385 DOI: 10.1186/s12876-017-0736-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/22/2017] [Indexed: 12/12/2022] Open
Abstract
Background Pathogenesis in non-alcoholic steatohepatitis (NASH) involves abnormal cholesterol metabolism and hepatic accumulation of toxic free cholesterol. Apical sodium-dependent bile acid transporter (ASBT) inhibition in the terminal ileum may facilitate removal of free cholesterol from the liver by reducing recirculation of bile acids (BAs) to the liver, thereby stimulating new BA synthesis from cholesterol. The aim of this phase 1 study in adult healthy volunteers (HVs) and patients with type 2 diabetes mellitus (T2DM) was to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of ASBT inhibition with volixibat (SHP626; formerly LUM002). Methods Participants were randomised 3:1 to receive once-daily oral volixibat (0.5 mg, 1 mg, 5 mg or 10 mg) or placebo for 28 days in two cohorts (HV and T2DM). Assessments included safety, faecal BA and serum 7α-hydroxy-4-cholesten-3-one (C4; BA synthesis biomarker). Results Sixty-one individuals were randomised (HVs: placebo, n = 12; volixibat, n = 38; T2DM: placebo, n = 3; volixibat, n = 8). No deaths or treatment-related serious adverse events were reported. Mild or moderate gastrointestinal adverse events were those most frequently reported with volixibat. With volixibat, mean total faecal BA excretion on day 28 was ~1.6–3.2 times higher in HVs (643.73–1239.3 μmol/24 h) and ~8 times higher in T2DM (1786.0 μmol/24 h) than with placebo (HVs: 386.93 μmol/24 h; T2DM: 220.00 μmol/24 h). With volixibat, mean C4 concentrations increased by ~1.3–5.3-fold from baseline to day 28 in HVs and by twofold in T2DM. Conclusions Volixibat was generally well tolerated. Increased faecal BA excretion and serum C4 levels support the mechanistic rationale for exploring ASBT inhibition in NASH. The study was registered with the Dutch clinical trial authority (Centrale Commissie Mensgebonden Onderzoek; trial registration number NL44732.056.13; registered 24 May 2013). Electronic supplementary material The online version of this article (10.1186/s12876-017-0736-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Renger G Tiessen
- Early Development Services, Pharmaceutical Research Associates (PRA) Health Sciences, Van Swietenlaan 6, 9728 NZ Groningen, PO Box 8144, 9702, Groningen, KC, Netherlands.
| | - Ciara A Kennedy
- Lumena Pharmaceuticals Inc. (part of the Shire group of companies), 12531 High Bluff Drive, Suite 110, San Diego, CA, 92130, USA
| | - Bradley T Keller
- Lumena Pharmaceuticals Inc. (part of the Shire group of companies), 12531 High Bluff Drive, Suite 110, San Diego, CA, 92130, USA
| | - Nancy Levin
- Lumena Pharmaceuticals Inc. (part of the Shire group of companies), 12531 High Bluff Drive, Suite 110, San Diego, CA, 92130, USA
| | - Lisette Acevedo
- Lumena Pharmaceuticals Inc. (part of the Shire group of companies), 12531 High Bluff Drive, Suite 110, San Diego, CA, 92130, USA
| | - Bronislava Gedulin
- Lumena Pharmaceuticals Inc. (part of the Shire group of companies), 12531 High Bluff Drive, Suite 110, San Diego, CA, 92130, USA
| | - Andre A van Vliet
- Early Development Services, Pharmaceutical Research Associates (PRA) Health Sciences, Van Swietenlaan 6, 9728 NZ Groningen, PO Box 8144, 9702, Groningen, KC, Netherlands
| | - Alejandro Dorenbaum
- Lumena Pharmaceuticals Inc. (part of the Shire group of companies), 12531 High Bluff Drive, Suite 110, San Diego, CA, 92130, USA
| | | |
Collapse
|
32
|
Sheikh IA, Ammoury R, Ghishan FK. Pathophysiology of Diarrhea and Its Clinical Implications. PHYSIOLOGY OF THE GASTROINTESTINAL TRACT 2018:1669-1687. [DOI: 10.1016/b978-0-12-809954-4.00068-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
33
|
Chothe PP, Czuba LC, Moore RH, Swaan PW. Human bile acid transporter ASBT (SLC10A2) forms functional non-covalent homodimers and higher order oligomers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:645-653. [PMID: 29198943 DOI: 10.1016/j.bbamem.2017.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 12/28/2022]
Abstract
The human apical sodium-dependent bile acid transporter, hASBT/SLC10A2, plays a central role in cholesterol homeostasis via the efficient reabsorption of bile acids from the distal ileum. hASBT has been shown to self-associate in higher order complexes, but while the functional role of endogenous cysteines has been reported, their implication in the oligomerization of hASBT remains unresolved. Here, we determined the self-association architecture of hASBT by site-directed mutagenesis combined with biochemical, immunological and functional approaches. We generated a cysteine-less form of hASBT by creating point mutations at all 13 endogenous cysteines in a stepwise manner. Although Cysless hASBT had significantly reduced function correlated with lowered surface expression, it featured an extra glycosylation site that facilitated its differentiation from wt-hASBT on immunoblots. Decreased protein expression was associated with instability and subsequent proteasome-dependent degradation of Cysless hASBT protein. Chemical cross-linking of wild-type and Cysless species revealed that hASBT exists as an active dimer and/or higher order oligomer with apparently no requirement for endogenous cysteine residues. This was further corroborated by co-immunoprecipitation of differentially tagged (HA-, Flag-) wild-type and Cysless hASBT. Finally, Cysless hASBT exhibited a dominant-negative effect when co-expressed with wild-type hASBT which validated heterodimerization/oligomerization at the functional level. Combined, our data conclusively demonstrate the functional existence of hASBT dimers and higher order oligomers irrespective of cysteine-mediated covalent bonds, thereby providing greater understanding of its topological assembly at the membrane surface.
Collapse
Affiliation(s)
- Paresh P Chothe
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, 20 Penn Street, Baltimore, MD 21201, USA
| | - Lindsay C Czuba
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, 20 Penn Street, Baltimore, MD 21201, USA
| | - Robyn H Moore
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, 20 Penn Street, Baltimore, MD 21201, USA
| | - Peter W Swaan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, 20 Penn Street, Baltimore, MD 21201, USA.
| |
Collapse
|
34
|
Xiao L, Pan G. An important intestinal transporter that regulates the enterohepatic circulation of bile acids and cholesterol homeostasis: The apical sodium-dependent bile acid transporter (SLC10A2/ASBT). Clin Res Hepatol Gastroenterol 2017; 41:509-515. [PMID: 28336180 DOI: 10.1016/j.clinre.2017.02.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/19/2017] [Accepted: 02/06/2017] [Indexed: 02/04/2023]
Abstract
The enterohepatic circulation of bile acids (BAs) is governed by specific transporters expressed in the liver and the intestine and plays a critical role in the digestion of fats and oils. During this process, the majority of the BAs secreted from the liver is reabsorbed in intestinal epithelial cells via the apical sodium-dependent bile acid transporter (ASBT/SLC10A2) and then transported into the portal vein. Previous studies revealed that regulation of the ASBT involves BAs and cholesterol. In addition, abnormal ASBT expression and function might lead to some diseases associated with disorders in the enterohepatic circulation of BAs and cholesterol homeostasis, such as diarrhoea and gallstones. However, decreasing cholesterol or BAs by partly inhibiting ASBT-mediated transport might be used for treatments of hypercholesterolemia, cholestasis and diabetes. This review mainly discusses the regulation of the ASBT by BAs and cholesterol and its relevance to diseases and treatment.
Collapse
Affiliation(s)
- Ling Xiao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
35
|
Chung SW, Kweon S, Lee BS, Kim GC, Mahmud F, Lee H, Cho YS, Choi JU, Jeon OC, Kim JW, Kim SW, Kim IS, Kim SY, Byun Y. Radiotherapy-assisted tumor selective metronomic oral chemotherapy. Int J Cancer 2017. [DOI: 10.1002/ijc.30842] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Seung Woo Chung
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University; Seoul South Korea
| | - Seho Kweon
- Department of Molecular Medicine and Biopharmaceutical Sciences; Graduate School of Convergent Science and Technology, Seoul National University; Seoul South Korea
| | - Beom Suk Lee
- Biomedical Research Institute, Korea Institute of Science and Technology; Seoul South Korea
- Department of Otolaryngology; Asan Medical Center, University of Ulsan College of Medicine; Seoul South Korea
| | - Gui Chul Kim
- Department of Otolaryngology; Asan Medical Center, University of Ulsan College of Medicine; Seoul South Korea
| | - Foyez Mahmud
- Department of Molecular Medicine and Biopharmaceutical Sciences; Graduate School of Convergent Science and Technology, Seoul National University; Seoul South Korea
| | - Hanul Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University; Seoul South Korea
| | - Young Seok Cho
- Department of Molecular Medicine and Biopharmaceutical Sciences; Graduate School of Convergent Science and Technology, Seoul National University; Seoul South Korea
| | - Jeong Uk Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University; Seoul South Korea
| | | | - Ji Won Kim
- Department of Otolaryngology; Asan Medical Center, University of Ulsan College of Medicine; Seoul South Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology; Asan Medical Center, College of Medicine, University of Ulsan; Seoul South Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology; Seoul South Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University; Seoul South Korea
| | - Sang Yoon Kim
- Department of Otolaryngology; Asan Medical Center, University of Ulsan College of Medicine; Seoul South Korea
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University; Seoul South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences; Graduate School of Convergent Science and Technology, Seoul National University; Seoul South Korea
| |
Collapse
|
36
|
Zhu R, Hou Y, Sun Y, Li T, Fan J, Chen G, Wei J. Pectin Penta-Oligogalacturonide Suppresses Intestinal Bile Acids Absorption and Downregulates the FXR-FGF15 Axis in High-Cholesterol Fed Mice. Lipids 2017; 52:489-498. [PMID: 28474246 DOI: 10.1007/s11745-017-4258-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
Haw pectin penta-oligogalacturonide (HPPS), purified from the hydrolysates of haw pectin, has important role in decreasing hepatic cholesterol accumulation and promoting bile acids (BA) excretion in the feces of mice fed a high-cholesterol diet (HCD). However, the mechanism is not clear. This study aims to investigate the effects of HPPS on BA reabsorption in ileum and biosynthesis in liver of mice. Results showed that HPPS increased fecal BA output by approximately 110%, but decreased ileal BA and the total BA pool size by approximately 47 and 36%, respectively, compared to HCD. Studies of molecular mechanism revealed that HPPS significantly decreased the mRNA and protein levels of farnesoid X receptor (FXR) in the small intestine of mice and inactivated the fibroblast growth factor 15 (FXR-FGF15) axis, which increased the mRNA and protein levels of CYP7A1 by approximately 204 and 104%, respectively, compared to HCD. Interestingly, the mRNA and protein levels of apical sodium-dependent bile acid transporter (ASBT) in the small intestine were approximately 128 and 73% higher in HPPS-fed mice than those in HCD-fed mice, respectively. However, no significant difference was detected for ASBT expression between HCD group and BA sequestrant cholestyramine group. These findings indicate that HPPS can suppress intestinal BA reabsorption and promoting hepatic BA biosynthesis. We speculated that HPPS could be ASBT competitive inhibitor rather than BA sequestrant in inhibiting BA reabsorption in ileum and improving cholesterol metabolism.
Collapse
Affiliation(s)
- Rugang Zhu
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China.
| | - Yuting Hou
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China
| | - Yandi Sun
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China
| | - Tuoping Li
- College of Food Science, Shenyang Agriculture University, Shenyang, 110032, China
| | - Jungang Fan
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang, 110032, China
| | - Gang Chen
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang, 110032, China
| | - Junxiu Wei
- Electrical Engineering and Automation, College of Light Industry, Liaoning University, Shenyang, 110032, China
| |
Collapse
|
37
|
Shi AX, Zhou Y, Zhang XY, Zhao YS, Qin HY, Wang YP, Wu XA. Irinotecan-induced bile acid malabsorption is associated with down-regulation of ileal Asbt ( Slc10a2 ) in mice. Eur J Pharm Sci 2017; 102:220-229. [DOI: 10.1016/j.ejps.2017.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/17/2022]
|
38
|
Lee Y, Yoshitsugu R, Kikuchi K, Joe GH, Tsuji M, Nose T, Shimizu H, Hara H, Minamida K, Miwa K, Ishizuka S. Combination of soya pulp and Bacillus coagulans lilac-01 improves intestinal bile acid metabolism without impairing the effects of prebiotics in rats fed a cholic acid-supplemented diet. Br J Nutr 2016; 116:603-10. [PMID: 27464459 DOI: 10.1017/s0007114516002270] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Intestinal bacteria are involved in bile acid (BA) deconjugation and/or dehydroxylation and are responsible for the production of secondary BA. However, an increase in the production of secondary BA modulates the intestinal microbiota due to the bactericidal effects and promotes cancer risk in the liver and colon. The ingestion of Bacillus coagulans improves constipation via the activation of bowel movement to promote defaecation in humans, which may alter BA metabolism in the intestinal contents. BA secretion is promoted with high-fat diet consumption, and the ratio of cholic acid (CA):chenodeoxycholic acid in primary BA increases with ageing. The dietary supplementation of CA mimics the BA environment in diet-induced obesity and ageing. We investigated whether B. coagulans lilac-01 and soya pulp influence both BA metabolism and the maintenance of host health in CA-supplemented diet-fed rats. In CA-fed rats, soya pulp significantly increased the production of secondary BA such as deoxycholic acid and ω-muricholic acids, and soya pulp ingestion alleviated problems related to plasma adiponectin and gut permeability in rats fed the CA diet. The combination of B. coagulans and soya pulp successfully suppressed the increased production of secondary BA in CA-fed rats compared with soya pulp itself, without impairing the beneficial effects of soya pulp ingestion. In conclusion, it is possible that a combination of prebiotics and probiotics can be used to avoid an unnecessary increase in the production of secondary BA in the large intestine without impairing the beneficial functions of prebiotics.
Collapse
Affiliation(s)
- Yeonmi Lee
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | - Reika Yoshitsugu
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | - Keidai Kikuchi
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | - Ga-Hyun Joe
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | - Misaki Tsuji
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | - Takuma Nose
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | - Hidehisa Shimizu
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | - Hiroshi Hara
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| | | | | | - Satoshi Ishizuka
- 1Research Faculty of Agriculture,Hokkaido University,Sapporo 060-8589,Japan
| |
Collapse
|
39
|
Ding L, Yang L, Wang Z, Huang W. Bile acid nuclear receptor FXR and digestive system diseases. Acta Pharm Sin B 2015; 5:135-44. [PMID: 26579439 PMCID: PMC4629217 DOI: 10.1016/j.apsb.2015.01.004] [Citation(s) in RCA: 291] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/31/2014] [Accepted: 01/05/2015] [Indexed: 12/14/2022] Open
Abstract
Bile acids (BAs) are not only digestive surfactants but also important cell signaling molecules, which stimulate several signaling pathways to regulate some important biological processes. The bile-acid-activated nuclear receptor, farnesoid X receptor (FXR), plays a pivotal role in regulating bile acid, lipid and glucose homeostasis as well as in regulating the inflammatory responses, barrier function and prevention of bacterial translocation in the intestinal tract. As expected, FXR is involved in the pathophysiology of a wide range of diseases of gastrointestinal tract, including inflammatory bowel disease, colorectal cancer and type 2 diabetes. In this review, we discuss current knowledge of the roles of FXR in physiology of the digestive system and the related diseases. Better understanding of the roles of FXR in digestive system will accelerate the development of FXR ligands/modulators for the treatment of digestive system diseases.
Collapse
Key Words
- 6-ECDCA, 6α-ethyl-chenodeoxycholic acid
- AF2, activation domain
- ANGTPL3, angiopoietin-like protein 3
- AOM, azoxymethane
- AP-1, activator protein-1
- ASBT, apical sodium-dependent bile salt transporter
- Apo, apolipoprotein
- BAAT, bile acid-CoA amino acid N-acetyltransferase
- BACS, bile acid-CoA synthetase
- BAs, bile acids
- BMI, body mass index
- BSEP, bile salt export pump
- Bile acids
- CA, cholic acid
- CD, Crohn׳s disease
- CDCA, chenodeoxycholic acid
- CREB, cAMP regulatory element-binding protein
- CYP7A1, cholesterol 7α-hydroxylase
- Colorectal cancer
- DBD, DNA binding domain
- DCA, deoxycholic acid
- DSS, dextrane sodium sulfate
- ERK, extracellular signal-regulated kinase
- FABP6, fatty acid-binding protein subclass 6
- FFAs, free fatty acids
- FGF19, fibroblast growth factor 19
- FGFR4, fibroblast growth factor receptor 4
- FXR, farnesoid X receptor
- FXRE, farnesoid X receptor response element
- Farnesoid X receptor
- G6Pase, glucose-6-phosphatase
- GLP-1, glucagon-like peptide 1
- GLUT2, glucose transporter type 2
- GPBAR, G protein-coupled BA receptor
- GPCRs, G protein-coupled receptors
- GSK3, glycogen synthase kinase 3
- Gastrointestinal tract
- HDL-C, high density lipoprotein cholesterol
- HNF4α, hepatic nuclear factor 4α
- I-BABP, intestinal bile acid-binding protein
- IBD, inflammatory bowel disease
- IL-1, interleukin 1
- Inflammatory bowel disease
- KLF11, Krüppel-like factor 11
- KRAS, Kirsten rat sarcoma viral oncogene homolog
- LBD, ligand binding domain
- LCA, lithocholic acid
- LPL, lipoprotein lipase
- LRH-1, liver receptor homolog-1
- MCA, muricholicacid
- MRP2, multidrug resistance-associated protein 2
- NF-κB, nuclear factor-kappa B
- NOD, non-obese diabetic
- NRs, nuclear receptors
- OSTα, organic solute transporter alpha
- OSTβ, organic solute transporter beta
- PEPCK, phosphoenol pyruvate carboxykinase
- PGC-1α, peroxisome proliferators-activated receptor γ coactivator protein-1α
- SHP, small heterodimer partner
- SREBP-1c, sterol regulatory element-binding protein 1c
- STAT3, signal transducers and activators of transcription 3
- T2D, type 2 diabetes
- TLCA, taurolithocholic acid
- TNBS, trinitrobenzensulfonic acid
- TNFα, tumor necrosis factors α
- Type 2 diabetes
- UC, ulcerative colitis
- UDCA, ursodeoxycholic acid
- VSG, vertical sleeve gastrectomy
- db/db, diabetic mice
Collapse
|
40
|
Nakao N, Kaneda H, Tsushima N, Ohta Y, Tanaka M. Characterization of primary structure and tissue expression profile of the chicken apical sodium-dependent bile acid transporter mRNA. Poult Sci 2015; 94:722-7. [PMID: 25681609 DOI: 10.3382/ps/pev027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ileal apical sodium-dependent bile acid cotransporter (ASBT) plays an essential role in the absorption of bile acids from intestinal lumina. ASBT cDNA has been cloned from mammalian and fish species, and the primary structure of the protein and expression properties of the mRNA have been characterized. In this study, we identified chicken ASBT mRNA by cDNA cloning. Chicken ASBT cDNA consisted of 91 bp of the 5'-untranslated region, 1,083 bp of the coding region, and 1,896 bp of the 3'-untranslated region. The cDNA encoded a protein of 360 amino acids showing significant sequence identity with mammalian and fish ASBT. The amino acid residues known to participate in the functions of mammalian ASBT were conserved in chicken ASBT. Real-time polymerase chain reaction analysis revealed that chicken ASBT mRNA was expressed at markedly higher levels in the ileum and proximal colon/rectum, relatively lower levels in the kidney, and very low levels in the jejunum and cecum. Expression levels in the ileum markedly increased after hatching, reached the highest levels on day 7 posthatching, and then decreased to adult levels. A similar expression pattern was observed in the proximal colon/rectum except for the significant decrease from day 7 posthatching to day 21 posthatching. These results suggest that chicken ASBT functions as a bile acid transporter in the ileum and proximal colon/rectum, particularly during the early posthatching period.
Collapse
Affiliation(s)
- N Nakao
- Laboratory of Animal Physiology, Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonancho, Musashino, Tokyo, 180-8602, Japan
| | - H Kaneda
- Laboratory of Animal Physiology, Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonancho, Musashino, Tokyo, 180-8602, Japan
| | - N Tsushima
- Laboratory of Animal Physiology, Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonancho, Musashino, Tokyo, 180-8602, Japan
| | - Y Ohta
- Laboratory of Applied Biochemistry, Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonancho, Musashino, Tokyo, 180-8602, Japan
| | - M Tanaka
- Laboratory of Animal Physiology, Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonancho, Musashino, Tokyo, 180-8602, Japan
| |
Collapse
|
41
|
Fattah S, Augustijns P, Annaert P. Age-dependent activity of the uptake transporters Ntcp and Oatp1b2 in male rat hepatocytes: from birth till adulthood. Drug Metab Dispos 2015; 43:1-8. [PMID: 25305012 DOI: 10.1124/dmd.114.059212] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Recognition of the role of hepatic drug transporters in elimination of xenobiotics continues to grow. Hepatic uptake transporters, such as hepatic isoforms of the organic anion-transporting polypeptide (Oatp) family as well as the bile acid transporter Na(+)-taurocholate cotransporting polypeptide (Ntcp) have been studied extensively both at the mRNA and protein expression levels in adults. However, in pediatric/juvenile populations, there continues to be a knowledge gap about the functional activity of these transporters. Therefore, the aim of this study was to examine the functional maturation of Ntcp and Oatp isoforms as major hepatic transporters. Hepatocytes were freshly isolated from rats aged between birth and 8 weeks. Transporter activities were assessed by measuring the initial uptake rates of known substrates: taurocholate (TCA) for Ntcp and sodium fluorescein (NaFluo) for Oatp. Relative to adult values, uptake clearance of TCA in hepatocytes from rats aged 0, 1, 2, 3, and 4 weeks reached 19, 43, 22, 46, and 63%, respectively. In contrast, Oatp-mediated NaFluo uptake showed a considerably slower developmental pattern: uptake clearance of NaFluo in hepatocytes from rats aged 0, 1, 2, 3, 4, and 6 weeks were 24, 20, 19, 8, 19, and 64%, respectively. Maturation of NaFluo uptake activity correlated with the previously reported ontogeny of Oatp1b2 mRNA expression, confirming the role of Oatp1b2 for NaFluo uptake in rat liver. The outcome of this project will help in understanding and predicting age-dependent drug exposure in juvenile animals and will eventually support safe and more effective drug therapies for children.
Collapse
Affiliation(s)
- Sarinj Fattah
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Patrick Augustijns
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| |
Collapse
|
42
|
Middendorp S, Schneeberger K, Wiegerinck CL, Mokry M, Akkerman RDL, van Wijngaarden S, Clevers H, Nieuwenhuis EES. Adult stem cells in the small intestine are intrinsically programmed with their location-specific function. Stem Cells 2014; 32:1083-91. [PMID: 24496776 DOI: 10.1002/stem.1655] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 12/21/2013] [Accepted: 12/28/2013] [Indexed: 12/22/2022]
Abstract
Differentiation and specialization of epithelial cells in the small intestine are regulated in two ways. First, there is differentiation along the crypt-villus axis of the intestinal stem cells into absorptive enterocytes, Paneth, goblet, tuft, enteroendocrine, or M cells, which is mainly regulated by WNT. Second, there is specialization along the cephalocaudal axis with different absorptive and digestive functions in duodenum, jejunum, and ileum that is controlled by several transcription factors such as GATA4. However, so far it is unknown whether location-specific functional properties are intrinsically programmed within stem cells or if continuous signaling from mesenchymal cells is necessary to maintain the location-specific identity of the small intestine. Using the pure epithelial organoid technique, we show that region-specific gene expression profiles are conserved throughout long-term cultures of both mouse and human intestinal stem cells and correlated with differential Gata4 expression. Furthermore, the human organoid culture system demonstrates that Gata4-regulated gene expression is only allowed in absence of WNT signaling. These data show that location-specific function is intrinsically programmed in the adult stem cells of the small intestine and that their differentiation fate is independent of location-specific extracellular signals. In light of the potential future clinical application of small intestine-derived organoids, our data imply that it is important to generate GATA4-positive and GATA4-negative cultures to regenerate all essential functions of the small intestine.
Collapse
Affiliation(s)
- Sabine Middendorp
- Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cai JS, Chen JH. The mechanism of enterohepatic circulation in the formation of gallstone disease. J Membr Biol 2014; 247:1067-82. [PMID: 25107305 PMCID: PMC4207937 DOI: 10.1007/s00232-014-9715-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/25/2014] [Indexed: 12/25/2022]
Abstract
Bile acids entering into enterohepatic circulating are primary acids synthesized from cholesterol in hepatocyte. They are secreted actively across canalicular membrane and carried in bile to gallbladder, where they are concentrated during digestion. About 95 % BAs are actively taken up from the lumen of terminal ileum efficiently, leaving only approximately 5 % (or approximately 0.5 g/d) in colon, and a fraction of bile acids are passively reabsorbed after a series of modifications in the human large intestine including deconjugation and oxidation of hydroxy groups. Bile salts hydrolysis and hydroxy group dehydrogenation reactions are performed by a broad spectrum of intestinal anaerobic bacteria. Next, hepatocyte reabsorbs bile acids from sinusoidal blood, which are carried to liver through portal vein via a series of transporters. Bile acids (BAs) transporters are critical for maintenance of the enterohepatic BAs circulation, where BAs exert their multiple physiological functions including stimulation of bile flow, intestinal absorption of lipophilic nutrients, solubilization, and excretion of cholesterol. Tight regulation of BA transporters via nuclear receptors (NRs) is necessary to maintain proper BA homeostasis. In conclusion, disturbances of enterohepatic circulation may account for pathogenesis of gallstones diseases, including BAs transporters and their regulatory NRs and the metabolism of intestinal bacterias, etc.
Collapse
Affiliation(s)
- Jian-Shan Cai
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai, 200040, People's Republic of China,
| | | |
Collapse
|
44
|
Resveratrol promotes degradation of the human bile acid transporter ASBT (SLC10A2). Biochem J 2014; 459:301-12. [PMID: 24498857 DOI: 10.1042/bj20131428] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The sodium/bile acid co-transporter ASBT [apical sodium-dependent bile acid transporter; SLC10A2 (solute carrier family 10 member 2)] plays a key role in the enterohepatic recycling of the bile acids and indirectly contributes to cholesterol homoeostasis. ASBT inhibitors reportedly lower plasma triglyceride levels and increase HDL (high-density lipoprotein) cholesterol levels. RSV (resveratrol), a major constituent of red wine, is known to lower LDL (low-density lipoprotein) cholesterol levels, but its mechanism of action is still unclear. In the present study, we investigated the possible involvement of ASBT in RSV-mediated cholesterol-lowering effects. We demonstrate that RSV inhibits ASBT protein expression and function via a SIRT1 (sirtuin 1)-independent mechanism. The effect was specific to ASBT since other transporters involved in cholesterol homoeostasis, NTCP (SLC10A1), OSTα (SLC51A) and ABCG1 (ATP-binding cassette G1), remained unaffected. ASBT inhibition by RSV was reversed by proteasome inhibitors (MG-132 and lactacystin) and the ubiquitin inhibitor LDN57444, suggesting involvement of the ubiquitin-proteasome pathway. Immunoprecipitation revealed high levels of ubiquitinated ASBT after RSV treatment. Phosphorylation at Ser335 and Thr339 was shown previously to play a role in proteosomal degradation of rat ASBT. However, mutation at corresponding residues in rat ASBT revealed that phosphorylation does not contribute to RSV-mediated degradation of ASBT. Combined, our data indicate that RSV promotes ASBT degradation via the ubiquitin-proteasome pathway without requiring phosphorylation. We conclude that regulation of ASBT expression by RSV may have clinical relevance with regard to the observed cholesterol-lowering effects of RSV.
Collapse
|
45
|
Ngo Sock ET, Farahnak Z, Lavoie JM. Exercise training decreases gene expression of endo- and xeno-sensors in rat small intestine. Appl Physiol Nutr Metab 2014; 39:1098-103. [PMID: 24933213 DOI: 10.1139/apnm-2013-0573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The purpose of the study was to test the hypothesis that gene expression of members of the nuclear receptor (NR) superfamily known to act as endo- and xeno-sensors is reduced in the ileum of exercise-trained (Tr) rats. Healthy female rats were either treadmill-trained for 8 weeks, 5 times/week, or remained sedentary (Sed). Training resulted in a significant (p < 0.05) decrease in plasma free fatty acid (0.18 ± 0.01 to 0.15 ± 0.01 mmol/L) and glycerol (24.8 ± 0.8 to 18.7 ± 0.8 mg/L) concentrations. Gene expressions of NRs farnesoid X receptor (FXR; p < 0.05), liver X receptor (LXR; p < 0.05), pregnane X receptor (PXR; p < 0.01), and retinoid X receptor (RXR; p < 0.06) were reduced in the ileum of Tr compared with Sed animals. Tr was also associated with a reduction (p < 0.05) in gene expression of FXR downstream heterodimeric organite solute transporters α (OSTα) and β (OSTβ) involved in the transport of bile acids, LXR downstream genes heterodimeric ATP-binding cassette transporters (ABCG5/G8) involved in transport of absorbed cholesterol back to the lumen, and Niemann-Pick C1-like 1 (NPC1L1) involved in cholesterol absorption. These data indicate that exercise training lowers the expression of molecules involved in the defense system of the ileum against endobiotic and xenobiotic insults under normal conditions, thus, suggesting that regular exercise contributes to the intestinal maintenance of cholesterol and bile acid homeostasis.
Collapse
Affiliation(s)
- Emilienne Tudor Ngo Sock
- Department of Kinesiology, Université de Montréal, P.O. Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada
| | | | | |
Collapse
|
46
|
Abstract
Enterohepatic circulation is responsible for the capture of bile acids and other steroids produced or metabolized in the liver and secreted to the intestine, for reabsorption back into the circulation and transport back to the liver. Bile acids are secreted from the liver in the form of mixed micelles that also contain phosphatidylcholines and cholesterol that facilitate the uptake of fats and vitamins from the diet due to the surfactant properties of bile acids and lipids. Bile acids are synthesized in the liver from cholesterol by a cascade of enzymes that carry out oxidation and conjugation reactions, and transported to the bile duct and gall bladder where they are stored before being released into the intestine. Bile flow from the gall bladder to the small intestine is triggered by food intake in accordance with its role in lipid and vitamin absorption from the diet. Bile acids are further metabolized by gut bacteria and are transported back to the circulation. Metabolites produced in the liver are termed primary bile acids or primary conjugated bile salts, while the metabolites generated by bacterial are called secondary bile acids. About 95% of bile acids are reabsorbed in the proximal and distal ileum into the hepatic portal vein and then into the liver sinusoids, where they are efficiently transported into the liver with little remaining in circulation. Each bile acid is reabsorbed about 20 times on average before being eliminated. Enterohepatic circulation is under tight regulation by nuclear receptor signaling, notably by the farnesoid X receptor (FXR).
Collapse
Affiliation(s)
- Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
47
|
YANG KYUNGHEE, KÖCK KATHLEEN, SEDYKH ALEXANDER, TROPSHA ALEXANDER, BROUWER KIML. An updated review on drug-induced cholestasis: mechanisms and investigation of physicochemical properties and pharmacokinetic parameters. J Pharm Sci 2013; 102:3037-57. [PMID: 23653385 PMCID: PMC4369767 DOI: 10.1002/jps.23584] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/13/2013] [Accepted: 04/16/2013] [Indexed: 12/15/2022]
Abstract
Drug-induced cholestasis is an important form of acquired liver disease and is associated with significant morbidity and mortality. Bile acids are key signaling molecules, but they can exert toxic responses when they accumulate in hepatocytes. This review focuses on the physiological mechanisms of drug-induced cholestasis associated with altered bile acid homeostasis due to direct (e.g., bile acid transporter inhibition) or indirect (e.g., activation of nuclear receptors, altered function/expression of bile acid transporters) processes. Mechanistic information about the effects of a drug on bile acid homeostasis is important when evaluating the cholestatic potential of a compound, but experimental data often are not available. The relationship between physicochemical properties, pharmacokinetic parameters, and inhibition of the bile salt export pump among 77 cholestatic drugs with different pathophysiological mechanisms of cholestasis (i.e., impaired formation of bile vs. physical obstruction of bile flow) was investigated. The utility of in silico models to obtain mechanistic information about the impact of compounds on bile acid homeostasis to aid in predicting the cholestatic potential of drugs is highlighted.
Collapse
Affiliation(s)
- KYUNGHEE YANG
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - KATHLEEN KÖCK
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - ALEXANDER SEDYKH
- Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - ALEXANDER TROPSHA
- Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - KIM L.R. BROUWER
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
48
|
Liu HT, He HW, Bai XG, Wang JX, Xu CL, Cai SY, Shao RG, Wang YC. Arylsulfonylamino-benzanilides as inhibitors of the apical sodium-dependent bile salt transporter (SLC10A2). Molecules 2013; 18:6883-97. [PMID: 23752471 PMCID: PMC6269792 DOI: 10.3390/molecules18066883] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/04/2013] [Accepted: 06/05/2013] [Indexed: 02/03/2023] Open
Abstract
The apical sodium-dependent bile salt transporter (ASBT) plays a pivotal role in maintaining bile acid homeostasis. Inhibition of ASBT would reduce bile acid pool size and lower cholesterol levels. In this report, a series of novel arylsulfonylaminobenzanilides were designed and synthesized as potential inhibitors of ASBT. Most of them demonstrated great potency against ASBT’s bile acid transport activity. In particular, compound 5g2 inhibited ASBT activity with an IC50 value of 0.11 μM. These compounds represent potential cholesterol-lowering drugs.
Collapse
Affiliation(s)
- Hong-Tao Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hong-Wei He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiao-Guang Bai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ju-Xian Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chang-Liang Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shi-Ying Cai
- Department of Internal Medicine and Liver Center, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Rong-Guang Shao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Authors to whom correspondence should be addressed; E-Mails: (R.G.S.); (Y.C.W.); Tel./Fax: +86-10-6302-6956 (R.G.S.); Tel./Fax: +86-10-6316-5263 (Y.C.W.)
| | - Yu-Cheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Authors to whom correspondence should be addressed; E-Mails: (R.G.S.); (Y.C.W.); Tel./Fax: +86-10-6302-6956 (R.G.S.); Tel./Fax: +86-10-6316-5263 (Y.C.W.)
| |
Collapse
|
49
|
Wu Y, Aquino CJ, Cowan DJ, Anderson DL, Ambroso JL, Bishop MJ, Boros EE, Chen L, Cunningham A, Dobbins RL, Feldman PL, Harston LT, Kaldor IW, Klein R, Liang X, McIntyre MS, Merrill CL, Patterson KM, Prescott JS, Ray JS, Roller SG, Yao X, Young A, Yuen J, Collins JL. Discovery of a highly potent, nonabsorbable apical sodium-dependent bile acid transporter inhibitor (GSK2330672) for treatment of type 2 diabetes. J Med Chem 2013; 56:5094-114. [PMID: 23678871 DOI: 10.1021/jm400459m] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The apical sodium-dependent bile acid transporter (ASBT) transports bile salts from the lumen of the gastrointestinal (GI) tract to the liver via the portal vein. Multiple pharmaceutical companies have exploited the physiological link between ASBT and hepatic cholesterol metabolism, which led to the clinical investigation of ASBT inhibitors as lipid-lowering agents. While modest lipid effects were demonstrated, the potential utility of ASBT inhibitors for treatment of type 2 diabetes has been relatively unexplored. We initiated a lead optimization effort that focused on the identification of a potent, nonabsorbable ASBT inhibitor starting from the first-generation inhibitor 264W94 (1). Extensive SAR studies culminated in the discovery of GSK2330672 (56) as a highly potent, nonabsorbable ASBT inhibitor which lowers glucose in an animal model of type 2 diabetes and shows excellent developability properties for evaluating the potential therapeutic utility of a nonabsorbable ASBT inhibitor for treatment of patients with type 2 diabetes.
Collapse
Affiliation(s)
- Yulin Wu
- GlaxoSmithKline Research & Development, Five Moore Drive, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Matsubara T, Li F, Gonzalez FJ. FXR signaling in the enterohepatic system. Mol Cell Endocrinol 2013; 368:17-29. [PMID: 22609541 PMCID: PMC3491147 DOI: 10.1016/j.mce.2012.05.004] [Citation(s) in RCA: 289] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 04/18/2012] [Accepted: 05/08/2012] [Indexed: 02/07/2023]
Abstract
Enterohepatic circulation serves to capture bile acids and other steroid metabolites produced in the liver and secreted to the intestine, for reabsorption back into the circulation and reuptake to the liver. This process is under tight regulation by nuclear receptor signaling. Bile acids, produced from cholesterol, can alter gene expression in the liver and small intestine via activating the nuclear receptors farnesoid X receptor (FXR; NR1H4), pregnane X receptor (PXR; NR1I2), vitamin D receptor (VDR; NR1I1), G protein coupled receptor TGR5, and other cell signaling pathways (JNK1/2, AKT and ERK1/2). Among these controls, FXR is known to be a major bile acid-responsive ligand-activated transcription factor and a crucial control element for maintaining bile acid homeostasis. FXR has a high affinity for several major endogenous bile acids, notably cholic acid, deoxycholic acid, chenodeoxycholic acid, and lithocholic acid. By responding to excess bile acids, FXR is a bridge between the liver and small intestine to control bile acid levels and regulate bile acid synthesis and enterohepatic flow. FXR is highly expressed in the liver and gut, relative to other tissues, and contributes to the maintenance of cholesterol/bile acid homeostasis by regulating a variety of metabolic enzymes and transporters. FXR activation also affects lipid and glucose metabolism, and can influence drug metabolism.
Collapse
Affiliation(s)
- Tsutomu Matsubara
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Fei Li
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
- Correspondence: Frank J. Gonzalez, Laboratory of Metabolism, National Cancer Institute, Building 37, Room 3106, Bethesda, MD 20892, Tel: 301-496-9067, Fax: 301-496-8419,
| |
Collapse
|